1
|
Ma L, Zhang H, Liu Y, Liu Y, Zhou J, Yang H, Xu B. Exploring the Molecular Mechanisms of Fisetin in Treating Periodontitis Through Multiomics and Network Pharmacology. Int Dent J 2025; 75:2204-2221. [PMID: 39755534 DOI: 10.1016/j.identj.2024.12.012] [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: 09/29/2024] [Revised: 11/20/2024] [Accepted: 12/08/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Periodontitis (PD) is a common chronic inflammatory oral disease that severely affects patients' quality of life. Fisetin has been shown to possess antioxidant and anti-inflammatory properties in various biological systems. METHODS This study first identified the molecular targets of fisetin for PD through network pharmacology analysis. The therapeutic effects of fisetin were then evaluated in an animal model of PD and validated through in vitro experiments. Additionally, we utilised single-cell and spatial transcriptomics technologies to identify key cell populations in PD and their spatial distribution. RESULTS The study demonstrated that fisetin significantly reduced alveolar bone destruction in the rat model of PD. Single-cell transcriptomics revealed that fisetin primarily affects fibroblast populations. In vitro experiments showed that fisetin alleviated the cytotoxicity caused by high oxidative stress levels in human periodontal ligament fibroblasts (PDLFs) . CONCLUSION Fisetin inhibits the progression of periodontitis by reducing oxidative stress levels in fibroblast populations. These findings support the potential of fisetin as a therapeutic agent for periodontitis and provide a scientific basis for future clinical trials and treatment strategies. CLINICAL RELEVANCE By significantly reducing alveolar bone destruction and modulating fibroblast function, fisetin presents a novel therapeutic strategy for managing periodontitis. These results provide a scientific foundation for the design of clinical trials aimed at evaluating the efficacy of fisetin in PD patients. If validated in clinical settings, fisetin could be incorporated into treatment regimens, offering a pharmacological option that complements conventional periodontal therapies, thereby improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Lingzhi Ma
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China; Yunnan Key Laboratory of Stomatology, Kunming, China
| | - Hongrong Zhang
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China; Yunnan Key Laboratory of Stomatology, Kunming, China
| | - Yali Liu
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China; Yunnan Key Laboratory of Stomatology, Kunming, China
| | - Yan Liu
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China; Yunnan Key Laboratory of Stomatology, Kunming, China
| | - Jing Zhou
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China; Yunnan Key Laboratory of Stomatology, Kunming, China
| | - Hefeng Yang
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China; Yunnan Key Laboratory of Stomatology, Kunming, China.
| | - Biao Xu
- Affiliated Stomatology Hospital of Kunming Medical University, Kunming, China; Yunnan Key Laboratory of Stomatology, Kunming, China.
| |
Collapse
|
2
|
Messeha SS, Fidudusola FF, Gendy S, Latinwo LM, Odewumi CO, Soliman KFA. Nrf2 Activation as a Therapeutic Target for Flavonoids in Aging-Related Osteoporosis. Nutrients 2025; 17:267. [PMID: 39861398 PMCID: PMC11767473 DOI: 10.3390/nu17020267] [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: 12/22/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Biological aging is a substantial change that leads to different diseases, including osteoporosis (OP), a condition involved in loss of bone density, deterioration of bone structure, and increased fracture risk. In old people, there is a natural decline in bone mineral density (BMD), exacerbated by hormonal changes, particularly during menopause, and it continues in the early postmenopausal years. During this transition time, hormonal alterations are linked to elevated oxidative stress (OS) and decreased antioxidant defenses, leading to a significant increase in OP. Aging is significantly associated with an abnormal ratio of oxidant/antioxidant and modified nuclear factor erythroid-derived two related factor2 (Nrf2)/Kelch-like ECH-associated protein 1 (Keap1) pathway. OS adversely affects bone health by promoting osteoclastic (bone resorbing) activity and impairing osteoblastic (bone-forming cells). Nrf2 is critical in controlling OS and various cellular processes. The expression of Nrf2 is linked to multiple age-related diseases, including OP, and Nrf2 deficiency leads to unbalanced bone formation/resorption and a consequent decline in bone mass. Various drugs are available for treating OP; however, long-term uses of these medicines are implicated in diverse illnesses such as cancer, cardiovascular, and stroke. At the same time, multiple categories of natural products, in particular flavonoids, were proposed as safe alternatives with antioxidant activity and substantial anti-osteoporotic effects.
Collapse
Affiliation(s)
- Samia S. Messeha
- College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA; (S.S.M.); (F.F.F.); (L.M.L.)
- College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA
| | - Fidara F. Fidudusola
- College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA; (S.S.M.); (F.F.F.); (L.M.L.)
| | - Sherif Gendy
- School of Allied Health Sciences, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Lekan M. Latinwo
- College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA; (S.S.M.); (F.F.F.); (L.M.L.)
| | - Caroline O. Odewumi
- College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA; (S.S.M.); (F.F.F.); (L.M.L.)
| | - Karam F. A. Soliman
- College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA
| |
Collapse
|
3
|
Sakai E, Tsukuba T. Transcriptomic Characterization Reveals Mitochondrial Involvement in Nrf2/Keap1-Mediated Osteoclastogenesis. Antioxidants (Basel) 2024; 13:1575. [PMID: 39765903 PMCID: PMC11673794 DOI: 10.3390/antiox13121575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Although osteoclasts play crucial roles in the skeletal system, the mechanisms that underlie oxidative stress during osteoclastogenesis remain unclear. The transcription factor Nrf2 and its suppressor, Keap1, function as central mediators of oxidative stress. To further elucidate the function of Nrf2/Keap1-mediated oxidative stress regulation in osteoclastogenesis, DNA microarray analysis was conducted in this study using wild-type (WT), Keap1 knockout (Keap1 KO), and Nrf2 knockout (Nrf2 KO) osteoclasts. Principal component analysis showed that 403 genes, including Nqo1, Il1f9, and Mmp12, were upregulated in Keap1 KO compared with WT osteoclasts, whereas 24 genes, including Snhg6, Ccdc109b, and Wfdc17, were upregulated in Nrf2 KO compared with WT osteoclasts. Moreover, 683 genes, including Car2, Calcr, and Pate4, were upregulated in Nrf2 KO cells compared to Keap1 KO cells. Functional analysis by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis showed upregulated genes in Nrf2 KO osteoclasts were mostly enriched in oxidative phosphorylation. Furthermore, GeneMANIA predicted the protein-protein interaction network of novel molecules such as Rufy4 from genes upregulated in Nrf2 KO osteoclasts. Understanding the complex interactions between these molecules may pave the way for developing promising therapeutic strategies against bone metabolic diseases caused by increased osteoclast differentiation under oxidative stress.
Collapse
Affiliation(s)
- Eiko Sakai
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1, Sakamoto, Nagasaki 852-8588, Japan;
| | | |
Collapse
|
4
|
Sakai E, Saito M, Koyanagi Y, Takayama Y, Farhana F, Yamaguchi Y, Tsukuba T. Autophagy Regulator Rufy 4 Promotes Osteoclastic Bone Resorption by Orchestrating Cytoskeletal Organization via Its RUN Domain. Cells 2024; 13:1766. [PMID: 39513873 PMCID: PMC11545195 DOI: 10.3390/cells13211766] [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: 07/18/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Rufy4, a protein belonging to the RUN and FYVE domain-containing protein family, participates in various cellular processes such as autophagy and intracellular trafficking. However, its role in osteoclast-mediated bone resorption remains uncertain. In this study, we investigated the expression and role of the Rufy4 gene in osteoclasts using small interfering RNA (siRNA) transfection and gene overexpression systems. Our findings revealed a significant increase in Rufy4 expression during osteoclast differentiation. Silencing Rufy4 enhanced osteoclast differentiation, intracellular cathepsin K levels, and formation of axial protrusive structures but suppressed bone resorption. Conversely, overexpressing wild-type Rufy4 in osteoclasts hindered differentiation while promoting podosome formation and bone resorption. Similarly, overexpression of a Rufy4 variant lacking the RUN domain mimics the effects of Rufy4 knockdown, significantly increasing intracellular cathepsin K levels, promoting osteoclastogenesis, and elongated axial protrusions formation, yet inhibiting bone resorption. These findings indicate that Rufy4 plays a critical role in osteoclast differentiation and bone resorption by regulating the cytoskeletal organization through its RUN domain. Our study provides new insights into the molecular mechanisms governing osteoclast activity and underscores Rufy4's potential as a novel therapeutic target for bone disorders characterized by excessive bone resorption.
Collapse
Affiliation(s)
- Eiko Sakai
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan (Y.K.); (F.F.); (Y.Y.)
| | - Minoru Saito
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan (Y.K.); (F.F.); (Y.Y.)
- Kondou Dental Clinic, 1154-5 Oozujinnai, Kikuchi 869-1221, Japan
| | - Yu Koyanagi
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan (Y.K.); (F.F.); (Y.Y.)
- Department of Prosthetic Dentistry, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | - Yoshitsugu Takayama
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan (Y.K.); (F.F.); (Y.Y.)
- Ito Dental Clinic Medical Corporation, 3-2-4 Kousienn, Nishinomiya 663-8152, Japan
| | - Fatima Farhana
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan (Y.K.); (F.F.); (Y.Y.)
| | - Yu Yamaguchi
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan (Y.K.); (F.F.); (Y.Y.)
| | - Takayuki Tsukuba
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan (Y.K.); (F.F.); (Y.Y.)
| |
Collapse
|
5
|
Xu K, Ren X, Wang J, Zhang Q, Fu X, Zhang PC. Clinical development and informatics analysis of natural and semi-synthetic flavonoid drugs: A critical review. J Adv Res 2024; 63:269-284. [PMID: 37949300 PMCID: PMC11380023 DOI: 10.1016/j.jare.2023.11.007] [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: 08/03/2023] [Revised: 10/17/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Flavonoids are one of the most important metabolites with vast structural diversity and a plethora of potential pharmacological applications, which have drawn considerable attention in the laboratory. Nevertheless, it remains uncertain how many candidates were progressed to clinical application. AIM OF REVIEW We carried out a critical review of natural and semi-synthetic flavonoid drugs and candidates undergoing different clinical phases worldwide by applying an adequate search method and conducted a brief cheminformatic and bioinformatic analysis. It was expected that the obtained results might narrow the screening scope and reduce the cost of drug research and development. KEY SCIENTIFIC CONCEPTS OF REVIEW To our knowledge, this is the most systematic summarization of flavonoid-based drugs and clinical candidates to date. It was found that a total of 19 flavonoid-based drugs have been approved for the market, and of these, natural flavonoids accounted for 52.6%. Besides, a total of 36 flavonoid-based clinical candidates are undergoing or suspended in different phases, and of these, natural flavonoids account for 44.4%. Thus, natural flavonoids remain the best option for finding novel agents/active templates, and when investigated in conjunction with synthetic chemicals and biologicals, they offer the potential to discover novel structures that can lead to effective agents against a variety of human diseases. Additionally, flavonoid-based marketed drugs have been successful in cardiovascular treatment, and the related drugs account for more than 30% of marketed drugs. However, the use of flavonoids as antineoplastic and immunomodulating agents is not likely for approximately 50% of the candidates suspended in the clinical stage. Interestingly, the marketed drugs covered a broader range of chemical spaces based on size, polarity, and three-dimensional structure compared to the clinical candidates. In addition, flavonoid glycosides with poor oral bioavailability account for 36.8% of the marketed drugs, and thus, they could be thoroughly investigated.
Collapse
Affiliation(s)
- Kuo Xu
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Qingdao Academy of Chinese Medical Sciences Shandong University of Traditional Chinese Medicine, Qingdao Key Laboratory of Research in Marine Traditional Chinese Medicine, Qingdao Key Technology Innovation Center of Marine Traditional Chinese Medicine's Deep Development and Industrialization, Qingdao 266114, China
| | - Xia Ren
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Qingdao Academy of Chinese Medical Sciences Shandong University of Traditional Chinese Medicine, Qingdao Key Laboratory of Research in Marine Traditional Chinese Medicine, Qingdao Key Technology Innovation Center of Marine Traditional Chinese Medicine's Deep Development and Industrialization, Qingdao 266114, China
| | - Jintao Wang
- Chongqing Kangzhou Big Data (Group) Co., Ltd., Chongqing 401336, China
| | - Qin Zhang
- Chongqing Kangzhou Big Data (Group) Co., Ltd., Chongqing 401336, China
| | - Xianjun Fu
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Qingdao Academy of Chinese Medical Sciences Shandong University of Traditional Chinese Medicine, Qingdao Key Laboratory of Research in Marine Traditional Chinese Medicine, Qingdao Key Technology Innovation Center of Marine Traditional Chinese Medicine's Deep Development and Industrialization, Qingdao 266114, China.
| | - Pei-Cheng Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China.
| |
Collapse
|
6
|
Su Z, Yao B, Liu G, Fang J. Polyphenols as potential preventers of osteoporosis: A comprehensive review on antioxidant and anti-inflammatory effects, molecular mechanisms, and signal pathways in bone metabolism. J Nutr Biochem 2024; 123:109488. [PMID: 37865383 DOI: 10.1016/j.jnutbio.2023.109488] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 10/03/2023] [Accepted: 10/17/2023] [Indexed: 10/23/2023]
Abstract
Osteoporosis (OP) is a skeletal disorder characterized by decreased bone density, alterations in bone microstructure, and increased damage to the bones. As the population ages and life expectancy increases, OP has become a global epidemic, drawing attention from scientists and doctors. Because of polyphenols have favorable antioxidant and anti-allergy effects, which are regarded as potential methods to prevent angiocardipathy and OP. Polyphenols offer a promising approach to preventing and treating OP by affecting bone metabolism, reducing bone resolution, maintaining bone density, and lowering the differentiation level of osteoclasts (OC). There are multiple ways in which polyphenols affect bone metabolism. This article provides an overview of how polyphenols inhibit oxidative stress, exert antibacterial effects, and prevent the occurrence of OP. Furthermore, we will explore the regulatory mechanisms and signaling pathways implicated in this process.
