1
|
Wu J, Ge Y, Huang W, Zhang L, Huang J, Huang N, Luo Y. Natural bioactive compounds modified with mesenchymal stem cells: new hope for regenerative medicine. Front Bioeng Biotechnol 2025; 13:1446537. [PMID: 40416310 PMCID: PMC12098461 DOI: 10.3389/fbioe.2025.1446537] [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: 06/10/2024] [Accepted: 04/25/2025] [Indexed: 05/27/2025] Open
Abstract
Mesenchymal stem cells (MSCs) have the potential to differentiate into various cell types, providing important sources of cells for the development of regenerative medicine. Although MSCs have various advantages, there are also various problems, such as the low survival rate of transplanted cells and poor migration and homing; therefore, determining how to reform MSCs to improve their utilization is particularly important. Although many natural bioactive compounds have shown great potential for improving MSCs, many mechanisms and pathways are involved; however, in the final analysis, natural bioactive compounds promoted MSC proliferation, migration and homing and promoted differentiation and antiaging. This article reviews the regulatory effects of natural bioactive compounds on MSCs to provide new ideas for the therapeutic effects of modified MSCs on diseases.
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Ying Ge
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Wendi Huang
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Li Zhang
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Juan Huang
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Nanqu Huang
- National Drug Clinical Trial Institution, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
- Department of Gerontology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| | - Yong Luo
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
- Department of Gerontology, Third Affiliated Hospital of Zunyi Medical University (The First People’s Hospital of Zunyi), Zunyi, Guizhou, China
| |
Collapse
|
2
|
Zhang P, Zhang Z, Li J, Xu M, Lu W, Chen M, Shi J, Wang Q, Zhang H, Huang S, Lian C, Liu J, Ma J, Liu J. Advanced PROTAC and Quantitative Proteomics Strategy Reveals Bax Inhibitor-1 as a Critical Target of Icaritin in Burkitt Lymphoma. Int J Mol Sci 2024; 25:12944. [PMID: 39684655 DOI: 10.3390/ijms252312944] [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: 11/08/2024] [Revised: 11/23/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Understanding the molecular targets of natural products is crucial for elucidating their mechanisms of action, mitigating toxicity, and uncovering potential therapeutic pathways. Icaritin (ICT), a bioactive flavonoid, demonstrates significant anti-tumor activity but lacks defined molecular targets. This study employs an advanced strategy integrating proteolysis targeting chimera (PROTAC) technology with quantitative proteomics to identify ICT's key targets. A library of 22 ICT-based PROTAC derivatives were synthesized, among which LJ-41 exhibited a superior IC50 of 5.52 μM against Burkitt lymphoma (CA-46) cells. Then, differential proteomic analysis identified Bax inhibitor-1 (BI-1) as a potential target. Target validation techniques, including cellular thermal shift assay (CETSA), drug affinity responsive target stability (DARTS) assay, surface plasmon resonance (SPR) assay, and molecular docking, confirmed LJ-41's high specificity for BI-1. Mechanistic investigations revealed that LJ-41 induces apoptosis through BI-1 degradation, triggering endoplasmic reticulum stress and activating inositol-requiring enzyme 1 α (IRE1α), activating transcription factor 6 (ATF6), and nuclear factor erythroid 2-related factor transcription factor heme oxygenase 1 (NRF2-HO-1) signaling pathways. This study establishes a refined methodological framework for natural product target discovery and highlights ICT-PROTAC derivatives' potential for clinical application in Burkitt lymphoma treatment.
Collapse
Affiliation(s)
- Peixi Zhang
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Ziqing Zhang
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Jie Li
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Meng Xu
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Weiming Lu
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Ming Chen
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Jiaqi Shi
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Qiaolai Wang
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Hengyuan Zhang
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Shi Huang
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Chenlei Lian
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Jia Liu
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Junjie Ma
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Jieqing Liu
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| |
Collapse
|
3
|
Reyes-Hernández OD, Figueroa-González G, Quintas-Granados LI, Hernández-Parra H, Peña-Corona SI, Cortés H, Kipchakbayeva A, Mukazhanova Z, Habtemariam S, Leyva-Gómez G, Büsselberg D, Sharifi-Rad J. New insights into the anticancer therapeutic potential of icaritin and its synthetic derivatives. Drug Dev Res 2024; 85:e22175. [PMID: 38567708 DOI: 10.1002/ddr.22175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/06/2024] [Accepted: 03/04/2024] [Indexed: 04/05/2024]
Abstract
Icaritin is a natural prenylated flavonoid derived from the Chinese herb Epimedium. The compound has shown antitumor effects in various cancers, especially hepatocellular carcinoma (HCC). Icaritin exerts its anticancer activity by modulating multiple signaling pathways, such as IL-6/JAK/STAT3, ER-α36, and NF-κB, affecting the tumor microenvironment and immune system. Several clinical trials have evaluated the safety and efficacy of icaritin in advanced HCC patients with poor prognoses, who are unsuitable for conventional therapies. The results have demonstrated that icaritin can improve survival, delay progression, and produce clinical benefits in these patients, with a favorable safety profile and minimal adverse events. Moreover, icaritin can enhance the antitumor immune response by regulating the function and phenotype of various immune cells, such as CD8+ T cells, MDSCs, neutrophils, and macrophages. These findings suggest that icaritin is a promising candidate for immunotherapy in HCC and other cancers. However, further studies are needed to elucidate the molecular mechanisms and optimal dosing regimens of icaritin and its potential synergistic effects with other agents. Therefore, this comprehensive review of the scientific literature aims to summarize advances in the knowledge of icaritin in preclinical and clinical studies as well as the pharmacokinetic, metabolism, toxicity, and mechanisms action to recognize the main challenge, gaps, and opportunities to develop a medication that cancer patients can use. Thus, our main objective was to clarify the current state of icaritin for use as an anticancer drug.