Collapse
Affiliation(s)
- Zhan Su
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan, China
| | - Bin Yao
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan, China
| | - Gang Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan, China
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, Hunan, China.
| |
Collapse
|
7
|
Farhana F, Sakai E, Koyanagi Y, Yamaguchi Y, Alam MI, Okamoto K, Tsukuba T. Abr, a Rho-regulating protein, modulates osteoclastogenesis by enhancing lamellipodia formation by interacting with poly(ADP-ribose) glycohydrolase. Mol Biol Rep 2023; 50:7557-7569. [PMID: 37507586 DOI: 10.1007/s11033-023-08690-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Osteoclasts are multinucleated bone-resorbing cells formed by the fusion of monocyte/macrophage lineage. During osteoclast differentiation, Rho GTPases are involved in various processes, including cell migration, adhesion, and polarity. However, the role of Rho-regulatory molecules in the regulation of osteoclast differentiation remains unclear. In this study, among these genes, we focused on active breakpoint cluster region-related (Abr) protein that is a multifunctional regulator of Rho GTPases. METHODS AND RESULTS We examined using knockdown and overexpression experiments in RANKL-stimulated RAW-D macrophages whether Abr regulates osteoclast differentiation and cell morphology. We observed an increase in Abr expression during osteoclast differentiation and identified expression of a variant of the Abr gene in osteoclasts. Knockdown of Abr suppressed osteoclast differentiation and resorption. Abr knockdown markedly inhibited the expression of osteoclast markers, such as Nfatc1, c-fos, Src, and Ctsk in osteoclasts. Conversely, overexpression of Abr enhanced the formation of multinucleated osteoclasts, bone resorption activity, and osteoclast marker gene expression. Moreover, Abr overexpression accelerated lamellipodia formation and induced the formation of well-developed actin in osteoclasts. Importantly, the Abr protein interacted with poly(ADP-ribose) glycohydrolase (PARG) and Rho GTPases, including RhoA, Rac1/2/3, and Cdc42 in osteoclasts. CONCLUSIONS Taken together, these results indicate that Abr modulates osteoclastogenesis by enhancing lamellipodia formation via its interaction with PARG.
Collapse
Affiliation(s)
- Fatima Farhana
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Eiko Sakai
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Yu Koyanagi
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Yu Yamaguchi
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Mohammad Ibtehaz Alam
- Department of Periodontology and Endodontology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kuniaki Okamoto
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, 700-8558, Japan
| | - Takayuki Tsukuba
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan.
| |
Collapse
|
8
|
Rauf A, Abu-Izneid T, Imran M, Hemeg HA, Bashir K, Aljohani ASM, Aljohani MSM, Alhumaydhi FA, Khan IN, Bin Emran T, Gondal TA, Nath N, Ahmad I, Thiruvengadam M. Therapeutic Potential and Molecular Mechanisms of the Multitargeted Flavonoid Fisetin. Curr Top Med Chem 2023; 23:2075-2096. [PMID: 37431899 DOI: 10.2174/1568026623666230710162217] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/22/2023] [Accepted: 06/07/2023] [Indexed: 07/12/2023]
Abstract
Flavonoids effectively treat cancer, inflammatory disorders (cardiovascular and nervous systems), and oxidative stress. Fisetin, derived from fruits and vegetables, suppresses cancer growth by altering cell cycle parameters that lead to cell death and angiogenesis without affecting healthy cells. Clinical trials are needed in humans to prove the effectiveness of this treatment for a wide range of cancers. According to the results of this study, fisetin can be used to prevent and treat a variety of cancers. Despite early detection and treatment advances, cancer is the leading cause of death worldwide. We must take proactive steps to reduce the risk of cancer. The natural flavonoid fisetin has pharmacological properties that suppress cancer growth. This review focuses on the potential drug use of fisetin, which has been extensively explored for its cancer-fighting ability and other pharmacological activities such as diabetes, COVID-19, obesity, allergy, neurological, and bone disorders. Researchers have focused on the molecular function of fisetin. In this review, we have highlighted the biological activities against chronic disorders, including cancer, metabolic illnesses, and degenerative illnesses, of the dietary components of fisetin.
Collapse
Affiliation(s)
- Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar, KPK, Pakistan
| | - Tareq Abu-Izneid
- Department of Pharmaceutical Sciences, College of Pharmacy, Al Ain University of Science and Technology, Al Ain Campus, Abu Dhabi, United Arab Emirates
| | - Muhammad Imran
- Department of Food Science and Technology, University of Narowal, Punjab, Pakistan
| | - Hassan A Hemeg
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Taibah University, P.O. Box 344, Al-Madinah Al-Monawra, 41411, Saudi Arabia
| | - Kashif Bashir
- Department of Health and Biological Sciences, Abasyn University, Peshawar, Pakistan
| | - Abdullah S M Aljohani
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Mona S M Aljohani
- Pharmaceutical Care Department, King Saud Hospital, Ministry of Health, Unaizah, Saudi Arabia
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Ishaq N Khan
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, 25100, Pakistan
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Tanweer Aslam Gondal
- School of Exercise and Nutrition, Faculty of Health, Deakin University, Victoria, 3125, Australia
| | - Nikhil Nath
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, 4318, Bangladesh
| | - Ishtiaque Ahmad
- Department of Dairy Technology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, 05029, South Korea
| |
Collapse
|
9
|
Ugan RA, Cadirci E, Un H, Cinar I, Gurbuz MA. Fisetin Attenuates Paracetamol-Induced Hepatotoxicity by Regulating CYP2E1 Enzyme. AN ACAD BRAS CIENC 2023; 95:e20201408. [PMID: 37018834 DOI: 10.1590/0001-3765202320201408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/14/2020] [Indexed: 04/07/2023] Open
Abstract
Paracetamol is one of the drugs that cause hepatic damage. Fisetin has wide pharmacological effects such as anticancer, antiinflammatory and antioxidant. We aimed to evaluate the possible protective effect of fisetin on paracetamol-induced hepatotoxicity. Fisetin was administered at 25 and 50 mg/kg doses. Paracetamol was administered orally at a dose of 2 g/kg for induce hepatotoxicity 1 h after the fisetin and NAC treatments. The rats were sacrificed 24h after the Paracetamol administration. Tumor necrosis factor-alpha (TNF-α), NFκB and CYP2E1 mRNA levels and Superoxide dismutase (SOD) activity, glutathione (GSH) and malondialdehyde (MDA) levels of livers were determined. Serum ALT, AST and ALP levels were measured. Histopathological examinations were also performed. Fisetin administration significantly decreased the ALT, AST and ALP levels in a dose dependent manner. In addition, SOD activity and GSH levels increased, and the MDA level decreased with the treatment of fisetin. The TNF-α, NFκB and CYP2E1 gene expressions were significantly lower in both doses of the fisetin groups compared with the PARA group. Histopathological examinations showed that fisetin has hepatoprotective effects. This study showed that fisetin has the liver protective effects by increasing GSH, decreasing inflammatory mediators and CYP2E1.
Collapse
Affiliation(s)
- Rustem A Ugan
- Ataturk University, Faculty of Pharmacy, Department of Pharmacology, 25240, Erzurum, Turkey
- Ataturk University, Clinical Research, Development and Design Application and Research Center, 25240, Erzurum, Turkey
| | - Elif Cadirci
- Ataturk University, Faculty of Medicine, Department of Pharmacology, 25240, Erzurum, Turkey
- Ataturk University, Clinical Research, Development and Design Application and Research Center, 25240, Erzurum, Turkey
| | - Harun Un
- Agri Ibrahim Cecen University, Faculty of Pharmacy, Department of Biochemistry, 04100, Agri, Turkey
| | - Irfan Cinar
- Kafkas University, Faculty of Medicine, Department of Pharmacology, Kars, 36200, Turkey
| | - Muhammet A Gurbuz
- Ataturk University, Faculty of Medicine, Department of Histology and Embryology, 25240, Erzurum, Turkey
| |
Collapse
|
10
|
Yamaura K, Nelson AL, Nishimura H, Rutledge JC, Ravuri SK, Bahney C, Philippon MJ, Huard J. The effects of fisetin on bone and cartilage: A systematic review. Pharmacol Res 2022; 185:106504. [DOI: 10.1016/j.phrs.2022.106504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/06/2022] [Accepted: 10/09/2022] [Indexed: 12/09/2022]
|
11
|
Hassan SSU, Samanta S, Dash R, Karpiński TM, Habibi E, Sadiq A, Ahmadi A, Bungau S. The neuroprotective effects of fisetin, a natural flavonoid in neurodegenerative diseases: Focus on the role of oxidative stress. Front Pharmacol 2022; 13:1015835. [PMID: 36299900 PMCID: PMC9589363 DOI: 10.3389/fphar.2022.1015835] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/08/2022] [Indexed: 12/13/2022] Open
Abstract
Oxidative stress (OS) disrupts the chemical integrity of macromolecules and increases the risk of neurodegenerative diseases. Fisetin is a flavonoid that exhibits potent antioxidant properties and protects the cells against OS. We have viewed the NCBI database, PubMed, Science Direct (Elsevier), Springer-Nature, ResearchGate, and Google Scholar databases to search and collect relevant articles during the preparation of this review. The search keywords are OS, neurodegenerative diseases, fisetin, etc. High level of ROS in the brain tissue decreases ATP levels, and mitochondrial membrane potential and induces lipid peroxidation, chronic inflammation, DNA damage, and apoptosis. The subsequent results are various neuronal diseases. Fisetin is a polyphenolic compound, commonly present in dietary ingredients. The antioxidant properties of this flavonoid diminish oxidative stress, ROS production, neurotoxicity, neuro-inflammation, and neurological disorders. Moreover, it maintains the redox profiles, and mitochondrial functions and inhibits NO production. At the molecular level, fisetin regulates the activity of PI3K/Akt, Nrf2, NF-κB, protein kinase C, and MAPK pathways to prevent OS, inflammatory response, and cytotoxicity. The antioxidant properties of fisetin protect the neural cells from inflammation and apoptotic degeneration. Thus, it can be used in the prevention of neurodegenerative disorders.
Collapse
Affiliation(s)
- Syed Shams ul Hassan
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
- Department of Natural Product Chemistry, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Saptadip Samanta
- Department of Physiology, Midnapore College, Midnapore, West Bengal, India
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju, South Korea
| | - Tomasz M. Karpiński
- Department of Medical Microbiology, Poznań University of Medical Sciences, Poznań, Poland
| | - Emran Habibi
- Department of Pharmacognosy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abdul Sadiq
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Centre, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
12
|
Kim SH, Huh CK. Isolation and Identification of Fisetin: An Antioxidative Compound Obtained from Rhus verniciflua Seeds. Molecules 2022; 27:molecules27144510. [PMID: 35889379 PMCID: PMC9318972 DOI: 10.3390/molecules27144510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 12/10/2022] Open
Abstract
The goal of this study was to provide basic data for the development of functional food and health materials for Rhus verniciflua (R. verniciflua) seeds. We investigated an antioxidative compound obtained from these seeds. Solvent fractionation was carried out on a 50%-ethanol extract of the seeds. The DPPH and ABTS radical-scavenging activity and superoxide dismutase (SOD) activity were measured, and high antioxidant activity was seen in the ethyl acetate fraction. The antioxidant compounds in the ethyl acetate fraction were isolated using silica-gel column chromatography by adjusting the solvent between chloroform and methanol. Fraction numbers 2–7 showed activity of more than 50%. Next, primary column chromatography was used to mix and concentrate the fractions that demonstrated antioxidant activity. The fractions were then subjected to secondary column chromatography to obtain subfraction 4, which showed high antioxidant activity. The separation of subfraction 4 was then performed using high-performance liquid chromatography (HPLC). Three peaks were identified and peak number 2 was judged to be the primary antioxidative compound, which was then isolated by pure separation. Finally, the purified subfraction peak number 2 was identified as a fisetin compound by liquid chromatography–mass spectrometry (LC–MS), nuclear magnetic resonance (NMR), and HPLC.
Collapse
Affiliation(s)
- Su-Hwan Kim
- Research Institute of Food Industry, Sunchon National University, Suncheon 57922, Korea;
| | - Chang-Ki Huh
- Department of Food Science and Technology, Sunchon National University, Suncheon 57922, Korea
- Correspondence: ; Tel.: +82-61-750-3251
| |
Collapse
|
13
|
McCarty MF, Lewis Lujan L, Iloki Assanga S. Targeting Sirt1, AMPK, Nrf2, CK2, and Soluble Guanylate Cyclase with Nutraceuticals: A Practical Strategy for Preserving Bone Mass. Int J Mol Sci 2022; 23:4776. [PMID: 35563167 PMCID: PMC9104509 DOI: 10.3390/ijms23094776] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 12/15/2022] Open
Abstract
There is a vast pre-clinical literature suggesting that certain nutraceuticals have the potential to aid the preservation of bone mass in the context of estrogen withdrawal, glucocorticoid treatment, chronic inflammation, or aging. In an effort to bring some logical clarity to these findings, the signaling pathways regulating osteoblast, osteocyte, and osteoclast induction, activity, and survival are briefly reviewed in the present study. The focus is placed on the following factors: the mechanisms that induce and activate the RUNX2 transcription factor, a key driver of osteoblast differentiation and function; the promotion of autophagy and prevention of apoptosis in osteoblasts/osteoclasts; and the induction and activation of NFATc1, which promotes the expression of many proteins required for osteoclast-mediated osteolysis. This analysis suggests that the activation of sirtuin 1 (Sirt1), AMP-activated protein kinase (AMPK), the Nrf2 transcription factor, and soluble guanylate cyclase (sGC) can be expected to aid the maintenance of bone mass, whereas the inhibition of the serine kinase CK2 should also be protective in this regard. Fortuitously, nutraceuticals are available to address each of these targets. Sirt1 activation can be promoted with ferulic acid, N1-methylnicotinamide, melatonin, nicotinamide riboside, glucosamine, and thymoquinone. Berberine, such as the drug metformin, is a clinically useful activator of AMPK. Many agents, including lipoic acid, melatonin, thymoquinone, astaxanthin, and crucifera-derived sulforaphane, can promote Nrf2 activity. Pharmacological doses of biotin can directly stimulate sGC. Additionally, certain flavonols, notably quercetin, can inhibit CK2 in high nanomolar concentrations that may be clinically relevant. Many, though not all, of these agents have shown favorable effects on bone density and structure in rodent models of bone loss. Complex nutraceutical regimens providing a selection of these nutraceuticals in clinically meaningful doses may have an important potential for preserving bone health. Concurrent supplementation with taurine, N-acetylcysteine, vitamins D and K2, and minerals, including magnesium, zinc, and manganese, plus a diet naturally high in potassium, may also be helpful in this regard.
Collapse
Affiliation(s)
| | - Lidianys Lewis Lujan
- Department of Research and Postgraduate in Food Science, Sonoran University, Hermosillo 83200, Mexico;
| | - Simon Iloki Assanga
- Department of Biological Chemical Sciences, Sonoran University, Hermosillo 83200, Mexico;
| |
Collapse
|
14
|
Xu Q, Cao Z, Xu J, Dai M, Zhang B, Lai Q, Liu X. Effects and mechanisms of natural plant active compounds for the treatment of osteoclast-mediated bone destructive diseases. J Drug Target 2021; 30:394-412. [PMID: 34859718 DOI: 10.1080/1061186x.2021.2013488] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Bone-destructive diseases, caused by overdifferentiation of osteoclasts, reduce bone mass and quality, and disrupt bone microstructure, thereby causes osteoporosis, Paget's disease, osteolytic bone metastases, and rheumatoid arthritis. Osteoclasts, the only multinucleated cells with bone resorption function, are derived from haematopoietic progenitors of the monocyte/macrophage lineage. The regulation of osteoclast differentiation is considered an effective target for the treatment of bone-destructive diseases. Natural plant-derived products have received increasing attention in recent years due to their good safety profile, the preference of natural compounds over synthetic drugs, and their potential therapeutic and preventive activity against osteoclast-mediated bone-destructive diseases. In this study, we reviewed the research progress of the potential antiosteoclast active compounds extracted from medicinal plants and their molecular mechanisms. Active compounds from natural plants that inhibit osteoclast differentiation and functions include flavonoids, terpenoids, quinones, glucosides, polyphenols, alkaloids, coumarins, lignans, and limonoids. They inhibit bone destruction by downregulating the expression of osteoclast-specific marker genes (CTSK, MMP-9, TRAP, OSCAR, DC-STAMP, V-ATPase d2, and integrin av3) and transcription factors (c-Fos, NFATc1, and c-Src), prevent the effects of local factors (ROS, LPS, and NO), and suppress the activation of various signalling pathways (MAPK, NF-κB, Akt, and Ca2+). Therefore, osteoclast-targeting natural products are of great value in the prevention and treatment of bone destructive diseases.
Collapse
Affiliation(s)
- Qiang Xu
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhiyou Cao
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - JiaQiang Xu
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Min Dai
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Bin Zhang
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qi Lai
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xuqiang Liu
- Department of Orthopedics, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
15
|
Ma X, Tian Y, Xue K, Huai Y, Patil S, Deng X, Hao Q, Li D, Miao Z, Zhang W, Qian A. Kaempferide enhances antioxidant capacity to promote osteogenesis through FoxO1/β-catenin signaling pathway. Eur J Pharmacol 2021; 911:174555. [PMID: 34627807 DOI: 10.1016/j.ejphar.2021.174555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Forkhead box O1 (FoxO1)/β-catenin signaling pathway is a main oxidative defense pathway, which plays essential roles in the regulation of osteoporosis (OP). The natural products possess quality therapeutic effects and few side effects. It is used as a novel strategy in the treatment of OP. However, there is no systematic study in the natural antioxidant drug based on the FoxO1/β-catenin signaling pathway. This paper aims to discover pro-osteogenesis natural antioxidants for the prevention and treatment of OP. METHODS Systems pharmacology; combined with reverse drug targeting, systems-ADME process, network analysis and molecular docking, was used to screen natural antioxidants based on the FoxO1/β-catenin signaling pathway. Then in vitro experiments were performed to evaluate the osteogenesis effects of screened natural antioxidants. RESULTS Kaempferide was screened as the most potential antioxidant to improve osteogenesis by the regulation of the FoxO1/β-catenin signaling pathway. In vitro experiments showed that kaempferide significantly increased the expression of antioxidant genes and promoted osteogenic differentiation. Furthermore, kaempferide also improved the osteogenic differentiation inhibited by H2O2 through the enhancement of antioxidant capacity. Notably, kaempferide promoted cell antioxidant capacity by the increased nuclear translocation of FoxO1 and β-catenin. CONCLUSIONS These findings suggest that kaempferide is the natural antioxidant to promote osteogenesis effectively through the FoxO1/β-catenin signaling pathway. Natural antioxidant therapy maybe a promising strategy for the prevention and treatment of OP.
Collapse
Affiliation(s)
- Xiaoli Ma
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Ye Tian
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Kaiyue Xue
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Ying Huai
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Suryaji Patil
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Xiaoni Deng
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.
| | - Danming Li
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210006, China.
| | - Zhiping Miao
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Wenjuan Zhang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| | - Airong Qian
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| |
Collapse
|
16
|
Liang R, Zhao Q, Zhu Q, He X, Gao M, Wang Y. Lycium barbarum polysaccharide protects ARPE‑19 cells against H 2O 2‑induced oxidative stress via the Nrf2/HO‑1 pathway. Mol Med Rep 2021; 24:769. [PMID: 34490478 PMCID: PMC8436232 DOI: 10.3892/mmr.2021.12409] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 08/13/2021] [Indexed: 01/03/2023] Open
Abstract
Age-related macular degeneration (AMD) is a global health problem. Lycium barbarum polysaccharide (LBP), a traditional Chinese herbal medicine, has been proven to be effective against several eye diseases. However, only a few studies have investigated the effectiveness of LBP for AMD. In the present study, the human retinal epithelial cell line, ARPE-19, was pretreated with LBP for 24 h before exposure to H2O2 (500 µM). Cell viability was assessed, and a series of oxidative and antioxidant indicators were evaluated to determine the influence of LBP on H2O2-triggered oxidative stress. The present study also determined the apoptosis status, as well as the expression levels of apoptotic proteins and nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway proteins. The present study aimed to determine the protective role for LBP pretreatment and its underlying molecular mechanism. The results of the present study suggest that pretreatment of ARPE-19 cells with LBP exhibit high efficacy at reducing oxidative damage and inhibiting cell apoptosis. Furthermore, LBP may modulate the expression of proteins involved in the apoptotic pathway and activate the Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Ran Liang
- Department of Ophthalmology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116021, P.R. China
| | - Qi Zhao
- Department of Ophthalmology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116021, P.R. China
| | - Qing Zhu
- Department of Ophthalmology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116021, P.R. China
| | - Xin He
- Department of Ophthalmology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116021, P.R. China
| | - Mingjun Gao
- Department of Ophthalmology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116021, P.R. China
| | - Yiru Wang
- Department of Ophthalmology, The Second Hospital of Dalian Medical University, Dalian, Liaoning 116021, P.R. China
| |
Collapse
|
17
|
Fisetin Inhibits NLRP3 Inflammasome by Suppressing TLR4/MD2-Mediated Mitochondrial ROS Production. Antioxidants (Basel) 2021; 10:antiox10081215. [PMID: 34439462 PMCID: PMC8389007 DOI: 10.3390/antiox10081215] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 12/25/2022] Open
Abstract
Fisetin has numerous therapeutic properties, such as anti-inflammatory, antioxidative, and anticancer effects. However, the mechanism by which fisetin inhibits NLRP3 inflammasome remains unclear. In this study, we observed that fisetin bound to TLR4 and occluded the hydrophobic pocket of MD2, which in turn inhibited the binding of LPS to the TLR4/MD2 complex. This prevented the initiation of scaffold formation by the inhibition of MyD88/IRAK4 and subsequently downregulated the NF-κB signaling pathway. The result also demonstrated that fisetin downregulated the activation of the NLRP3 inflammasome induced by LPS and ATP (LPS/ATP) and the subsequent maturation of IL-1β. Fisetin also activated mitophagy and prevented the accumulation of damaged mitochondria and the excessive production of mitochondrial reactive oxygen species. The transient knockdown of p62 reversed the inhibitory activity of fisetin on the LPS/ATP-induced formation of the NLRP3 inflammasome. This indicated that fisetin induces p62-mediated mitophagy for eliminating damaged mitochondria. Recently, the existence of inflammasomes in non-mammalian species including zebrafish have been identified. Treatment of an LPS/ATP-stimulated zebrafish model with fisetin aided the recovery of the impaired heart rate, decreased the recruitment of macrophage to the brain, and gradually downregulated the expression of inflammasome-related genes. These results indicated that fisetin inhibited the TLR4/MD2-mediated activation of NLRP3 inflammasome by eliminating damaged mitochondria in a p62-dependent manner.