Collapse
Affiliation(s)
- Octavio Daniel Reyes-Hernández
- Laboratorio de Biología Molecular del Cáncer, UMIEZ, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Gabriela Figueroa-González
- Laboratorio de Farmacogenética, UMIEZ, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Laura Itzel Quintas-Granados
- Colegio de Ciencias y Humanidades, Plantel Cuautepec, Universidad Autónoma de la Ciudad de México. Ciudad de México, México, México
| | - Hector Hernández-Parra
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México, Mexico
| | - Sheila I Peña-Corona
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de Mexico, Mexico
| | - Aliya Kipchakbayeva
- Faculty of Chemistry and Chemical Technology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Zhazira Mukazhanova
- Higher School of IT and Natural Sciences, Sarsen Amanzholov East Kazakhstan University, Ust-Kamenogorsk, Kazakhstan
| | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, Central Avenue, Chatham-Maritime, London, UK
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | | | | |
Collapse
|
4
|
Tian Z, Li Y, Wang X, Cui K, Guo J, Wang M, Hao Y, Zhang F. Exploring the mechanism of Astragali radix for promoting osteogenic differentiation based on network pharmacology, molecular docking, and experimental validation. Chem Biol Drug Des 2023; 102:1489-1505. [PMID: 37690812 DOI: 10.1111/cbdd.14340] [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: 04/10/2023] [Revised: 07/20/2023] [Accepted: 08/18/2023] [Indexed: 09/12/2023]
Abstract
The present study used network pharmacology and molecular docking to predict the active ingredients and mechanisms of action of Astragalus radix (AR) to promote osteogenic differentiation of bone marrow mesenchymal stem cells (BM-MSCs), and cell experiments were conducted for verification. First, network pharmacology was used to predict the effective components, targets, and mechanisms of action of AR to promote osteogenic differentiation. The effective components and corresponding target proteins of AR, and the target proteins of osteogenic differentiation were collected through the database. The intersection targets of the two were used for the construction and analysis of a protein-protein interaction (PPI) network. Gene Oncology (GO) and Kyoto Encyclopedia of Genes, and Genomes (KEGG) enrichment analyses were conducted. Next, molecular docking technology was carried out to verify the interaction between the active ingredient and the target protein, and to select the appropriate effective active ingredient. Finally, the results of network pharmacology analysis were verified by in vitro experiments. A total of 95 potential targets were retrieved by searching the intersection of AR and osteogenic differentiation targets. PPI network analysis indicated that RAC-α-serine-threonine-protein kinase (Akt1) was considered to be the most reliable target for AR to regulate osteogenic differentiation. GO enrichment analysis included 21 biological processes, 21 cellular components and 100 molecular functions. KEGG enrichment analysis indicated that the class I phosphatidylinositol-3 kinase (PI3K)-serine-threonine kinase (Akt) signaling pathway may play an important role in promoting osteogenic differentiation. The results of molecular docking showed that quercetin's performance was improved compared with that of kaempferol. In vitro experiments showed that quercetin promoted the expression of osteogenic marker proteins (including collagen I, Runt-related transcription factor 2 and osteopontin) in BMSCs and activated the PI3K/Akt signaling pathway. AR acted on Akt1 targets through its main active component quercetin, and promoted the osteogenic differentiation of BM-MSCs by activating the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Zenghui Tian
- College of First Clinical Medical, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yingying Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaoying Wang
- Teaching and Research Department of Internal Medicine, Jinan Vocational College of Nursing, Jinan, China
| | - Kaiying Cui
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinxing Guo
- College of First Clinical Medical, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mingliang Wang
- Department of Orthopedics, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, China
| | - Yanke Hao
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Farong Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
5
|
Huong NT, Son NT. Icaritin: A phytomolecule with enormous pharmacological values. PHYTOCHEMISTRY 2023:113772. [PMID: 37356700 DOI: 10.1016/j.phytochem.2023.113772] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 05/24/2023] [Accepted: 06/10/2023] [Indexed: 06/27/2023]
Abstract
Pharmacological studies on flavonoids have always drawn much interest for many years. Icaritin (ICT), a representative flavone containing an 8-prenyl group, is a principal compound detected in medicinal plants of the genus Epimedum, the family Berberidaceae. Experimental results in the phytochemistry and pharmacology of this molecule are abundant now, but a deep overview has not been carried out. The goal of this review is to provide an insight into the natural observation, biosynthesis, biotransformation, synthesis, pharmacology, and pharmacokinetics of prenyl flavone ICT. The relevant data on ICT was collected from bibliographic sources, like Google Scholar, Web of Science, Sci-Finder, and various published journals. "Icaritin" alone or in combination is the main keyword to seek for references, and references have been updated till now. ICT is among the characteristic phytomolecules of Epimedum plants. Bacteria monitored its biosynthesis and biotransformation, while this agent was rapidly synthesized from phloroglucinol by microwave-assistance Claisen rearrangement. ICT is a potential agent in numerous in vitro and in vivo pharmacological records, which demonstrated its role in cancer treatments via apoptotic-related mechanisms. It also brings in various health benefits since it reduced harmful effects on the liver, lung, heart, bone, blood, and skin, and improved immune responses. Pharmacokinetic outcomes indicated that its metabolic pathway involved hydration, hydroxylation, dehydrogenation, glycosylation, and glucuronidation. Molecule mechanisms of action at a cellular level are predominant, but clinical studies are expected to get more. Structure-activity relationship records seem insufficient, and the studies on nano-combined approaches to improve its soluble property in living bodied medium are needed.
Collapse
Affiliation(s)
- Nguyen Thi Huong
- Faculty of Chemical Technology, Hanoi University of Industry, Hanoi, Viet Nam
| | - Ninh The Son
- Institute of Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Caugiay, Hanoi, Viet Nam.
| |
Collapse
|
6
|
Sun C, Cao N, Wang Q, Liu N, Yang T, Li S, Pan L, Yao J, Zhang L, Liu M, Zhang G, Xiao X, Liu C. Icaritin induces resolution of inflammation by targeting cathepsin B to prevents mice from ischemia-reperfusion injury. Int Immunopharmacol 2023. [DOI: 10.1016/j.intimp.2023.109850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
|
7
|
Xiao Y, Yao W, Lin M, Huang W, Li B, Peng B, Ma Q, Zhou X, Liang M. Icaritin-loaded PLGA nanoparticles activate immunogenic cell death and facilitate tumor recruitment in mice with gastric cancer. Drug Deliv 2022; 29:1712-1725. [PMID: 35635307 PMCID: PMC9176696 DOI: 10.1080/10717544.2022.2079769] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/04/2022] [Accepted: 05/09/2022] [Indexed: 12/21/2022] Open
Abstract
This study aimed to explore the anti-tumor effect of icaritin loading poly (lactic-co-glycolic acid) nanoparticles (refer to PLGA@Icaritin NPs) on gastric cancer (GC) cells. Transmission Electron Microscope (TEM), size distribution, zeta potential, drug-loading capability, and other physicochemical characteristics of PLGA@Icaritin NPs were carried out. Furthermore, flow cytometry, confocal laser scanning microscope (CLSM), Cell Counting Kit-8 (CCK-8), Transwell, Elisa assay and Balb/c mice were applied to explore the cellular uptake, anti-proliferation, anti-metastasis, immune response activation effects, and related anti-tumor mechanism of PLGA@Icaritin NPs in vitro and in vivo. PLGA@Icaritin NPs showed spherical shape, with appropriate particle sizes and well drug loading and releasing capacities. Flow cytometry and CLSM results indicated that PLGA@Icaritin could efficiently enter into GC cells. CCK-8 proved that PLGA@Icaritin NPs dramatically suppressed cell growth, induced Lactic dehydrogenase (LDH) leakage, arrested more GC cells at G2 phase, and inhibited the invasion and metastasis of GC cells, compared to free icaritin. In addition, PLGA@Icaritin could help generate dozens of reactive oxygen species (ROS) within GC cells, following by significant mitochondrial membrane potentials (MMPs) loss and excessive production of oxidative-mitochondrial DNA (Ox-mitoDNA). Since that, Ox-mitoDNA further activated the releasing of damage associated molecular pattern molecules (DAMPs), and finally led to immunogenic cell death (ICD). Our in vivo data also elaborated that PLGA@Icaritin exerted a powerful inhibitory effect (∼80%), compared to free icaritin (∼60%). Most importantly, our results demonstrated that PLGA@Icaritin could activate the anti-tumor immunity via recruitment of infiltrating CD4+ cells, CD8+ T cells and increased secretion of cytokine immune factors, including interferon-γ (IFN-γ) tumor necrosis factor-α (TNF-α) and interleukin-1 (IL-1).++ Our findings validate that the successful design of PLGA@Icaritin, which can effectively active ICD and facilitate tumor recruitment in GC through inducing mitoDNA oxidative damage.