Collapse
|
18
|
Yang MY, Fan Z, Zhang Z, Fan J. MitoQ protects against high glucose-induced brain microvascular endothelial cells injury via the Nrf2/HO-1 pathway. J Pharmacol Sci 2020; 145:105-114. [PMID: 33357768 DOI: 10.1016/j.jphs.2020.10.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/07/2020] [Accepted: 10/23/2020] [Indexed: 12/13/2022] Open
Abstract
Brain microvascular endothelial cells (BMECs) dysfunction is related to the pathogenesis of neurovascular complication of diabetes mellitus that adversely lead to various CNS disorders. Mitoquinone (MitoQ) is a mitochondria targeted antioxidant that exerts multiple protective effects in many oxidative damage-related diseases. In this study, we determined the protective effects of MitoQ on high glucose (HG)-induced BMECs injury and investigated the underlying mechanism. We found that HG significantly reduced the expression of Nrf2 and HO-1, decreased mitochondrial membrane potential, increased intracellular and mitochondrial reactive oxygen species (ROS) generation, induced cytoskeletal damage and apoptosis in BMECs. In addition, Mito tempol, a mitochondrial ROS scavenger, significantly reduced HG-induced mitochondrial ROS production and attenuated cytoskeletal damage and cell apoptosis, suggesting MtROS production was involved in HG-induced BMECs injury. Moreover, we found that MitoQ treatment significantly upregulated the expression of Nrf2 and HO-1 in HG-induced BMECs, which is accompanied by improved mitochondrial membrane potential and decreased MtROS production. Meanwhile, MitoQ treatment also remarkably attenuated HG-induced cytoskeletal damage and cell apoptosis in BMECs. However, inhibitor of Nrf2 with ML385 impaired the protective effects of MitoQ in HG-induced BMECs. In conclusion, our results suggest that MitoQ exerts protective effect on HG-induced BMECs injury via activating Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Min-Yan Yang
- Department of Internal Medicine, The Fourth People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Zhen Fan
- Department of Geriatrics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Zhao Zhang
- Department of Emergency, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Jin Fan
- Department of Neurology, The General Hospital of Western Theater Command, Chendu, Sichuan, China.
| |
Collapse
|
19
|
Zhou L, Song H, Zhang Y, Ren Z, Li M, Fu Q. Polyphyllin VII attenuated RANKL-induced osteoclast differentiation via inhibiting of TRAF6/c-Src/PI3K pathway and ROS production. BMC Musculoskelet Disord 2020; 21:112. [PMID: 32075617 PMCID: PMC7031869 DOI: 10.1186/s12891-020-3077-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 01/20/2020] [Indexed: 11/24/2022] Open
Abstract
Background Osteoporosis is a worldwide severe bone disease. This study aimed to evaluate the effect of polyphyllin VII on the genesis of osteoclasts from bone marrow macrophages (BMMs) and its potentiality as a therapeutic drug for osteoporosis. Methods BMMs were induced to differentiate into osteoclasts by RANKL and M-CSF. The cells were then treated with various concentrations of polyphyllin VII. Intracellular reactive oxygen species (ROS) measurement assay, resorption pit formation assay, tartrate-resistant acid phosphatase (TRAP) staining and TRAP activity assessment, cell viability assay, active GTPase pull-down assay, immunofluorescent staining, immunoblotting, and RT-PCR were performed. Results RANKL + M-CSF significantly increased TRAP activity, number of osteoclasts, number and area of lacunae, intracellular content of ROS, protein levels of Nox1, TRAF6, c-Src and p-PI3K, as well as the content of activated GTP-Rac1, which were significantly blocked by polyphyllin VII in a concentration-dependent manner. Conclusion These findings suggested that polyphyllin VII inhibited differentiation of BMMs into osteoclasts through suppressing ROS synthesis, which was modulated by TRAF6–cSrc–PI3k signal transduction pathway including GTP-Rac1 and Nox1. Polyphyllin VII could be a therapeutic drug for osteoporosis.
Collapse
Affiliation(s)
- Long Zhou
- Department of Orthopedics, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China
| | - Hanyi Song
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yiqi Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China
| | - Zhaozhou Ren
- Department of Orthopedics, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China
| | - Minghe Li
- Department of Clinical Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qin Fu
- Department of Orthopedics, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning Province, China.
| |
Collapse
|
20
|
Tong H, Zhang X, Tan L, Jin R, Huang S, Li X. Multitarget and promising role of dihydromyricetin in the treatment of metabolic diseases. Eur J Pharmacol 2019; 870:172888. [PMID: 31866404 DOI: 10.1016/j.ejphar.2019.172888] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/01/2019] [Accepted: 12/18/2019] [Indexed: 12/30/2022]
Abstract
Dihydromyricetin (DMY or DHM), also known as ampelopsin, is the main natural flavonol compound extracted from the plant Ampelopsis grossedentata (Hand. -Mazz) W.T. Wang. In recent years, accumulating studies have been conducted to explore the extensive biological functions of DMY, including antitumor, anti-inflammation, organ-protective, and metabolic regulation effects. DMY acts as a potential preventive or therapeutic agent in treating multiple diseases, such as diabetes mellitus, atherosclerosis, nonalcoholic fatty liver disease and osteoporosis. This review article summarizes the preventive and therapeutic potential of DMY in multiple metabolic diseases and the main signaling pathways in which DMY participates to offer a comprehensive understanding and guidance for future studies.
Collapse
Affiliation(s)
- Haihui Tong
- Department of Pediatrics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jie Fang Avenue, Wuhan, Hubei Province, 430022, China.
| | - Xuejun Zhang
- Department of Orthopedics, The People's Hospital of China Three Gorges University, First People's Hospital of Yichang, No. 4 Hudi Street, Yichang, Hubei Province, 443000, China.
| | - Lingfang Tan
- Department of Pediatrics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jie Fang Avenue, Wuhan, Hubei Province, 430022, China.
| | - Runming Jin
- Department of Pediatrics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jie Fang Avenue, Wuhan, Hubei Province, 430022, China.
| | - Shilong Huang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jie Fang Avenue, Wuhan, Hubei Province, 430030, China.
| | - Xin Li
- Department of Pediatrics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jie Fang Avenue, Wuhan, Hubei Province, 430022, China.
| |
Collapse
|
21
|
Zhang H, Zheng W, Feng X, Yang F, Qin H, Wu S, Hou DX, Chen J. Nrf2⁻ARE Signaling Acts as Master Pathway for the Cellular Antioxidant Activity of Fisetin. Molecules 2019; 24:molecules24040708. [PMID: 30781396 PMCID: PMC6413105 DOI: 10.3390/molecules24040708] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/12/2019] [Accepted: 02/12/2019] [Indexed: 12/22/2022] Open
Abstract
Fisetin, a dietary flavonoid, is reported to have cellular antioxidant activity with an unclear mechanism. In this study, we investigated the effect of fisetin on the nuclear factor, erythroid 2-like 2 (Nrf2) signaling pathway in HepG2 cells to explore the cellular antioxidant mechanism. Fisetin upregulated the mRNA expression of heme oxygenase-1 (HO-1), glutamate-cysteine ligase catalytic subunit (GCLC), glutamate-cysteine ligase modifier subunit (GCLM), and NAD(P)H quinone oxidoreductase-1 (NQO1), and induced the protein of HO-1 but had no significant effect on the protein of GCLC, GCLM and NQO1. Moreover, nuclear accumulation of Nrf2 was clearly observed by immunofluorescence analysis and western blotting after fisetin treatment, and an enhanced luciferase activity of antioxidant response element (ARE)-regulated transactivation was obtained by dual-luciferase reporter gene assays. In addition, fisetin upregulated the protein level of Nrf2 and downregulated the protein level of Kelch-like ECH-associated protein 1 (Keap1). However, fisetin had no significant effect on Nrf2 mRNA expression. When protein synthesis was inhibited with cycloheximide (CHX), fisetin prolonged the half-life of Nrf2 from 15 min to 45 min. When blocking Nrf2 degradation with proteasome inhibitor MG132, ubiquitinated proteins were enhanced, and fisetin reduced ubiquitination of Nrf2. Taken together, fisetin translocated Nrf2 into the nucleus and upregulated the expression of downstream HO-1 gene by inhibiting the degradation of Nrf2 at the post-transcriptional level. These data provide the molecular mechanism to understand the cellular antioxidant activity of fisetin.
Collapse
Affiliation(s)
- Huihui Zhang
- Xiangya School of Public Health, Central South University, Changsha 410128, China.
| | - Wan Zheng
- Xiangya School of Public Health, Central South University, Changsha 410128, China.
| | - Xiangling Feng
- Xiangya School of Public Health, Central South University, Changsha 410128, China.
| | - Fei Yang
- Xiangya School of Public Health, Central South University, Changsha 410128, China.
| | - Hong Qin
- Xiangya School of Public Health, Central South University, Changsha 410128, China.
| | - Shusong Wu
- 1515 Core Research Program, Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| | - De-Xing Hou
- 1515 Core Research Program, Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
- Course of Biological Science and Technology, The United Graduate School of Agricultural Sciences, Department of Food Science and Biotechnology, Faculty of Agriculture, Kagoshima University, Korimoto 1-21-24, Kagoshima 890-0065, Japan.
| | - Jihua Chen
- Xiangya School of Public Health, Central South University, Changsha 410128, China.
- 1515 Core Research Program, Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| |
Collapse
|
22
|
Zhang X, Li X, Fang J, Hou X, Fang H, Guo F, Li F, Chen A, Huang S. (2R,3R)Dihydromyricetin inhibits osteoclastogenesis and bone loss through scavenging LPS-induced oxidative stress and NF-κB and MAPKs pathways activating. J Cell Biochem 2018; 119:8981-8995. [PMID: 30076654 DOI: 10.1002/jcb.27154] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 05/18/2018] [Indexed: 12/26/2022]
Abstract
Osteolysis is a serious complication of several chronic inflammatory diseases and is closely associated with a local chronic inflammatory reaction with a variety of causes. However, similarities exist in the mechanisms of their pathological processes. Inflammatory factors and oxidative stress-induced nuclear factor κB (NF-κB) and mitogen-activated protein kinases (MAPKs) signaling pathways play a center role in bone erosion. Dihydromyricetin (DMY) is a natural compound with anti-inflammatory and antioxidative effect, which are commonly used in chronic pharyngitis and alcohol use disorders. In the current study, we identified that DMY attenuated lipopolysaccharide (LPS)-induced oxidative stress through inhibiting the production of reactive oxygen species (ROS) and nitric oxide (NO), downregulated COX-2 and iNOS, and promoted the activity of the antioxidative system by activating superoxide dismutase (SOD) and Nrf2/HO-1 pathway. To further investigate the underlying mechanism, we found that DMY inhibits osteoclast (OC) differentiation and bone resorption activity through blocking the RANKL-induced activation of the NF-κB and MAPKs signaling pathways and then downregulated c-Fos and NFATc1, which is essential for OC differentiation. Furthermore, DMY inhibited LPS-induced osteolysis in vivo. Collectively, these results indicate that DMY might be a promising prophylactic antiosteoclastic/resorptive agent in preventing or treating bone lysis diseases.
Collapse
Affiliation(s)
- Xuejun Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Li
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianguo Fang
- Department of Pharmacy, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaolong Hou
- Department of Pharmacy, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huang Fang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anmin Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shilong Huang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Pandey MK, Gupta SC, Karelia D, Gilhooley PJ, Shakibaei M, Aggarwal BB. Dietary nutraceuticals as backbone for bone health. Biotechnol Adv 2018; 36:1633-1648. [PMID: 29597029 DOI: 10.1016/j.biotechadv.2018.03.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/13/2018] [Accepted: 03/21/2018] [Indexed: 12/11/2022]
|
24
|
Komaki S, Sakai E, Fukuma Y, Nishishita K, Okamoto K, Tsukuba T. Dihydroartemisinin represses osteoclastogenesis of bone marrow macrophages through reduced NFATc1 expression and impaired phosphorylation of IκBα. Biomed Res 2018; 39:169-177. [PMID: 30101837 DOI: 10.2220/biomedres.39.169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Osteoclasts are multinucleated bone resorbing cells whose differentiation is regulated by several important signaling pathways. Several lines of evidence indicate that dihydroartemisinin (DHA), an anti-malarial drug, inhibits osteoclast differentiation with little cytotoxicity. However, the detailed inhibitory mechanisms of DHA on osteoclastogenesis from native cells remain to be elucidated. In this study, we investigated the effects of DHA on the differentiation of bone marrow-derived macrophages into osteoclasts. DHA inhibited receptor activator of nuclear factor κ-B ligand (RANKL)-induced osteoclast formation and its bone resorbing activity. Mechanistically, DHA treatment markedly abolished phosphorylation of IκBα, and slightly affected a p38 MAPK dependent pathway. Moreover, DHA treatment induced down-regulation of nuclear factor of activated T cells cytoplasmic-1 (NFATc1), a master regulator for osteoclast differentiation and its target proteins, such as Src and cathepsin K. These results indicate that DHA represses RANKL-induced osteoclastogenesis of bone marrow macrophages through reduced NFATc1 expression and impaired phosphorylation of IκBα.
Collapse
Affiliation(s)
- Shunsuke Komaki
- Department of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences
| | - Eiko Sakai
- Department of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences
| | - Yutaka Fukuma
- Department of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences
| | - Kazuhisa Nishishita
- Department of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences
| | - Kuniaki Okamoto
- Department of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences
| | - Takayuki Tsukuba
- Department of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences
| |
Collapse
|
25
|
Huang W, Li ML, Xia MY, Shao JY. Fisetin-treatment alleviates airway inflammation through inhbition of MyD88/NF-κB signaling pathway. Int J Mol Med 2018; 42:208-218. [PMID: 29568921 PMCID: PMC5979929 DOI: 10.3892/ijmm.2018.3582] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/31/2018] [Indexed: 11/23/2022] Open
Abstract
Asthma is a common chronic airway inflammation disease and is considered as a major public health problem. Fisetin (3,3′,4′,7-tetrahydroxyflavone) is a naturally occurring flavonoid abundantly found in different vegetables and fruits. Fisetin has been reported to exhibit various positive biological effects, including anti-proliferative, anticancer, anti-oxidative and neuroprotective effects. We evaluated the effects of fisetin on allergic asthma regulation in mice. Mice were first sensi-tized, then airway-challenged with ovalbumin (OVA). Whether fisetin treatment attenuated OVA-induced airway inflammation was examined via inflammation inhibition through MyD88-related NF-κB (p65) signaling pathway. Mice were divided into the control (Con), OVA-induced asthma (Mod), 40 (FL) and 50 (FH) mg/kg fisetin-treated OVA-induced asthma groups. Our results found that OVA-induced airway inflammation in mice caused a significant inflammatory response via the activation of MyD88 and NF-κB signaling pathways, leading to release of pro-inflammatory cytokines. In contrast, fisetin-treated mice after OVA induction inhibited activation of MyD88 and NF-κB signaling pathways, resulting in downregulation of pro-inflammatory cytokine secretion. Further, fisetin significantly ameliorated the airway hyperresponsiveness (AHR) towards methacholine (Mch). In addition, fisetin reduced the number of eosinophil, monocyte, neutrophil and total white blood cell in the bronchoalveolar lavage fluid (BALF) of OVA-induced mice. The serum and BALF samples obtained from the OVA-induced mice with fisetin showed lower levels of pro-inflammatory cytokines. The results of our study illustrated that fisetin may be a new promising candidate to inhibit airway inflammation response induced by OVA.
Collapse
Affiliation(s)
- Wei Huang
- Department of Pediatrics, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei 066000, P.R. China
| | - Ming-Li Li
- Department of Pediatrics, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei 066000, P.R. China
| | - Ming-Yue Xia
- Department of Pediatrics, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei 066000, P.R. China
| | - Jian-Ying Shao
- Department of Pediatrics, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei 066000, P.R. China
| |
Collapse
|
26
|
Ghosh M, Kim IS, Lee YM, Hong SM, Lee TH, Lim JH, Debnath T, Lim BO. The Effects of Aronia melanocarpa 'Viking' Extracts in Attenuating RANKL-Induced Osteoclastic Differentiation by Inhibiting ROS Generation and c-FOS/NFATc1 Signaling. Molecules 2018. [PMID: 29518052 PMCID: PMC6017336 DOI: 10.3390/molecules23030615] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
This study aimed to determine the anti-osteoclastogenic effects of extracts from Aronia melanocarpa ‘Viking’ (AM) and identify the underlying mechanisms in vitro. Reactive oxygen species (ROS) are signal mediators in osteoclast differentiation. AM extracts inhibited ROS production in RAW 264.7 cells in a dose-dependent manner and exhibited strong radical scavenging activity. The extracts also attenuated the number of tartrate-resistant acid phosphatase (TRAP)-positive multinucleated osteoclasts. To attain molecular insights, the effect of the extracts on the signaling pathways induced by receptor activator of nuclear factor kappa B ligand (RANKL) were also investigated. RANKL triggers many transcription factors through the activation of mitogen-activated protein kinase (MAPK) and ROS, leading to the induction of osteoclast-specific genes. The extracts significantly suppressed RANKL-induced activation of MAPKs, such as extracellular signal-regulated kinase (ERK), c-Jun-N-terminal kinase (JNK) and p38 and consequently led to the downregulation of c-Fos and nuclear factor of activated T cells 1 (NFATc1) protein expression which ultimately suppress the activation of the osteoclast-specific genes, cathepsin K, TRAP, calcitonin receptor and integrin β3. In conclusion, our findings suggest that AM extracts inhibited RANKL-induced osteoclast differentiation by downregulating ROS generation and inactivating JNK/ERK/p38, nuclear factor kappa B (NF-κB)-mediated c-Fos and NFATc1 signaling pathway.
Collapse
Affiliation(s)
- Mithun Ghosh
- Department of Applied Life Science, College of Biomedical & Health Science, Konkuk University, Chungju-si, Chungcheongbuk-do 27478, Korea.
| | - In Sook Kim
- Department of Applied Life Science, College of Biomedical & Health Science, Konkuk University, Chungju-si, Chungcheongbuk-do 27478, Korea.
| | - Young Min Lee
- Department of Applied Life Science, College of Biomedical & Health Science, Konkuk University, Chungju-si, Chungcheongbuk-do 27478, Korea.
| | - Seong Min Hong
- Department of Applied Life Science, College of Biomedical & Health Science, Konkuk University, Chungju-si, Chungcheongbuk-do 27478, Korea.
| | - Taek Hwan Lee
- Department of Applied Life Science, College of Biomedical & Health Science, Konkuk University, Chungju-si, Chungcheongbuk-do 27478, Korea.
- Ahn-Gook Health, LTD, Seoul 07445, Korea.
| | - Ji Hong Lim
- Department of Applied Life Science, College of Biomedical & Health Science, Konkuk University, Chungju-si, Chungcheongbuk-do 27478, Korea.
| | - Trishna Debnath
- Department of Food Science and Biotechnology, Dongguk University-Seoul, Goyang-si, Gyeonggi-do 10326, Korea.
| | - Beong Ou Lim
- Department of Applied Life Science, College of Biomedical & Health Science, Konkuk University, Chungju-si, Chungcheongbuk-do 27478, Korea.
| |
Collapse
|
27
|
Kashyap D, Sharma A, Sak K, Tuli HS, Buttar HS, Bishayee A. Fisetin: A bioactive phytochemical with potential for cancer prevention and pharmacotherapy. Life Sci 2018; 194:75-87. [PMID: 29225112 DOI: 10.1016/j.lfs.2017.12.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/28/2017] [Accepted: 12/05/2017] [Indexed: 02/07/2023]
Abstract
A wide variety of chronic diseases, such as neurodegenerative and cardiovascular disorders, diabetes mellitus, osteoarthtitis, obesity and various cancers, are now being treated with cost effective phytomedicines. Since synthetic medicines are very expensive, concerted efforts are being made in developing and poor countries to discover cost effective medicines for the treatment of non-communicable diseases (NCDs). Understanding the underlying mechanisms of bioactive medicines from natural sources would not only open incipient avenues for the scientific community and pharmaceutical industry to discover new drug molecules for the therapy of NCDs, but also help to garner knowledge for alternative therapeutic approaches for the management of chronic diseases. Fisetin is a polyphenolic molecule of flavonoids class, and belongs to the bioactive phytochemicals that have potential to block multiple signaling pathways associated with NCDs such as cell division, angiogenesis, metastasis, oxidative stress, and inflammation. The emerging evidence suggests that fisetin may be useful for the prevention and management of several types of human malignancies. Efforts are being made to enhance the bioavailability of fisetin after oral administration to prevent and/or treat cancer of the liver, breast, ovary and other organs. The intent of this review is to highlight the in vitro and in vivo activities of fisetin and to provide up-to-date information about the molecular interactions of fisetin with its cellular targets involved in cancer initiation, promotion and progression as well as to focus on strategies underway to increase the bioavailability and reduce the risk of deleterious effects, if any, associated with fisetin administration.
Collapse
Affiliation(s)
- Dharambir Kashyap
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh 160 012, Punjab, India
| | - Ajay Sharma
- Department of Chemistry, Career Point University, Tikker-Kharwarian, Hamirpur 176 041, Himachal Pradesh, India
| | | | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar University, Mullana-Ambala 133 207, Haryana, India.
| | - Harpal Singh Buttar
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ontario, K1N 6N5, Canada
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL 33169, USA
| |
Collapse
|
28
|
Yamaguchi Y, Sakai E, Okamoto K, Kajiya H, Okabe K, Naito M, Kadowaki T, Tsukuba T. Rab44, a novel large Rab GTPase, negatively regulates osteoclast differentiation by modulating intracellular calcium levels followed by NFATc1 activation. Cell Mol Life Sci 2018; 75:33-48. [PMID: 28791425 PMCID: PMC11105776 DOI: 10.1007/s00018-017-2607-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 07/04/2017] [Accepted: 08/02/2017] [Indexed: 11/29/2022]
Abstract
Rab44 is an atypical Rab GTPase that contains some additional domains such as the EF-hand and coiled-coil domains as well as Rab-GTPase domain. Although Rab44 genes have been found in mammalian genomes, no studies concerning Rab44 have been reported yet. Here, we identified Rab44 as an upregulated protein during osteoclast differentiation. Knockdown of Rab44 by small interfering RNA promotes RANKL-induced osteoclast differentiation of the murine monocytic cell line, RAW-D or of bone marrow-derived macrophages (BMMs). In contrast, overexpression of Rab44 prevents osteoclast differentiation. Rab44 was localized in the Golgi complex and lysosomes, and Rab44 overexpression caused an enlargement of early endosomes. A series of deletion mutant studies of Rab44 showed that the coiled-coil domain and lipidation sites of Rab44 is important for regulation of osteoclast differentiation. Mechanistically, Rab44 affects nuclear factor of activated T-cells c1 (NFATc1) signaling in RANKL-stimulated macrophages. Moreover, Rab44 depletion caused an elevation in intracellular Ca2+ transients upon RANKL stimulation, and particularly regulated lysosomal Ca2+ influx. Taken together, these results suggest that Rab44 negatively regulates osteoclast differentiation by modulating intracellular Ca2+ levels followed by NFATc1 activation.
Collapse
Affiliation(s)
- Yu Yamaguchi
- Division of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Eiko Sakai
- Division of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Kuniaki Okamoto
- Division of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Hiroshi Kajiya
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, 814-0193, Japan
| | - Koji Okabe
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, 814-0193, Japan
| | - Mariko Naito
- Division of Microbiology and Oral Infection, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Tomoko Kadowaki
- Division of Frontier Life Science, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Takayuki Tsukuba
- Division of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan.
| |
Collapse
|
29
|
Ferreira de Oliveira JMP, Pacheco AR, Coutinho L, Oliveira H, Pinho S, Almeida L, Fernandes E, Santos C. Combination of etoposide and fisetin results in anti-cancer efficiency against osteosarcoma cell models. Arch Toxicol 2017; 92:1205-1214. [PMID: 29270805 DOI: 10.1007/s00204-017-2146-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/14/2017] [Indexed: 01/26/2023]
Abstract
Osteosarcoma chemotherapy is often limited by chemoresistance, resulting in poor prognosis. Combined chemotherapy could, therefore, be used to prevent resistance to chemotherapeutics. Here, the effects of fisetin on osteosarcoma cells were investigated, as well as cytostatic potential in combination with the anti-cancer drug etoposide. For this, different osteosarcoma cell lines were treated with fisetin, with etoposide and with respective combinations. Fisetin was associated with decrease in colony formation in Saos-2 and in U2OS cells but not in MG-63 cells. Notwithstanding, upon evaluation of cellular growth by crystal violet assay, MG-63 and Saos-2 cells showed decreased cell proliferation at 40 and 20 µM fisetin, respectively. Depending on the relative concentrations, fisetin:etoposide combinations showed negative-to-positive interactions on the inhibition of cell proliferation. In addition, fisetin treatment up to 50 µM for 48 h resulted in G2-phase cell cycle arrest. Regardless of the combination, fisetin:etoposide increased % cells in G2-phase and decreased % cells in G1-phase. In addition, mixtures with more positive combined effects induced increased % cells in S-phase. Compared to etoposide treatment, these combinations resulted in decreased levels of cyclins B1 and E1, pointing to the role of these regulators in fisetin-induced cell cycle arrest. In conclusion, these results show that the combination of fisetin with etoposide has higher anti-proliferative effects in osteosarcoma associated with cell cycle arrest, allowing the use of lower doses of the chemotherapeutic agent, which has important implications for osteosarcoma treatment.