Collapse
Affiliation(s)
- Yao Xiao
- Department of Oncology, Innovation centre for Advanced Interdisciplinary Medicine, Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510700, China
| | - Wenxia Yao
- Department of Oncology, Innovation centre for Advanced Interdisciplinary Medicine, Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510700, China
| | - Mingzhen Lin
- Department of Oncology, Innovation centre for Advanced Interdisciplinary Medicine, Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510700, China
| | - Wei Huang
- Department of Oncology, Innovation centre for Advanced Interdisciplinary Medicine, Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510700, China
| | - Ben Li
- Department of Oncology, Innovation centre for Advanced Interdisciplinary Medicine, Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510700, China
| | - Bin Peng
- Department of Oncology, Innovation centre for Advanced Interdisciplinary Medicine, Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510700, China
| | - Qinhai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510062, China
| | - Xinke Zhou
- Department of Oncology, Innovation centre for Advanced Interdisciplinary Medicine, Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510700, China
| | - Min Liang
- Department of Oncology, Innovation centre for Advanced Interdisciplinary Medicine, Guangzhou Key Laboratory of Enhanced Recovery after Abdominal Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510700, China
| |
Collapse
|
8
|
Shanmugavadivu A, Balagangadharan K, Selvamurugan N. Angiogenic and Osteogenic Effects of Flavonoids in Bone Regeneration. Biotechnol Bioeng 2022; 119:2313-2330. [PMID: 35718883 DOI: 10.1002/bit.28162] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 11/10/2022]
Abstract
Bone is a highly vascularised tissue that relies on a close spatial and temporal interaction between blood vessels and bone cells. As a result, angiogenesis is critical for bone formation and healing. The vascular system supports bone regeneration by delivering oxygen, nutrients, and growth factors, as well as facilitating efficient cell-cell contact. Most clinical applications of engineered bone grafts are hampered by insufficient vascularization after implantation. Over the last decade, a number of flavonoids have been reported to have osteogenic-angiogenic potential in bone regeneration because of their excellent bioactivity, low cost, availability, and minimal in vivo toxicity. During new bone formation, the osteoinductive nature of certain flavonoids is involved in regulating multiple signaling pathways contributing toward the osteogenic-angiogenic coupling. This review briefly outlines the osteogenic-angiogenic potential of those flavonoids and the mechanisms of their action in promoting bone regeneration. However, further studies are needed to investigate their delivery strategies and establish their clinical efficacy. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Abinaya Shanmugavadivu
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - K Balagangadharan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| |
Collapse
|
9
|
Li K, Xiao K, Zhu S, Wang Y, Wang W. Chinese Herbal Medicine for Primary Liver Cancer Therapy: Perspectives and Challenges. Front Pharmacol 2022; 13:889799. [PMID: 35600861 PMCID: PMC9117702 DOI: 10.3389/fphar.2022.889799] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/21/2022] [Indexed: 12/17/2022] Open
Abstract
Primary liver cancer (PLC) is one of the most common solid malignancies. However, PLC drug development has been slow, and first-line treatments are still needed; thus, studies exploring and developing alternative strategies for effective PLC treatment are urgently needed. Chinese herbal medicine (CHM) has long been applied in the clinic due to its advantages of low toxicity and targeting of multiple factors and pathways, and it has great potential for the development of novel natural drugs against PLC. Purpose: This review aims to provide an update on the pharmacological mechanisms of Chinese patent medicines (CPMs) and the latest CHM-derived compounds for the treatment of PLC and relevant clinical evaluations. Materials and Methods: A systematic search of English literature databases, Chinese literature, the Clinical Trials Registry Platform, and the Chinese Clinical Trial Registry for studies of CHMs for PLC treatment was performed. Results: In this review, we summarize the clinical trials and mechanisms of CPMs for PLC treatment that have entered the clinic with the approval of the Chinese medicine regulatory authority. These CPMs included Huaier granules, Ganfule granules, Fufang Banmao capsules, Jinlong capsules, Brucea javanica oil emulsions, and compound kushen injections. We also summarize the latest in vivo, in vitro, and clinical studies of CHM-derived compounds against PLC: icaritin and ginsenoside Rg3. Dilemmas facing the development of CHMs, such as drug toxicity and low oral availability, and future developments are also discussed. Conclusion: This review provides a deeper the understanding of CHMs as PLC treatments and provides ideas for the development of new natural drugs against PLC.
Collapse
Affiliation(s)
- Kexin Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Kunmin Xiao
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shijie Zhu
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Yong Wang, ; Wei Wang,
| | - Wei Wang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Institute of Prescription and Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provinvial Key Laboratory of TCM Pathogenesis and Prescriptions of Heart and Spleen Diseases, Guangzhou, China
- *Correspondence: Yong Wang, ; Wei Wang,
| |
Collapse
|
10
|
Zhang J, Liu Z, Luo Y, Li X, Huang G, Chen H, Li A, Qin S. The Role of Flavonoids in the Osteogenic Differentiation of Mesenchymal Stem Cells. Front Pharmacol 2022; 13:849513. [PMID: 35462886 PMCID: PMC9019748 DOI: 10.3389/fphar.2022.849513] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/16/2022] [Indexed: 01/02/2023] Open
Abstract
Mesenchymal stem cells (MSCs) play an important role in developing bone tissue engineered constructs due to their osteogenic and chondrogenic differentiation potential. MSC-based tissue engineered constructs are generally considered a safe procedure, however, the long-term results obtained up to now are far from satisfactory. The main causes of these therapeutic limitations are inefficient homing, engraftment, and directional differentiation. Flavonoids are a secondary metabolite, widely existed in nature and have many biological activities. For a long time, researchers have confirmed the anti-osteoporosis effect of flavonoids through in vitro cell experiments, animal studies. In recent years the regulatory effects of flavonoids on mesenchymal stem cells (MSCs) differentiation have been received increasingly attention. Recent studies revealed flavonoids possess the ability to modulate self-renewal and differentiation potential of MSCs. In order to facilitate further research on MSCs osteogenic differentiation of flavonoids, we surveyed the literature published on the use of flavonoids in osteogenic differentiation of MSCs, and summarized their pharmacological activities as well as the underlying mechanisms, aimed to explore their promising therapeutic application in bone disorders and bone tissue engineered constructs.