Collapse
Affiliation(s)
- José Miguel P Ferreira de Oliveira
- UCIBIO, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.,Department of Biology, Faculty of Sciences, University of Porto, 4150-171, Porto, Portugal
| | - Ana Rita Pacheco
- Department of Biology and CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Laura Coutinho
- Department of Biology and CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Helena Oliveira
- Department of Biology and CESAM, University of Aveiro, 3810-193, Aveiro, Portugal.,CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 4200-319, Porto, Portugal
| | - Sónia Pinho
- Department of Biology and CESAM, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Luis Almeida
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Eduarda Fernandes
- UCIBIO, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Conceição Santos
- Department of Biology, Faculty of Sciences, University of Porto, 4150-171, Porto, Portugal. .,LAQV, REQUIMTE, Faculty of Sciences, University of Porto, Porto, Portugal.
| |
Collapse
|
30
|
Antioxidant properties of the flavonoid fisetin: An updated review of in vivo and in vitro studies. Trends Food Sci Technol 2017. [DOI: 10.1016/j.tifs.2017.10.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
31
|
Brito C, Stavroullakis A, Oliveira T, Prakki A. Cytotoxicity and potential anti-inflammatory activity of velutin on RAW 264.7 cell line differentiation: Implications in periodontal bone loss. Arch Oral Biol 2017; 83:348-356. [PMID: 28898790 DOI: 10.1016/j.archoralbio.2017.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/31/2017] [Accepted: 09/01/2017] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Hypoxia-inducible factor-1α (HIF-1α) has been implicated in periodontal tissue inflammation and possibly in osteoclast differentiation, while polyphenols are known to be anti-inflammatory natural compounds that are capable of regulating the NF-κB protein complex pathway. The objective of this study was to investigate cytotoxicity and HIF-1α expression through the NF-κB pathway by polyphenol velutin (Euterpe oleracea Mart.), found in the pulp of acai fruit, during inflammatory RAW 264.7 differentiation. DESIGN RAW 264.7 mouse monocyte macrophage cells were stimulated with RANKL (30ng/mL) and Porphyromonas gingivalis lipopolysaccharide (1μg/mL). Cells were treated with various concentrations of velutin (0.5-2μM) to check for viability, morphology, osteoclast differentiation, and HIF-1α expression (Western blot). RESULTS Alamar blue cell viability assay showed no toxicity to RAW cells with the use of velutin in all concentrations tested (p>0.05). Velutin did not induce cell apoptosis based on caspase 3/7 assay (p>0.05). Fluorescence images stained by DAPI showed no alteration in the morphology of RAW cell nuclei (p>0.05) treated with velutin. TRAP assays demonstrated a dose-dependent reduction in osteoclast formation by velutin when compared with control (p<0.05). Velutin showed a reduction in HIF-1α expression related to IκB phosphorylation when compared with control (p<0.001). CONCLUSIONS At the tested concentrations, velutin was not cytotoxic to RAW 264.7 and differentiated cells. Velutin reduced osteoclast differentiation and downregulated HIF-1α through the NF-κB pathway.
Collapse
Affiliation(s)
- Carlos Brito
- Department of Clinical Sciences, Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Alexander Stavroullakis
- Department of Clinical Sciences, Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Tatiane Oliveira
- Department of Clinical Sciences, Faculty of Dentistry, University of Toronto, Toronto, Canada; Institute of Health Sciences, Federal University of Bahia, Bahia, Brazil
| | - Anuradha Prakki
- Department of Clinical Sciences, Faculty of Dentistry, University of Toronto, Toronto, Canada.
| |
Collapse
|
32
|
Torre E. Molecular signaling mechanisms behind polyphenol-induced bone anabolism. PHYTOCHEMISTRY REVIEWS : PROCEEDINGS OF THE PHYTOCHEMICAL SOCIETY OF EUROPE 2017; 16:1183-1226. [PMID: 29200988 PMCID: PMC5696504 DOI: 10.1007/s11101-017-9529-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 08/20/2017] [Indexed: 05/08/2023]
Abstract
For millennia, in the different cultures all over the world, plants have been extensively used as a source of therapeutic agents with wide-ranging medicinal applications, thus becoming part of a rational clinical and pharmacological investigation over the years. As bioactive molecules, plant-derived polyphenols have been demonstrated to exert many effects on human health by acting on different biological systems, thus their therapeutic potential would represent a novel approach on which natural product-based drug discovery and development could be based in the future. Many reports have provided evidence for the benefits derived from the dietary supplementation of polyphenols in the prevention and treatment of osteoporosis. Polyphenols are able to protect the bone, thanks to their antioxidant properties, as well as their anti-inflammatory actions by involving diverse signaling pathways, thus leading to bone anabolic effects and decreased bone resorption. This review is meant to summarize the research works performed so far, by elucidating the molecular mechanisms of action of polyphenols in a bone regeneration context, aiming at a better understanding of a possible application in the development of medical devices for bone tissue regeneration.
Collapse
Affiliation(s)
- Elisa Torre
- Nobil Bio Ricerche srl, Via Valcastellana, 26, 14037 Portacomaro, AT Italy
| |
Collapse
|
33
|
Kanzaki H, Wada S, Narimiya T, Yamaguchi Y, Katsumata Y, Itohiya K, Fukaya S, Miyamoto Y, Nakamura Y. Pathways that Regulate ROS Scavenging Enzymes, and Their Role in Defense Against Tissue Destruction in Periodontitis. Front Physiol 2017; 8:351. [PMID: 28611683 PMCID: PMC5447763 DOI: 10.3389/fphys.2017.00351] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/15/2017] [Indexed: 01/04/2023] Open
Abstract
Periodontitis, an inflammatory disease that affects the tissues surrounding the teeth, is a common disease worldwide. It is caused by a dysregulation of the host inflammatory response to bacterial infection, which leads to soft and hard tissue destruction. In particular, it is the excessive inflammation in response to bacterial plaque that leads to the release of reactive oxygen species (ROS) from neutrophils, which, then play a critical role in the destruction of periodontal tissue. Generally, ROS produced from immune cells exhibit an anti-bacterial effect and play a role in host defense and immune regulation. Excessive ROS, however, can exert cytotoxic effects, cause oxidative damage to proteins, and DNA, can interfere with cell growth and cell cycle progression, and induce apoptosis of gingival fibroblasts. Collectively, these effects enable ROS to directly induce periodontal tissue damage. Some ROS also act as intracellular signaling molecules during osteoclastogenesis, and can thus also play an indirect role in bone destruction. Cells have several protective mechanisms to manage such oxidative stress, most of which involve production of cytoprotective enzymes that scavenge ROS. These enzymes are transcriptionally regulated via NRF2, Sirtuin, and FOXO. Some reports indicate an association between periodontitis and these cytoprotective enzymes' regulatory axes, with superoxide dismutase (SOD) the most extensively investigated. In this review article, we discuss the role of oxidative stress in the tissue destruction manifest in periodontitis, and the mechanisms that protect against this oxidative stress.
Collapse
Affiliation(s)
- Hiroyuki Kanzaki
- Maxillo-Oral Disorders, Tohoku University HospitalSendai, Japan.,Department of Orthodontics, School of Dental Medicine, Tsurumi UniversityYokohama, Japan
| | - Satoshi Wada
- Department of Orthodontics, School of Dental Medicine, Tsurumi UniversityYokohama, Japan
| | - Tsuyoshi Narimiya
- Department of Orthodontics, School of Dental Medicine, Tsurumi UniversityYokohama, Japan
| | - Yuuki Yamaguchi
- Department of Orthodontics, School of Dental Medicine, Tsurumi UniversityYokohama, Japan
| | - Yuta Katsumata
- Department of Orthodontics, School of Dental Medicine, Tsurumi UniversityYokohama, Japan
| | - Kanako Itohiya
- Department of Orthodontics, School of Dental Medicine, Tsurumi UniversityYokohama, Japan
| | - Sari Fukaya
- Department of Orthodontics, School of Dental Medicine, Tsurumi UniversityYokohama, Japan
| | - Yutaka Miyamoto
- Department of Orthodontics, School of Dental Medicine, Tsurumi UniversityYokohama, Japan
| | - Yoshiki Nakamura
- Department of Orthodontics, School of Dental Medicine, Tsurumi UniversityYokohama, Japan
| |
Collapse
|
34
|
Xu Z, Kong XQ. Bixin ameliorates high fat diet-induced cardiac injury in mice through inflammation and oxidative stress suppression. Biomed Pharmacother 2017; 89:991-1004. [PMID: 28292028 DOI: 10.1016/j.biopha.2017.02.052] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 02/05/2017] [Accepted: 02/15/2017] [Indexed: 12/27/2022] Open
Abstract
Diabetic cardiomyopathy is known as an essential complication of diabetes, a main reason leading to mortality for diabetic patients, and novel therapeutic strategies for treatment are urgently required. Bixin (BX), isolated from the seeds of Bixa orellana, is a carotenoid, possessing anti-inflammatory, anti-tumor and anti-oxidant activities. In our study, we attempted to calculate the role of bixin in cardiac injury progression, and reveal the possible molecular mechanism. Bixin treatment ameliorated cardiac dysfunction through inhibiting fibrosis, inflammation and reactive oxygen species (ROS) generation. It reduced fibrosis levels via collagen deposition down-regulation. Inflammatory response was attenuated by reducing pro-inflammatory cytokines secretion via Toll-like receptor 4/nuclear factor kappa B (TLR4/NF-κB) signaling pathway inactivation in mice induced by high fat diet. Also, in in vitro studies, lipopolysaccharide (LPS)-treated cardiac muscle cells exhibits pro-inflammatory cytokines over-expression, which was reduced by bixin through blocking TLR4/NF-κB pathway. Additionally, oxidative stress triggered by high fat in vivo and LPS in vitro was down-regulated for bixin administration via nuclear factor-E2-related factor 2 (Nrf2) signaling pathway activation. Our study suggested that bixin might be a novel and protective agent with therapeutic activity against cardiac injury by suppressing fibrosis, inflammation and oxidative stress.
Collapse
Affiliation(s)
- Zhou Xu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210009, China; Department of Cardiology, Huai'an First People's Hospital, Nanjing Medical University, 6 Beijing Road West, Huai'an 223300, China
| | - Xiang-Qing Kong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210009, China.
| |
Collapse
|
35
|
Lee SH, Lee JY, Kwon YI, Jang HD. Anti-Osteoclastic Activity of Artemisia capillaris Thunb. Extract Depends upon Attenuation of Osteoclast Differentiation and Bone Resorption-Associated Acidification Due to Chlorogenic Acid, Hyperoside, and Scoparone. Int J Mol Sci 2017; 18:ijms18020322. [PMID: 28165389 PMCID: PMC5343858 DOI: 10.3390/ijms18020322] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 01/23/2017] [Accepted: 01/26/2017] [Indexed: 12/31/2022] Open
Abstract
The present study attempts to elucidate the anti-osteoporotic activity of Artemisia capillaris Thunb. in the form of anti-osteoclastic effect and responsible bioactive compounds. The contents of chlorogenic acid, caffeic acid, hyperoside, isoquercitrin, isochlorogenic acid A, and scoparone in Artemisia capillaris hydroethanolic extract (ACHE) were 38.53, 0.52, 4.07, 3.03, 13.90, and 6.59 mg/g, respectively. ACHE diminished osteoclast differentiation and bone resorption due to chlorogenic acid, hyperoside, and scoparone. In addition, ACHE attenuated acidification as well as reducing tumor necrosis factor receptor-associated factor 6 (TRAF6) expression and its association with vacuolar H+-adenosine triphosphatase (V-ATPase). Furthermore, chlorogenic acid, hyperoside, and scoparone from A. capillaris abrogated the association of V-ATPase with TRAF6, suggesting that the blockage of bone resorption by A. capillaris was partially mediated by reducing acidification through down-regulating interaction of V-ATPase with TRAF6 due to scoparone as well as chlorogenic acid and hyperoside. These results imply that the anti-osteoclastic effect of A. capillaris through down-regulating osteoclast differentiation and bone resorption may contribute to its anti-osteoporotic effect.
Collapse
Affiliation(s)
- Sang-Hyun Lee
- Department of Food and Nutrition, Hannam University, Daejeon 34430, Korea.
| | - Jung-Yun Lee
- Department of Food and Nutrition, Hannam University, Daejeon 34430, Korea.
| | - Young-In Kwon
- Department of Food and Nutrition, Hannam University, Daejeon 34430, Korea.
| | - Hae-Dong Jang
- Department of Food and Nutrition, Hannam University, Daejeon 34430, Korea.
| |
Collapse
|
36
|
The flavonoid fisetin as an anticancer agent targeting the growth signaling pathways. Eur J Pharmacol 2016; 789:8-16. [DOI: 10.1016/j.ejphar.2016.07.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 06/30/2016] [Accepted: 07/01/2016] [Indexed: 12/29/2022]
|
37
|
Alliin Attenuated RANKL-Induced Osteoclastogenesis by Scavenging Reactive Oxygen Species through Inhibiting Nox1. Int J Mol Sci 2016; 17:ijms17091516. [PMID: 27657047 PMCID: PMC5037793 DOI: 10.3390/ijms17091516] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/21/2016] [Accepted: 09/06/2016] [Indexed: 01/22/2023] Open
Abstract
The healthy skeleton requires a perfect coordination of the formation and degradation of bone. Metabolic bone disease like osteoporosis is resulted from the imbalance of bone formation and/or bone resorption. Osteoporosis also reflects lower level of bone matrix, which is contributed by up-regulated osteoclast-mediated bone resorption. It is reported that monocytes/macrophage progenitor cells or either hematopoietic stem cells (HSCs) gave rise to multinucleated osteoclasts. Thus, inhibition of osteoclastic bone resorption generally seems to be a predominant therapy for treating osteoporosis. Recently, more and more natural compounds have been discovered, which have the ability of inhibiting osteoclast differentiation and fusion. Alliin (S-allyl-l-cysteine sulfoxides, SACSO) is the major component of aged garlic extract (AGE), bearing broad-spectrum natural antioxidant properties. However, its effects on bone health have not yet been explored. Hence, we designed the current study to explore its effects and role in receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast fusion and differentiation. It was revealed that alliin had an inhibitory effect in osteoclasteogenesis with a dose-dependent manner via blocking the c-Fos-NFATc1 signaling pathway. In addition, alliin decreased the generation of reactive oxygen species (ROS) and down-regulated the expression of NADPH oxidase 1 (Nox1). The overall results revealed that alliin could be a potential therapeutic agent in the treatment of osteoporosis.