Collapse
Affiliation(s)
- Jinli Zhang
- Guangzhou Institute of Traumatic Surgery, Department of Orthopedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Zhihe Liu
- Guangzhou Institute of Traumatic Surgery, Department of Orthopedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Yang Luo
- School of Physical Education, Southwest University, Guangzhou, China
| | - Xiaojian Li
- Department of Burn and Plastic Surgery, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Guowei Huang
- Guangzhou Institute of Traumatic Surgery, Department of Orthopedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Huan Chen
- Guangzhou Institute of Traumatic Surgery, Department of Orthopedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Aiguo Li
- Guangzhou Institute of Traumatic Surgery, Department of Orthopedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| | - Shengnan Qin
- Guangzhou Institute of Traumatic Surgery, Department of Orthopedics, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, China
| |
Collapse
|
11
|
He X, Liao Y, Liu J, Sun S. Research Progress of Natural Small-Molecule Compounds Related to Tumor Differentiation. Molecules 2022; 27:2128. [PMID: 35408534 PMCID: PMC9000768 DOI: 10.3390/molecules27072128] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 11/25/2022] Open
Abstract
Tumor differentiation is a therapeutic strategy aimed at reactivating the endogenous differentiation program of cancer cells and inducing cancer cells to mature and differentiate into other types of cells. It has been found that a variety of natural small-molecule drugs can induce tumor cell differentiation both in vitro and in vivo. Relevant molecules involved in the differentiation process may be potential therapeutic targets for tumor cells. Compared with synthetic drugs, natural small-molecule antitumor compounds have the characteristics of wide sources, structural diversity and low toxicity. In addition, natural drugs with structural modification and transformation have relatively concentrated targets and enhanced efficacy. Therefore, using natural small-molecule compounds to induce malignant cell differentiation represents a more targeted and potential low-toxicity means of tumor treatment. In this review, we focus on natural small-molecule compounds that induce differentiation of myeloid leukemia cells, osteoblasts and other malignant cells into functional cells by regulating signaling pathways and the expression of specific genes. We provide a reference for the subsequent development of natural small molecules for antitumor applications and promote the development of differentiation therapy.
Collapse
Affiliation(s)
- Xiaoli He
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China; (X.H.); (Y.L.)
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha 410078, China
| | - Yongkang Liao
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China; (X.H.); (Y.L.)
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha 410078, China
| | - Jing Liu
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China; (X.H.); (Y.L.)
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha 410078, China
| | - Shuming Sun
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha 410078, China; (X.H.); (Y.L.)
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha 410078, China
| |
Collapse
|
12
|
Cheng T, Cao J, Jiang X, Yarmolenko M, Rogachev A, Rogachev A. Study of Icaritin Films by Low-Energy Electron Beam Deposition. EURASIAN CHEMICO-TECHNOLOGICAL JOURNAL 2021. [DOI: 10.18321/ectj1077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In this paper, icaritin film was prepared by low-energy beam electron beam deposition (EBD). The material test showed that the structure and composition of icaritin were not changed after electron beam deposition. Then, the film was sliced and immersed in simulated body fluids, it can be seen that the film was released quickly in the first 7 days. With the extension of soaking time, the release rate gradually slowed down, and the release amount exceeded 90% in about 20 days. In vitro cytotoxicity test showed that the relative cell viability rate of the film was still 92.32±1.30% (p<0.05), indicating that the film possessed excellent cytocompatibility.
Collapse
|
13
|
Wu Y, Yang Y, Li F, Zou J, Wang YH, Xu MX, Wang YL, Li RX, Sun YT, Lu S, Zhang YY, Sun XD. Icaritin Attenuates Lipid Accumulation by Increasing Energy Expenditure and Autophagy Regulated by Phosphorylating AMPK. J Clin Transl Hepatol 2021; 9:373-383. [PMID: 34221923 PMCID: PMC8237137 DOI: 10.14218/jcth.2021.00050] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND AIMS Lipid accumulation is the major characteristic of non-alcoholic fatty liver disease, the prevalence of which continues to rise. We aimed to investigate the effects and mechanisms of icaritin on lipid accumulation. METHODS Cells were treated with icaritin at 0.7, 2.2, 6.7, or 20 µM for 24 h. The effects on lipid accumulation in L02 and Huh-7 cells were detected by Bodipy and oil red O staining, respectively. Mitochondria biogenesis of L02 cells was detected by MitoTracker Orange staining. Glucose uptake and adenosine triphosphate content of 3T3-L1 adipocytes and C2C12 myotubes were detected. The expression levels of proteins in the adenosine 5'-monophosphate-activated protein kinase (AMPK) signaling pathway, biomarkers of autophagy, and mitochondria biogenesis were measured by western blotting. LC3 puncta were detected by immunofluorescence. RESULTS Icaritin significantly attenuated lipid accumulation in L02 and Huh-7 cells and boosted the mitochondria biogenesis of L02 cells. Icaritin enhanced glucose uptake, decreased adenosine triphosphate content, and activated the AMPK signaling pathway in 3T3-L1 adipocytes and C2C12 myotubes. Icaritin boosted autophagy and also enhanced the initiation of autophagic flux in 3T3-L1 preadipocytes and C2C12 myoblasts. However, icaritin decreased autophagy and promoted mitochondria biogenesis in 3T3-L1 adipocytes and C2C12 myotubes. CONCLUSIONS Icaritin attenuates lipid accumulation by increasing energy expenditure and regulating autophagy by activating the AMPK pathway.
Collapse
Affiliation(s)
- Yue Wu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Ying Yang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Fang Li
- Department of Medical Oncology, Sichuan Cancer Hospital & Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jie Zou
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Yu-Hao Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Meng-Xia Xu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Yong-Lun Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Rui-Xi Li
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Yu-Ting Sun
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Shun Lu
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Department of Radiological Protection, Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
| | - Yuan-Yuan Zhang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
- Department of Gastroenterology, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
- Correspondence to: Yuan-Yuan Zhang, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China. ORCID: http://orcid.org/0000-0002-9263-6262. Tel: +86-28-8550-1278, Fax: +86-28-8550-1278, E-mail: , ; Xiao-Dong Sun, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China. ORCID: http://orcid.org/0000-0002-7062-8931. Tel: +86-28-8550-1278, Fax: +86-28-8550-1278, E-mail:
| | - Xiao-Dong Sun
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
- Correspondence to: Yuan-Yuan Zhang, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China. ORCID: http://orcid.org/0000-0002-9263-6262. Tel: +86-28-8550-1278, Fax: +86-28-8550-1278, E-mail: , ; Xiao-Dong Sun, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China. ORCID: http://orcid.org/0000-0002-7062-8931. Tel: +86-28-8550-1278, Fax: +86-28-8550-1278, E-mail:
| |
Collapse
|
14
|
Sharifi S, Moghaddam FA, Abedi A, Maleki Dizaj S, Ahmadian S, Abdolahinia ED, Khatibi SMH, Samiei M. Phytochemicals impact on osteogenic differentiation of mesenchymal stem cells. Biofactors 2020; 46:874-893. [PMID: 33037744 DOI: 10.1002/biof.1682] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 12/19/2022]
Abstract
Medicinal plants have always been utilized for the prevention and treatment of the spread of different diseases all around the world. To name some traditional medicine that has been used over centuries, we can refer to phytochemicals such as naringin, icariin, genistein, and resveratrol gained from plants. Osteogenic differentiation and mineralization of stem cells can be the result of specific bioactive compounds from plants. One of the most appealing choices for therapy can be mesenchymal stem cells (MSCs) because it has a great capability of self-renewal and differentiation into three descendants, namely, endoderm, mesoderm, and ectoderm. Stem cell gives us the glad tidings of great advances in tissue regeneration and transplantation field for treatment of diseases. Using plant bioactive phytochemicals also holds tremendous promises in treating diseases such as osteoporosis. The purpose of the present review article thus is to investigate what are the roles and consequences of phytochemicals on osteogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Simin Sharifi
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Atefeh Abedi
- Department of Endodontics, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Maleki Dizaj
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahin Ahmadian
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Elaheh Dalir Abdolahinia
- Research Center of Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Samiei
- Department of Endodontics, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
15
|
Sareethammanuwat M, Boonyuen S, Arpornmaeklong P. Effects of beta-tricalcium phosphate nanoparticles on the properties of a thermosensitive chitosan/collagen hydrogel and controlled release of quercetin. J Biomed Mater Res A 2020; 109:1147-1159. [PMID: 32985073 DOI: 10.1002/jbm.a.37107] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/22/2020] [Accepted: 09/26/2020] [Indexed: 12/11/2022]
Abstract
In the present study, an inorganic matrix of beta-tricalcium phosphate (bTCP) nanoparticles and quercetin was incorporated into an organic matrix of 2:1 (w/w) chitosan/collagen composite to fabricate thermosensitive bTCP-chitosan/collagen-quercetin hydrogels. A sol-gel transition of the hydrogels was stimulated by beta-glycerophosphate (bGP) and temperature changes at physiological temperature and pH levels. Thereafter, the effects of 1%-3% (w/v) bTCP on properties of the bTCP-bGP-2:1 (w/w) chitosan/collagen hydrogels were investigated. Notably, the incorporation of 1%-3% (w/v) bTCP in the hydrogels did not interfere with the gelation process and time of the hydrogels at physiological temperature and pH levels. The bTCP-hydrogels exhibited a porous structure, interconnecting pore architecture, and median pore size of 100-200 μm. The incorporation of 3% bTCP increased the mechanical strength but decreased the swelling and degradation rates, pore size, permeability, and quercetin release rate of the hydrogels. The hydrogels were noncytotoxic and able to support cell encapsulation. A sustained quercetin release profile of the 3% bTCP-hydrogel further suggested the applicability of the hydrogel as a delivery vehicle of natural flavonoids for bone regeneration.
Collapse
Affiliation(s)
- Maytha Sareethammanuwat
- Master of Science Program in Dental Implantology, Faculty of Dentistry, Thammasat University Rangsit campus, Pathum Thani, Thailand
| | - Supakorn Boonyuen
- Department of Chemistry, Faculty of Science and Technology, Thammasat University Rangsit campus, Pathum Thani, Thailand
| | - Premjit Arpornmaeklong
- Oral and Maxillofacial Surgery Division, Faculty of Dentistry, Thammasat University Rangsit campus, Pathum Thani, Thailand
| |
Collapse
|
16
|
Lu Q, Zhu H, Liu X, Tang C. Icariin sustains the proliferation and differentiation of Aβ 25-35-treated hippocampal neural stem cells via the BDNF-TrkB-ERK/Akt signaling pathway. Neurol Res 2020; 42:936-945. [PMID: 32727295 DOI: 10.1080/01616412.2020.1792701] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Icariin (ICA) can be potentially used to treat Alzheimer's disease (AD), but the mechanism was not clear. The current study explored the effects of ICA on hippocampal neural stem cells, aiming to provide a comprehensive basis for its clinical application. METHODS Hippocampal neural stem cells were isolated from newborn rats and their differentiation ability was evaluated by performing immunofluorescence staining. Next, Aβ cell model was constructed by treating the cells with Aβ25-35, and then the model was further treated by ICA or shBDNF or the two in combination. The viability and differentiation of the cells were, respectively, analyzed by 3-(4, 5-Dimethylthiazol-2-yl)-2, 5-Diphenyltetrazolium Bromide (MTT) and flow cytometry. The expression of BDNF-TrkB-ERK/Akt signaling pathway was assessed by quantitative real-time polymerase chain reaction (qRT-PCR) or Western blot (WB). RESULTS The hippocampal neural stem cells can differentiate into neurons and astrocytes. ICA effectively promoted the viability and differentiation of Aβ cell models. The expression levels of BDNF and TrkB in Aβ cell models were obviously decreased, which were noticeably increased by ICA. Moreover, BDNF knockdown further inhibited the viability and differentiation of Aβ model cells, which could be reversed by ICA. BDNF knockdown not only suppressed the expressions of BDNF and TrkB in Aβ cell models but also effectively prevented the phosphorylation of ERK/Akt; however, these phenomena were significantly alleviated by ICA treatment. DISCUSSION ICA promoted the proliferation and differentiation of Aβ25-35-treated hippocampal neural stem cells through BDNF-TrkB-ERK/Akt signaling pathway. The current findings might contribute to the treatment of AD.