Collapse
|
38
|
Sakai E, Aoki Y, Yoshimatsu M, Nishishita K, Iwatake M, Fukuma Y, Okamoto K, Tanaka T, Tsukuba T. Sanguiin H-6, a constituent of Rubus parvifolius L., inhibits receptor activator of nuclear factor-κB ligand-induced osteoclastogenesis and bone resorption in vitro and prevents tumor necrosis factor-α-induced osteoclast formation in vivo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:828-837. [PMID: 27288918 DOI: 10.1016/j.phymed.2016.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 03/11/2016] [Accepted: 04/06/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Osteoclasts are multinucleated bone-resorbing cells that differentiate in response to receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL). Enhanced osteoclastogenesis contributes to bone diseases, such as osteoporosis and rheumatoid arthritis. Rubus parvifolius L. is traditionally used as an herbal medicine for rheumatism; however, its detailed chemical composition and the molecular mechanisms responsible for its biological action have not been elucidated. PURPOSE To investigate the mechanisms by which R. parvifolius L. extract and its major constituent sanguiin H-6, inhibit osteoclastogenesis and bone resorption. METHODS Cell proliferation, cell differentiation, and bone resorption were detected in vitro. Inhibition of signaling pathways, marker protein expression, and protein nuclear translocation were evaluated by western blot analysis. Tumor necrosis factor-α (TNF-α)-mediated osteoclastogenesis was examined in vivo. RESULTS R. parvifolius L. extract inhibited the bone-resorption activity of osteoclasts. In addition, sanguiin H-6 markedly inhibited RANKL-induced osteoclast differentiation and bone resorption, reduced reactive oxygen species production, and inhibited the phosphorylation of inhibitor of NF-κB alpha (IκBα) and p38 mitogen-activated protein kinase. Sanguiin H-6 also decreased the protein levels of nuclear factor of activated T cells cytoplasmic-1 (NFATc1), cathepsin K, and c-Src. Moreover, sanguiin H-6 inhibited the nuclear translocation of NFATc1, c-Fos, and NF-κB in vitro, as well as TNF-α-mediated osteoclastogenesis in vivo. CONCLUSIONS Our data revealed that R. parvifolius L. has anti-bone resorption activity and suggest that its constituent, sanguiin H-6, can potentially be used for the prevention and treatment of bone diseases associated with excessive osteoclast formation and subsequent bone destruction.
Collapse
Affiliation(s)
- Eiko Sakai
- Division of Dental Pharmacology, Department of Developmental and Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan.
| | - Yuri Aoki
- Division of Dental Pharmacology, Department of Developmental and Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Masako Yoshimatsu
- Division of Orthodontics and Dentofacial Orthopedics, Department of Developmental and Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Kazuhisa Nishishita
- Division of Dental Pharmacology, Department of Developmental and Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Mayumi Iwatake
- Division of Dental Pharmacology, Department of Developmental and Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Yutaka Fukuma
- Division of Dental Pharmacology, Department of Developmental and Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Kuniaki Okamoto
- Division of Dental Pharmacology, Department of Developmental and Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Takashi Tanaka
- Division of Natural Product Chemistry, Department of Molecular Medicinal Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8131, Japan
| | - Takayuki Tsukuba
- Division of Dental Pharmacology, Department of Developmental and Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| |
Collapse
|
39
|
An J, Hao D, Zhang Q, Chen B, Zhang R, Wang Y, Yang H. Natural products for treatment of bone erosive diseases: The effects and mechanisms on inhibiting osteoclastogenesis and bone resorption. Int Immunopharmacol 2016; 36:118-131. [PMID: 27131574 DOI: 10.1016/j.intimp.2016.04.024] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 03/28/2016] [Accepted: 04/18/2016] [Indexed: 01/13/2023]
Abstract
Excessive bone resorption plays a central role on the development of bone erosive diseases, including osteoporosis, rheumatoid arthritis, and periodontitis. Osteoclasts, bone-resorbing multinucleated cells, are differentiated from hemopoietic progenitors of the monocyte/macrophage lineage. Regulation of osteoclast differentiation is considered an effective therapeutic target to the treatment of pathological bone loss. Natural plant-derived products, with potential therapeutic and preventive activities against bone-lytic diseases, have received increasing attention in recent years because of their whole regulative effects and specific pharmacological activities, which are more suitable for long-term use than chemically synthesized medicines. In this review, we summarized the detailed research progress on the active compounds derived from medical plants with potential anti-resorptive effects and their molecular mechanisms on inhibiting osteoclast formation and function. The active ingredients derived from natural plants that are efficacious in suppressing osteoclastogenesis and bone resorption include flavonoids, terpenoids (sesquiterpenoids, diterpenoids, triterpenoids), glycosides, lignans, coumarins, alkaloids, polyphenols, limonoids, quinones and others (steroid, oxoxishhone, fatty acid). Studies have shown that above natural products exert the inhibitory effects via regulating many factors involved in the process of osteoclast differentiation and bone resorption, including the essential cytokines (RANKL, M-CSF), transcription factors (NFATc1, c-Fos), signaling pathways (NF-κB, MAPKs, Src/PI3K/Akt, the calcium ion signaling), osteoclast-specific genes (TRAP, CTSK, MMP-9, integrin β3, OSCAR, DC-STAMP, Atp6v0d2) and local factors (ROS, LPS, NO). The development of osteoclast-targeting natural products is of great value for the prevention or treatment of bone diseases and for bone regenerative medicine.
Collapse
Affiliation(s)
- Jing An
- Translational Medicine Centre, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China
| | - Dingjun Hao
- Translational Medicine Centre, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China
| | - Qian Zhang
- Translational Medicine Centre, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China
| | - Bo Chen
- Translational Medicine Centre, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China
| | - Rui Zhang
- Translational Medicine Centre, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China
| | - Yi Wang
- Translational Medicine Centre, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China
| | - Hao Yang
- Translational Medicine Centre, Hong-Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an 710054, China.
| |
Collapse
|
40
|
Fisetin regulates TPA-induced breast cell invasion by suppressing matrix metalloproteinase-9 activation via the PKC/ROS/MAPK pathways. Eur J Pharmacol 2015; 764:79-86. [DOI: 10.1016/j.ejphar.2015.06.038] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 06/17/2015] [Accepted: 06/18/2015] [Indexed: 12/23/2022]
|
41
|
Uchino K, Okamoto K, Sakai E, Yoneshima E, Iwatake M, Fukuma Y, Nishishita K, Tsukuba T. Dual Effects of Liquiritigenin on the Proliferation of Bone Cells: Promotion of Osteoblast Differentiation and Inhibition of Osteoclast Differentiation. Phytother Res 2015; 29:1714-21. [PMID: 26172226 DOI: 10.1002/ptr.5416] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 05/29/2015] [Accepted: 06/20/2015] [Indexed: 01/25/2023]
Abstract
Bone is constantly controlled by a balance between osteoblastic bone formation and osteoclastic bone resorption. Liquiritigenin is a plant-derived flavonoid and has various pharmacological effects, such as antioxidative, antitumor, and antiinflammatory effects. Here, we show that liquiritigenin has dual effects on the proliferation of bone cells, regarding the promotion of osteoblast differentiation and the inhibition of osteoclast differentiation. Liquiritigenin-treated murine osteoblastic MC3T3-E1 cells showed an increased alkaline phosphatase activity and enhanced phosphorylation of Smad1/5 compared with untreated cells. Moreover, liquiritigenin inhibited osteoclast differentiation, its bone-resorption activity through slightly decreased the phosphorylation of extracellular signal-regulated kinase, c-Jun N-terminal kinase, and inhibitor of nuclear factor kappa Bα; however, the phosphorylation of Akt and p38 slightly increased in bone marrow-derived osteoclasts. The expression levels of the osteoclast marker proteins nuclear factor of activated T-cell cytoplasmic-1, Src, and cathepsin K diminished. These results suggest that liquiritigenin may be useful as a therapeutic and/or preventive agent for osteoporosis or inflammatory bone diseases.
Collapse
Affiliation(s)
- Kaho Uchino
- Department of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, 852-8588, Japan
| | - Kuniaki Okamoto
- Department of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, 852-8588, Japan
| | - Eiko Sakai
- Department of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, 852-8588, Japan
| | - Erika Yoneshima
- Department of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, 852-8588, Japan
| | - Mayumi Iwatake
- Department of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, 852-8588, Japan
| | - Yutaka Fukuma
- Department of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, 852-8588, Japan
| | - Kazuhisa Nishishita
- Department of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, 852-8588, Japan
| | - Takayuki Tsukuba
- Department of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, 852-8588, Japan
| |
Collapse
|
42
|
Moon SH, Choi SW, Kim SH. In vitro anti-osteoclastogenic activity of p38 inhibitor doramapimod via inhibiting migration of pre-osteoclasts and NFATc1 activity. J Pharmacol Sci 2015; 129:135-42. [PMID: 26232862 DOI: 10.1016/j.jphs.2015.06.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 06/18/2015] [Accepted: 06/24/2015] [Indexed: 12/13/2022] Open
Abstract
The mitogen activated protein kinase p38 plays a role in the receptor activator of NF-ĸB ligand (RANKL)-induced osteoclast differentiation. In this study, we investigated the effect of p38 inhibitor doramapimod on the osteoclast differentiation. Doramapimod significantly inhibited the osteoclastogenesis of bone marrow macrophages (BMMs) via attenuating the activation of p38 induced by M-CSF and RANKL. Importantly, doramapimod blocked the migration and fusion in pre-osteoclasts via the down-regulating NFATc1. The inhibitory effect of doramapimod on the migration/fusion of pre-osteoclasts via inhibiting NFATc1 activity were confirmed by measuring NFATc1 luciferase activity and evaluating the mRNA expression of NFATc1-responsive genes related to the osteoclastic migration/fusion. These results suggested anti-osteoclastogenic activity of doramapimod via inhibiting migration/fusion of pre-osteoclasts and NFATc1 activity.
Collapse
Affiliation(s)
- Seong-Hee Moon
- Laboratory of Translational Therapeutics, Pharmacology Research Center, Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 305-600, Republic of Korea; Department of Biology, Chungnam National University, Daejeon 305-764, Republic of Korea
| | - Sik-Won Choi
- Laboratory of Translational Therapeutics, Pharmacology Research Center, Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 305-600, Republic of Korea
| | - Seong Hwan Kim
- Laboratory of Translational Therapeutics, Pharmacology Research Center, Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon 305-600, Republic of Korea.
| |
Collapse
|
43
|
Fukuma Y, Sakai E, Nishishita K, Okamoto K, Tsukuba T. Cafestol has a weaker inhibitory effect on osteoclastogenesis than kahweol and promotes osteoblast differentiation. Biofactors 2015; 41:222-31. [PMID: 26154488 DOI: 10.1002/biof.1218] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/08/2015] [Accepted: 05/27/2015] [Indexed: 01/09/2023]
Abstract
Bone homeostasis is regulated by a balance between osteoclast (OCL)-mediated bone resorption and osteoblast (OBL)-mediated bone formation. Thus, developing a compound that simultaneously inhibits OCL function and promotes OBL function would be useful as a new medical therapy for bone diseases. Here, we examined the effects of cafestol, a coffee diterpene, on the differentiation of OCLs and OBLs. Cafestol prevented OCL formation in a dose-dependent manner and suppressed the bone-resorbing activity of OCLs. Interestingly, the viability of OCLs treated with 10-50 µM cafestol was significantly higher than that of untreated cells. At the molecular level, cafestol markedly decreased RANKL-induced phosphorylation of extracellular signal-regulated kinase (Erk) and inhibitor of nuclear factor kappa B alpha (IκBα). Compared to kahweol, another coffee-specific diterpene, the inhibitory effects of cafestol were milder on OCL differentiation, and cafestol and kahweol showed different characteristics in induction of the phase ΙΙ antioxidant enzymes and sensitivities in nuclear factor-erythroid 2-related factor 2 (Nrf2)-deficient BMMs. In addition to inhibiting OCLs, cafestol enhanced the differentiation of osteoblastic cells by increasing the mRNA levels of differentiation markers. Thus, cafestol inhibits OCL differentiation and promotes OBL differentiation, suggesting that cafestol may be a novel agent for bone diseases.