Collapse
Affiliation(s)
- Quan Lu
- Department of Neurology, Jingmen No.1 People's Hospital , Jingmen, Hubei, China
| | - Hailing Zhu
- Department of Emergency, Jingmen No.1 People's Hospital , Jingmen, Hubei, China
| | - Xuejiao Liu
- Department of Urology, Jingmen No.1 People's Hospital , Jingmen, Hubei, China
| | - Congfeng Tang
- Department of Neurology, Jingmen No.1 People's Hospital , Jingmen, Hubei, China
| |
Collapse
|
17
|
Icariin/Aspirin Composite Coating on TiO2 Nanotubes Surface Induce Immunomodulatory Effect of Macrophage and Improve Osteoblast Activity. COATINGS 2020. [DOI: 10.3390/coatings10040427] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Surface coating modification of titanium-based alloys is an efficient way to accelerate early osseointegration in dental implant fields. Icariin (ICA) is a traditional Chinese medicine that has bone activating functions, while aspirin (ASP) is a classical non-steroidal anti-inflammatory drug with good antipyretic and analgesic capabilities. Moreover, poly(lactic–co–glycolic acid) (PLGA) has attracted great attention due to its excellent biocompatibility and biodegradability. We superimposed an ASP/PLGA coating onto ICA loaded TiO2 nanotubes structure so as to establish an icariin/aspirin composite coating on TiO2 nanotubes surface. Scanning electron microscopy, X-ray photoelectron spectroscopy, a contact angle test and a drug release test confirmed the successful preparation of the NT–ICA–ASP/PLGA substrate, with a sustained release pattern of both ICA and ASP. Compared to those cultured on the Ti surface, macrophage cells on the NT-ICA-ASP/PLGA substrate displayed decreased M1 proinflammatory and enhanced M2 proregenerative genes and proteins expression, which implied activated immunomodulatory effect. Moreover, when cultured with conditioned medium from macrophages, osteoblast cells on the NT-ICA-ASP/PLGA substrate revealed improved cell proliferation, adhesion and osteogenic genes and proteins expression, compared with those on the Ti surface. The abovementioned results suggest that the established NT-ICA-ASP/PLGA substrate is a promising candidate for functionalized coating material in Ti implant surface modification.
Collapse
|
18
|
Agbeboh NI, Oladele IO, Daramola OO, Adediran AA, Olasukanmi OO, Tanimola MO. Environmentally sustainable processes for the synthesis of hydroxyapatite. Heliyon 2020; 6:e03765. [PMID: 32368642 PMCID: PMC7184159 DOI: 10.1016/j.heliyon.2020.e03765] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/15/2020] [Accepted: 04/06/2020] [Indexed: 11/03/2022] Open
Abstract
Hard tissue regeneration and regrowth have continued to be a challenge in the field of conventional medicine in this 21st century. Over the years, the regrowth of broken bones and diseased hard tissue has remained a major concern in medical research. Since the discovery of hydroxyapatite (HA), a bioceramic compound that possesses the ability to activate bone regrowth and bond directly with regenerated bone, it has subsequently become an indispensable biomaterial. Currently, it is being used across the medical fields due to its exceptional biocompatibility. This became plausible because the main mineral phase of mammalian bones is HA. It has found application in various medical fields like medical instruments, drug delivery, bone and tooth fillers, prosthetics, orthotics, and in-vitro implants. As the importance of HA geometrically increases, it is necessary to critically evaluate and propose the most economic process of synthesizing and manufacturing this important bioceramic material. This review, therefore, highlights the different sources of HA and the synthesis/production methods for each source with a strong emphasis on the environment. Thus, the appraisal was carried out based on the properties of the derived HA. Such properties include but are not limited to geometry, particle size, morphology, thermal stability, and stoichiometry to suggest the most economic and environmentally sustainable sources and processing routes.
Collapse
Affiliation(s)
- N I Agbeboh
- Department of Metallurgical and Materials Engineering, Federal University of Technology, Akure, Ondo State, Nigeria.,Department of Mechanical and Mechatronics Engineering, Federal University Otuoke, Ogbia, Bayelsa State, Nigeria
| | - I O Oladele
- Department of Metallurgical and Materials Engineering, Federal University of Technology, Akure, Ondo State, Nigeria
| | - O O Daramola
- Department of Metallurgical and Materials Engineering, Federal University of Technology, Akure, Ondo State, Nigeria.,Institute of Nano-Engineering Research (INER), Department of Chemical, Metallurgical and Materials Engineering (Polymer Division), Tshwane University of Technology, Pretoria, South Africa
| | - A A Adediran
- Department of Mechanical Engineering, Landmark University, Omuaran, Kwara State, Nigeria
| | - O O Olasukanmi
- Department of Industrial Chemistry, Federal University of Technology, Akure, Ondo State, Nigeria
| | - M O Tanimola
- Department of Civil Engineering, Federal University of Technology, Akure, Ondo State, Nigeria
| |
Collapse
|
19
|
Wei Q, Wang B, Hu H, Xie C, Ling L, Gao J, Cao Y. Icaritin promotes the osteogenesis of bone marrow mesenchymal stem cells via the regulation of sclerostin expression. Int J Mol Med 2020; 45:816-824. [PMID: 31985018 PMCID: PMC7015123 DOI: 10.3892/ijmm.2020.4470] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 09/30/2019] [Indexed: 12/11/2022] Open
Abstract
Icaritin, a metabolite of icariin, is a potent promoter of bone marrow-derived mesenchymal stem cells (BMSCs) osteogenesis, but the underlying mechanisms remain unclear. To examine the effects of icaritin on osteogenic differentiation, BMSCs were exposed to osteogenic induction medium with or without icaritin pretreatment in the present study. It was identified that icaritin (0.01-1 µM) exhibited no cytotoxicity on the proliferative abilities of the BMSCs. Icaritin at 1 µM increased alkaline phosphatase activity, mineral deposition and osteoblast-specific gene expression. Treatment with 1 µM Icaritin upregulated osteocalcin, RUNX family transcription factor 2, tissue-nonspecific alkaline phosphatase and β-catenin, and suppressed sclerostin (SOST) gene expression in different stages of osteogenic differentiation. It was also demonstrated that SOST over-expression inhibited icaritin-induced osteogenesis. The western blot analysis data suggested that ICI 182780, which causes estrogen receptor α (ERα) degradation, reversed the icaritin-induced decrease in SOST expression, which was inconsistent with the results of immunofluorescence analysis. In conclusion, icaritin was demonstrated to promote the osteogenesis of hBMSCs by downregulating SOST expression, and icaritin-induced suppression of SOST was regulated in part via the Wnt/β-catenin/ERα axis.