Collapse
Affiliation(s)
- Yutaka Fukuma
- Division of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan
| | - Eiko Sakai
- Division of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan
| | - Kazuhisa Nishishita
- Division of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan
| | - Kuniaki Okamoto
- Division of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan
| | - Takayuki Tsukuba
- Division of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, Sakamoto 1-7-1, Nagasaki, Japan
| |
Collapse
|
44
|
Callaway DA, Jiang JX. Reactive oxygen species and oxidative stress in osteoclastogenesis, skeletal aging and bone diseases. J Bone Miner Metab 2015; 33:359-70. [PMID: 25804315 DOI: 10.1007/s00774-015-0656-4] [Citation(s) in RCA: 292] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/12/2015] [Indexed: 12/11/2022]
Abstract
Osteoclasts are cells derived from bone marrow macrophages and are important in regulating bone resorption during bone homeostasis. Understanding what drives osteoclast differentiation and activity is important when studying diseases characterized by heightened bone resorption relative to formation, such as osteoporosis. In the last decade, studies have indicated that reactive oxygen species (ROS), including superoxide and hydrogen peroxide, are crucial components that regulate the differentiation process of osteoclasts. However, there are still many unanswered questions that remain. This review will examine the mechanisms by which ROS can be produced in osteoclasts as well as how it may affect osteoclast differentiation and activity through its actions on osteoclastogenesis signaling pathways. In addition, the contribution of ROS to the aging-associated disease of osteoporosis will be addressed and how targeting ROS may lead to the development of novel therapeutic treatment options.
Collapse
Affiliation(s)
- Danielle A Callaway
- Department of Biochemistry, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, TX, 78229-3900, USA
| | | |
Collapse
|
45
|
Yashima Y, Okamoto K, Sakai E, Iwatake M, Fukuma Y, Nishishita K, Tsukuba T. Cobalt protoporphyrin represses osteoclastogenesis through blocking multiple signaling pathways. Biometals 2015; 28:725-32. [PMID: 25981584 DOI: 10.1007/s10534-015-9861-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/09/2015] [Indexed: 12/14/2022]
Abstract
Cobalt protoporphyrin (CoPP) is a metallo-protoporphyrin that works as a powerful inducer of heme oxigenase-1 (HO-1) in various tissues and cells. Our recent studies have demonstrated that induction of HO-1 by several reagents inhibited differentiation and activation of osteoclasts (OCLs), which are multinucleated bone resorbing cells. However, the effects of CoPP on osteoclastogenesis remain to be elucidated. In this study, we report that CoPP inhibits receptor activator of nuclear factor κB ligand (RANKL)-induced OCL formation in a dose dependent manner. Importantly, CoPP had little cytotoxicity, but rather enhanced cell proliferation of OCLs. CoPP suppressed the protein levels of nuclear factor of activated T cells cytoplasmic-1 (NFATc1) as well as those of OCLs markers such as Src and cathepsin K, which are transcriptionally regulated by NFATc1 in mature OCLs. Western blot analyses also showed that CoPP abolished RANKL-stimulated phosphorylation of several major signaling pathways such as IκB, Akt, ERK, JNK and p38 MAPKs in OCL precursor cells. Thus, our results show that CoPP represses osteoclastogenesis through blocking multiple signaling pathways.
Collapse
Affiliation(s)
- Yuka Yashima
- Division of Dental Pharmacology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1, Sakamoto, Nagasaki, 852-8588, Japan
| | | | | | | | | | | | | |
Collapse
|
46
|
Shimada-Sugawara M, Sakai E, Okamoto K, Fukuda M, Izumi T, Yoshida N, Tsukuba T. Rab27A regulates transport of cell surface receptors modulating multinucleation and lysosome-related organelles in osteoclasts. Sci Rep 2015; 5:9620. [PMID: 25882854 PMCID: PMC5381753 DOI: 10.1038/srep09620] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 03/10/2015] [Indexed: 11/21/2022] Open
Abstract
Rab27A regulates transport of lysosome-related organelles (LROs) and release of secretory granules in various types of cells. Here, we identified up-regulation of Rab27A during differentiation of osteoclasts (OCLs) from bone-marrow macrophages (BMMs), by DNA microarray analysis. Rab27A deficiency in OCLs, using small interfering RNA (siRNA) knockdown in RAW-D cell line or BMMs derived from ashen mice, which display genetic defects in Rab27A expression, induced multinucleated and giant cells. Upon stimulation with macrophage-colony stimulating factor (M-CSF) and receptor activator of nuclear factor kappa-B ligand (RANKL), essential cytokines for OCL differentiation, phosphorylation levels of extracellular signal-regulated kinase (Erk), proto-oncogene tyrosine-protein kinase (Src), and p-38 were slightly enhanced in ashen BMMs than in wild-type BMMs. The cell surface level of c-fms, an M-CSF receptor, was slightly higher in ashen BMMs than in wild-type BMMs, and down-regulation of RANK, a RANKL receptor, was delayed. In addition to receptors, OCLs derived from ashen mice exhibited aberrant actin ring formation, abnormal subcellular localization of lysosome-associated membrane protein (LAMP2) and cathepsin K (CTSK), and marked reduction in resorbing activity. Thus, these findings suggest that Rab27A regulates normal transport of cell surface receptors modulating multinucleation and LROs in OCLs.
Collapse
Affiliation(s)
- Megumi Shimada-Sugawara
- 1] Division of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan [2] Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| | - Eiko Sakai
- Division of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| | - Kuniaki Okamoto
- Division of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Developmental Biology and Neurosciences, Graduate School of Life Sciences, Tohoku University, Aobayama, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Tetsuro Izumi
- Department of Molecular Medicine, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8512, Japan
| | - Noriaki Yoshida
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| | - Takayuki Tsukuba
- Division of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| |
Collapse
|
47
|
Iwatake M, Okamoto K, Tanaka T, Tsukuba T. Castalagin Exerts Inhibitory Effects on Osteoclastogenesis Through Blocking a Broad Range of Signaling Pathways with Low Cytotoxicity. Phytother Res 2015; 29:917-24. [DOI: 10.1002/ptr.5333] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 02/21/2015] [Accepted: 02/27/2015] [Indexed: 01/16/2023]
Affiliation(s)
- Mayumi Iwatake
- Division of Dental Pharmacology; Nagasaki University Graduate School of Biomedical Sciences; 1-7-1 Sakamoto Nagasaki 852-8588 Japan
| | - Kuniaki Okamoto
- Division of Dental Pharmacology; Nagasaki University Graduate School of Biomedical Sciences; 1-7-1 Sakamoto Nagasaki 852-8588 Japan
| | - Takashi Tanaka
- Division of Natural Product Chemistry; Nagasaki University Graduate School of Biomedical Sciences; 1-14 Bunkyo Nagasaki 852-8521 Japan
| | - Takayuki Tsukuba
- Division of Dental Pharmacology; Nagasaki University Graduate School of Biomedical Sciences; 1-7-1 Sakamoto Nagasaki 852-8588 Japan
| |
Collapse
|
48
|
Chuang JY, Chang PC, Shen YC, Lin C, Tsai CF, Chen JH, Yeh WL, Wu LH, Lin HY, Liu YS, Lu DY. Regulatory effects of fisetin on microglial activation. Molecules 2014; 19:8820-39. [PMID: 24972270 PMCID: PMC6271444 DOI: 10.3390/molecules19078820] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 06/13/2014] [Accepted: 06/18/2014] [Indexed: 12/31/2022] Open
Abstract
Increasing evidence suggests that inflammatory processes in the central nervous system that are mediated by microglial activation play a key role in neurodegeneration. Fisetin, a plant flavonol commonly found in fruits and vegetables, is frequently added to nutritional supplements due to its antioxidant properties. In the present study, treatment with fisetin inhibited microglial cell migration and ROS (reactive oxygen species) production. Treatment with fisetin also effectively inhibited LPS plus IFN-γ-induced nitric oxide (NO) production, and inducible nitric oxide synthase (iNOS) expression in microglial cells. Furthermore, fisetin also reduced expressions of iNOS and NO by stimulation of peptidoglycan, the major component of the Gram-positive bacterium cell wall. Fisetin also inhibited the enhancement of LPS/IFN-γ- or peptidoglycan-induced inflammatory mediator IL (interlukin)-1 β expression. Besides the antioxidative and anti-inflammatory effects of fisetin, our study also elucidates the manner in fisetin-induced an endogenous anti-oxidative enzyme HO (heme oxygenase)-1 expression. Moreover, the regulatory molecular mechanism of fisetin-induced HO-1 expression operates through the PI-3 kinase/AKT and p38 signaling pathways in microglia. Notably, fisetin also significantly attenuated inflammation-related microglial activation and coordination deficit in mice in vivo. These findings suggest that fisetin may be a candidate agent for the development of therapies for inflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Jing-Yuan Chuang
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 40402, Taiwan.
| | - Pei-Chun Chang
- Department of Bioinformatics, Asia University, Taichung 41354, Taiwan.
| | - Yi-Chun Shen
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 40402, Taiwan.
| | - Chingju Lin
- Department of Physiology, School of Medicine, China Medical University, Taichung 40402, Taiwan.
| | - Cheng-Fang Tsai
- Department of Biotechnology, Asia University, Taichung 41354, Taiwan.
| | - Jia-Hong Chen
- Department of General Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 42743, Taiwan.
| | - Wei-Lan Yeh
- Department of Cell and Tissue Engineering, Changhua Christian Hospital, Changhua 500, Taiwan.
| | - Ling-Hsuan Wu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
| | - Hsiao-Yun Lin
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan.
| | - Yu-Shu Liu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
| | - Dah-Yuu Lu
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
49
|
Genistein inhibits osteoclastic differentiation of RAW 264.7 cells via regulation of ROS production and scavenging. Int J Mol Sci 2014; 15:10605-21. [PMID: 24927148 PMCID: PMC4100171 DOI: 10.3390/ijms150610605] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 05/22/2014] [Accepted: 05/30/2014] [Indexed: 11/19/2022] Open
Abstract
Genistein, a phytoestrogen, has been demonstrated to have a bone-sparing and antiresorptive effect. Genistein can inhibit the osteoclast formation of receptor activator of nuclear factor-κB ligand (RANKL)-induced RAW 264.7 cells by preventing the translocation of nuclear factor-κB (NF-κB), a redox-sensitive factor, to the nucleus. Therefore, the suppressive effect of genistein on the reactive oxygen species (ROS) level during osteoclast differentiation and the mechanism associated with the control of ROS levels by genistein were investigated. The cellular antioxidant capacity and inhibitory effect of genistein were confirmed. The translation and activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 1 (Nox1), as well as the disruption of the mitochondrial electron transport chain system were obviously suppressed by genistein in a dose-dependent manner. The induction of phase II antioxidant enzymes, such as superoxide dismutase 1 (SOD1) and heme oxygenase-1 (HO-1), was enhanced by genistein. In addition, the translational induction of nuclear factor erythroid 2-related factor 2 (Nrf2) was notably increased by genistein. These results provide that the inhibitory effects of genistein on RANKL-stimulated osteoclast differentiation is likely to be attributed to the control of ROS generation through suppressing the translation and activation of Nox1 and the disruption of the mitochondrial electron transport chain system, as well as ROS scavenging through the Nrf2-mediated induction of phase II antioxidant enzymes, such as SOD1 and HO-1.
Collapse
|
50
|
Heme oxygenase-1 (HO-1) expression in prostate cancer cells modulates the oxidative response in bone cells. PLoS One 2013; 8:e80315. [PMID: 24224047 PMCID: PMC3817116 DOI: 10.1371/journal.pone.0080315] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/06/2013] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer (PCa) is a leading cause of death among males. It is currently estimated that inflammatory responses are linked to 15-20% of all deaths from cancer worldwide. PCa is dominated by complications arising from metastasis to the bone where the tumor cells interact with the bone microenvironment impairing the balance between bone formation and degradation. However, the molecular nature of this interaction is not completely understood. Heme oxygenase-1 (HO-1) counteracts oxidative damage and inflammation. Previous studies from our laboratory showed that HO-1 is implicated in PCa, demonstrating that endogenous HO-1 inhibits bone derived-prostate cancer cells proliferation, invasion and migration and decreases tumor growth and angiogenesis in vivo. The aim of this work was to analyze the impact of HO-1 modulated PCa cells on osteoblasts proliferation in vitro and on bone remodeling in vivo. Using a co-culture system of PC3 cells with primary mice osteoblasts (PMOs), we demonstrated that HO-1 pharmacological induction (hemin treatment) abrogated the diminution of PMOs proliferation induced by PCa cells and decreased the expression of osteoclast-modulating factors in osteoblasts. No changes were detected in the expression of genes involved in osteoblasts differentiation. However, co-culture of hemin pre-treated PC3 cells (PC3 Hem) with PMOs provoked an oxidative status and activated FoxO signaling in osteoblasts. The percentage of active osteoblasts positive for HO-1 increased in calvarias explants co-cultured with PC3 Hem cells. Nuclear HO-1 expression was detected in tumors generated by in vivo bone injection of HO-1 stable transfected PC3 (PC3HO-1) cells in the femur of SCID mice. These results suggest that HO-1 has the potential to modify the bone microenvironment impacting on PCa bone metastasis.
Collapse
|