Collapse
Affiliation(s)
- Qiushi Wei
- Hip Preserving Ward, No. 3 Orthopedic Region, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510407, P.R. China
| | - Bin Wang
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Hailan Hu
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Chuhai Xie
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Long Ling
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Jianliang Gao
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| | - Yanming Cao
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510260, P.R. China
| |
Collapse
|
20
|
Ning Y, Qin W, Ren Y, Li C, Chen W, Zhao H. [Effect of icariin/attapulgite/collagen type Ⅰ/polycaprolactone composite scaffold in repair of rabbit tibia defect]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:1181-1189. [PMID: 31512463 PMCID: PMC8355846 DOI: 10.7507/1002-1892.201902044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 07/11/2019] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To investigate the effect of icarin/attapulgite/collagen type Ⅰ/polycaprolactone (ICA/ATP/Col Ⅰ/PCL) composite scaffold in repair of rabbit tibia defect. METHODS The ICA/20%ATP/Col Ⅰ/PCL (scaffold 1), ICA/30%ATP/Col Ⅰ/PCL (scaffold 2), 20%ATP/Col Ⅰ/PCL (scaffold 3), and 30%ATP/Col Ⅰ/PCL (scaffold 4) composite scaffolds were constructed by solution casting-particle filtration method. The structure characteristics of the scaffold 2 before and after cross-linking were observed by scanning electron microscopy, and the surface contact angles of the scaffold 2 and the scaffold 4 were used to evaluate the water absorption performance of the material. The in vitro degradation test was used to evaluate the sustained-release effect of the scaffold 2. Thirty male Japanese white rabbits, weighing (2.0±0.1) kg, were randomly divided into groups A, B, C, D, and E, 6 in each group. After making a 1 cm- diameter bilateral tibial defects model, group A was the defect control group without any material implanted. Groups B, C, D, and E were implanted with scaffolds 3, 4, 1, and 2 at the defect sites, respectively. At 4, 8, and 12 weeks after operation, the repairing effects of 4 scaffolds were observed by gross observation, histological observation of HE and Masson staining, and immunohistochemical staining of osteogenic specific transcription factor (runt-related transcription factor 2, RUNX2), osteogenic related transcription factor [Osterix (OSX), Col Ⅰ, osteopontin (OPN)]. RESULTS Scanning electron microscopy observation showed that the scaffolds were all porous. The structure of the material was loose before and after cross-linking. The surface contact angle showed that the scaffold was hydrophobic, and the scaffold 2 was more hydrophobic than scaffold 4. The sustained-release effect in vitro showed that the drug could be released in a micro and long-term manner. In the animal implantation experiment, the gross observation showed that the defects were significantly smaller in groups D and E than in groups A, B, and C at 4 and 12 weeks after operation. HE and Masson staining showed that the defect of group A was full of connective tissue at 4 weeks after operation, a large number of fibers were seen in groups B and C, and the new bone formation was observed in groups D and E. The increase of new bone was observed in each group at 8 weeks after operation. The defect of group A was still dominated by connective tissue at 12 weeks after operation, and a small amount of new bone tissue was observed in groups B and C, and a large number of new bone tissue was observed in groups D and E, especially in group E, and most of the materials degraded. Immunohistochemical staining showed that the expressions of RUNX2 and OSX in the new tissues of groups D and E were significantly higher than those of the other groups at 4 weeks after operation. The expression of RUNX2 decreased at 8 and 12 weeks after operation. After 8 weeks and 12 weeks, the expressions of Col Ⅰand OPN increased than in 4 weeks. And the expressions of Col Ⅰ and OPN in the new tissues of groups D and E were significantly more than those of the other groups. CONCLUSION ICA/ATP/Col I/PCL composite scaffolds have good porosity and biocompatibility, can promote bone formation, and have good bone regeneration and repair effect.
Collapse
Affiliation(s)
- Yu Ning
- School of Basic Medical Sciences, Gansu University of Traditional Chinese Medicine, Lanzhou Gansu, 730000, P.R.China;Department of Orthopaedic Laboratory, Changzhou Second People's Hospital, Changzhou Jiangsu, 213000, P.R.China
| | - Wen Qin
- Department of Orthopaedic Laboratory, Changzhou Second People's Hospital, Changzhou Jiangsu, 213000, P.R.China
| | - Yahui Ren
- Department of Orthopaedic Laboratory, Changzhou Second People's Hospital, Changzhou Jiangsu, 213000, P.R.China
| | - Chenkai Li
- Department of Orthopaedic Laboratory, Changzhou Second People's Hospital, Changzhou Jiangsu, 213000, P.R.China
| | - Wenyang Chen
- Department of Orthopaedic Laboratory, Changzhou Second People's Hospital, Changzhou Jiangsu, 213000, P.R.China
| | - Hongbin Zhao
- School of Basic Medical Sciences, Gansu University of Traditional Chinese Medicine, Lanzhou Gansu, 730000, P.R.China;Department of Orthopaedic Laboratory, Changzhou Second People's Hospital, Changzhou Jiangsu, 213000,
| |
Collapse
|
21
|
Liu Y, Mi B, Lv H, Liu J, Xiong Y, Hu L, Xue H, Panayi AC, Liu G, Zhou W. Shared KEGG pathways of icariin-targeted genes and osteoarthritis. J Cell Biochem 2019; 120:7741-7750. [PMID: 30506715 DOI: 10.1002/jcb.28048] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 10/22/2018] [Indexed: 01/24/2023]
Abstract
The beneficial effects of icariin in the management of many diseases, such as chronic renal failure and heart failure, are well known. Icariin has also been shown to ameliorate osteoarthritis (OA) symptoms; however, the underlying mechanisms remain unclear. In this study, a bioinformatics analysis was performed to investigate the KEGG pathways of icariin-targeted genes involved in OA. Our study suggests that icariin plays a role in OA by regulating inflammatory cytokine production, insulin resistance, and cell survival through modulation of the NF-κB, MAPK, and Akt signaling pathways. Importantly, IKBKB, NFKBIA, MAPK8, MAPK9, and MAPK10 may be the hub genes affected by icariin when providing its beneficial effects on OA. In addition, we found that icariin decreases proinflammatory factors and inhibits chondrocyte apoptosis through suppression of the NF-κB pathway. Our study highlights a set of KEGG pathways that could explain the molecular mechanism of icariin's action on OA, suggesting that icariin could be considered as a promising therapeutic option for OA.
Collapse
Affiliation(s)
- Yi Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bobin Mi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Huijuan Lv
- Department of Rheumatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jing Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liangcong Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hang Xue
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Adriana C Panayi
- Department of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Guohui Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wu Zhou
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Mechanism of Action of Icariin in Bone Marrow Mesenchymal Stem Cells. Stem Cells Int 2019; 2019:5747298. [PMID: 31089330 PMCID: PMC6476003 DOI: 10.1155/2019/5747298] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/28/2019] [Accepted: 03/12/2019] [Indexed: 12/19/2022] Open
Abstract
Osteoporosis, femoral head necrosis, and congenital bone defects are orthopedic disorders characterized by reduced bone generation and insufficient bone mass. Bone regenerative therapy primarily relies on the bone marrow mesenchymal stem cells (BMSCs) and their ability to differentiate osteogenically. Icariin (ICA) is the active ingredient of Herba epimedii, a common herb used in traditional Chinese medicine (TCM) formulations, and can effectively enhance BMSC proliferation and osteogenesis. However, the underlying mechanism of ICA action in BMSCs is not completely clear. In this review, we provide an overview of the studies on the role and mechanism of action of ICA in BMSCs, to provide greater insights into its potential clinical use in bone regeneration.
Collapse
|
23
|
|
24
|
Icaritin: A Novel Natural Candidate for Hematological Malignancies Therapy. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4860268. [PMID: 31032347 PMCID: PMC6458936 DOI: 10.1155/2019/4860268] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/05/2019] [Accepted: 03/13/2019] [Indexed: 01/14/2023]
Abstract
Hematological malignancies including leukemia and lymphoma can severely impact human health. With the current therapies combined with chemotherapy, stem cell transplantation, radiotherapy, and immunotherapy, the prognosis of hematologic malignancies improved significantly. However, most hematological malignancies are still incurable. Therefore, research for novel treatment options was continuing with the natural product as one source. Icaritin is a compound extracted from a traditional Chinese herb, Epimedium Genus, and demonstrated an antitumor effect in various neoplasms including hematological malignancies such as leukemia, lymphoma, and multiple myeloma. In hematological malignancies, icaritin showed multiple cytotoxic effects to induce apoptosis, arrest the cell cycle, inhibit proliferation, promote differentiation, restrict metastasis and infiltration, and suppress the oncogenic virus. The proved underlying mechanisms of the cytotoxic effects of icaritin are different in various cell types of hematological malignancies but associated with the critical cell signal pathway, including PI3K/Akt, JAK/STAT3, and MAPK/ERK/JNK. Although the primary target of icaritin is still unspecified, the existing evidence indicates that icaritin is a potential novel therapeutic agent for neoplasms as with hematological malignancies. Here, in the field of hematology, we reviewed the reported activity of icaritin in hematologic malignancies and the underlying mechanisms and recognized icaritin as a candidate for therapy of hematological malignancies.
Collapse
|
25
|
Arumugam B, Balagangadharan K, Selvamurugan N. Syringic acid, a phenolic acid, promotes osteoblast differentiation by stimulation of Runx2 expression and targeting of Smad7 by miR-21 in mouse mesenchymal stem cells. J Cell Commun Signal 2018; 12:561-573. [PMID: 29350343 PMCID: PMC6039342 DOI: 10.1007/s12079-018-0449-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 01/10/2018] [Indexed: 01/10/2023] Open
Abstract
Syringic acid (SA), a phenolic acid, has been used in Chinese and Indian medicine for treating diabetes but its role in osteogenesis has not yet been investigated. In the present study, at the molecular and cellular levels, we evaluated the effects of SA on osteoblast differentiation. At the cellular level, there was increased alkaline phosphatase (ALP) activity and calcium deposition by SA treatment in mouse mesenchymal stem cells (mMSCs). At the molecular level, SA treatment of these cells stimulated expression of Runx2, a bone transcription factor, and of osteoblast differentiation marker genes such as ALP, type I collagen, and osteocalcin. It is known that Smad7 is an antagonist of TGF-β/Smad signaling and is a negative regulator of Runx2. microRNAs (miRNAs) play a key role in the regulation of osteogenesis genes at the post-transcriptional level and studies have reported that Smad7 is one of the target genes of miR-21. We found that there was down regulation of Smad7 and up regulation of miR-21 in SA-treated mMSCs. We further identified that the 3'-untranslated region (UTR) of Smad7 was directly targeted by miR-21 in these cells. Thus, our results suggested that SA promotes osteoblast differentiation via increased expression of Runx2 by miR-21-mediated down regulation of Smad7. Hence, SA may have potential in orthopedic applications.
Collapse
Affiliation(s)
- B Arumugam
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603 203, India
| | - K Balagangadharan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603 203, India
| | - N Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603 203, India.
| |
Collapse
|
26
|
IO O, OG A, OG O, AO B, MO P. Non-synthetic sources for the development of hydroxyapatite. ACTA ACUST UNITED AC 2018. [DOI: 10.15406/jabb.2018.05.00122] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
27
|
Zhao H, Zhao N, Zheng P, Xu X, Liu M, Luo D, Xu H, Ju D. Prevention and Treatment of Osteoporosis Using Chinese Medicinal Plants: Special Emphasis on Mechanisms of Immune Modulation. J Immunol Res 2018; 2018:6345857. [PMID: 29675436 PMCID: PMC5838472 DOI: 10.1155/2018/6345857] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 11/05/2017] [Accepted: 12/06/2017] [Indexed: 01/18/2023] Open
Abstract
Numerous studies have examined the pathogenesis of osteoporosis. The causes of osteoporosis include endocrine factors, nutritional status, genetic factors, physical factors, and immune factors. Recent osteoimmunology studies demonstrated that the immune system and immune factors play important regulatory roles in the occurrence of osteoporosis, and people should pay more attention to the relationship between immunity and osteoporosis. Immune and bone cells are located in the bone marrow and share numerous regulatory molecules, signaling molecules, and transcription factors. Abnormal activation of the immune system alters the balance between osteoblasts and osteoclasts, which results in an imbalance of bone remodeling and osteoporosis. The incidence of osteoporosis is also increasing with the aging of China's population, and traditional Chinese medicine has played a vital role in the prevention and treatment of osteoporosis for centuries. Chinese medicinal plants possess unique advantages in the regulation of the immune system and the relationships between osteoporosis and the immune system. In this review, we provide a general overview of Chinese medicinal plants in the prevention and treatment of osteoporosis, focusing on immunological aspects.
Collapse
Affiliation(s)
- Hongyan Zhao
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Science, Beijing 100700, China
| | - Ning Zhao
- Institute of Clinical Basic Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Peng Zheng
- Jilin Provincial Hospital of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Xiaohong Xu
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Meijie Liu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Science, Beijing 100700, China
| | - Dan Luo
- Traditional Chinese Medicine Hospital of Changping District, Beijing 102200, China
| | - Huihui Xu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Dahong Ju
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Science, Beijing 100700, China
| |
Collapse
|
28
|
Icariin Regulates the Bidirectional Differentiation of Bone Marrow Mesenchymal Stem Cells through Canonical Wnt Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:8085325. [PMID: 29445413 PMCID: PMC5763109 DOI: 10.1155/2017/8085325] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/28/2017] [Accepted: 09/12/2017] [Indexed: 01/13/2023]
Abstract
Fat infiltration within the bone marrow is easily observed in some postmenopausal women. Those fats are mainly derived from bone marrow mesenchymal stem cells (BMMSCs). The increment of adipocytes derived from BMMSCs leads to decreased osteoblasts derived from BMMSCs, so the bidirectional differentiation of BMMSCs significantly contributes to osteoporosis. Icariin is the main extractive of Herba Epimedii which is widely used in traditional Chinese medicine. In this experiment, we investigated the effect of icariin on the bidirectional differentiation of BMMSCs through quantitative real-time PCR, immunofluorescence, western blot, and tissue sections in vitro and in vivo. We found that icariin obviously promotes osteogenesis and inhibits adipogenesis through detecting staining and gene expression. Micro-CT analysis showed that icariin treatment alleviated the loss of cancellous bone of the distal femur in ovariectomized (OVX) mice. H&E staining analysis showed that icariin-treated OVX mice obtained higher bone mass and fewer bone marrow lipid droplets than OVX mice. Western blot and immunofluorescence showed that icariin regulates the bidirectional differentiation of BMMSCs via canonical Wnt signaling. This study demonstrates that icariin exerts its antiosteoporotic effect by regulating the bidirectional differentiation of BMMSCs through the canonical Wnt signaling pathway.
Collapse
|