1
|
Kirk B, Lombardi G, Duque G. Bone and muscle crosstalk in ageing and disease. Nat Rev Endocrinol 2025; 21:375-390. [PMID: 40011751 DOI: 10.1038/s41574-025-01088-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/30/2025] [Indexed: 02/28/2025]
Abstract
Interorgan communication between bone and skeletal muscle is central to human health. A dysregulation of bone-muscle crosstalk is implicated in several age-related diseases. Ageing-associated changes in endocrine, inflammatory, nutritional and biomechanical stimuli can influence the differentiation capacity, function and survival of mesenchymal stem cells and bone-forming and muscle-forming cells. Consequently, the secretome phenotype of bone and muscle cells is altered, leading to impaired crosstalk and, ultimately, catabolism of both tissues. Adipose tissue acts as a third player in the bone-muscle interaction by secreting factors that affect bone and muscle cells. Physical exercise remains the key biological stimulus for bone-muscle crosstalk, either directly via the release of cytokines from bone, muscle or adipocytes, or indirectly through extracellular vesicles. Overall, bone-muscle crosstalk is considered an inherent process necessary to maintain the structure and function of both tissues across the life cycle. This Review summarizes the latest biomedical advances in bone-muscle crosstalk as it pertains to human ageing and disease. We also outline future research priorities to accommodate the understanding of this rapidly emerging field.
Collapse
Affiliation(s)
- Ben Kirk
- Department of Medicine, Western Health, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, Melbourne, Victoria, Australia
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry & Advanced Diagnostics, IRCCS Ospedale Galeazzi-Sant'Ambrogio, Milan, Italy
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| | - Gustavo Duque
- Department of Medicine, Western Health, Melbourne Medical School, University of Melbourne, Melbourne, Victoria, Australia.
- Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, Melbourne, Victoria, Australia.
- Bone, Muscle & Geroscience Group, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
- Dr. Joseph Kaufmann Chair in Geriatric Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
2
|
Li XY, Gu XY, Li XM, Yan JG, Mao XL, Yu Q, Du YL, Kurihara H, Yan CY, Li WX. Supplementation with carnosine, a food-derived bioactive dipeptide, alleviates dexamethasone-induced oxidative stress and bone impairment via the NRF2 signaling pathway. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:1091-1104. [PMID: 39291490 DOI: 10.1002/jsfa.13899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/07/2024] [Accepted: 08/31/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Carnosine, a natural bioactive dipeptide derived from meat muscle, possesses strong antioxidant properties. Dexamethasone, widely employed for treating various inflammatory diseases, raises concerns regarding its detrimental effects on bone health. This study aimed to investigate the protective effects of carnosine against dexamethasone-induced oxidative stress and bone impairment, along with its underlying mechanisms, utilizing chick embryos and a zebrafish model in vivo, as well as MC3T3-E1 cells in vitro. RESULTS Our findings revealed that carnosine effectively mitigated bone injury in dexamethasone-exposed chick embryos, accompanied by reduced oxidative stress. Further investigation demonstrated that carnosine alleviated impaired osteoblastic differentiation in MC3T3-E1 cells and zebrafish by suppressing the excessive production of reactive oxygen species (ROS) and enhancing the activity of antioxidant enzymes such as superoxide dismutase (SOD) and glutathione peroxidase (GPX). Moreover, mechanistic studies elucidated that carnosine promoted the expression and nuclear translocation of nuclear factor erythroid 2-related factor 2 (NRF2), thereby facilitating the transcription of its downstream antioxidant response elements, including heme oxyense-1 (HO-1), glutamate cysteine ligase modifier (GCLM), and glutamate cysteine ligase catalytic (GCLC) to counteract dexamethasone-induced oxidative stress. CONCLUSION Overall, this study underscores the potential therapeutic efficacy of carnosine in mitigating oxidative stress and bone damage induced by dexamethasone exposure, shedding light on its underlying mechanism of action by activating the NRF2 signaling pathway. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xi-You Li
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Xiao-Yuan Gu
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Xiao-Min Li
- Perfect (Guangdong) Commodity Co., LTD, Zhongshan, China
| | - Jian-Gang Yan
- Perfect (Guangdong) Commodity Co., LTD, Zhongshan, China
| | - Xin-Liang Mao
- Perfect (Guangdong) Commodity Co., LTD, Zhongshan, China
| | - Qin Yu
- Perfect (Guangdong) Commodity Co., LTD, Zhongshan, China
| | - Yu-Lan Du
- Perfect (Guangdong) Commodity Co., LTD, Zhongshan, China
| | - Hiroshi Kurihara
- Perfect (Guangdong) Commodity Co., LTD, Zhongshan, China
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, College of Pharmacy, Jinan University, Guangzhou, China
| | - Chang-Yu Yan
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, College of Pharmacy, Jinan University, Guangzhou, China
| | - Wei-Xi Li
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China
| |
Collapse
|
3
|
Zhang X, Li H, Chen L, Wu Y, Li Y. NRF2 in age-related musculoskeletal diseases: Role and treatment prospects. Genes Dis 2024; 11:101180. [PMID: 39281838 PMCID: PMC11400624 DOI: 10.1016/j.gendis.2023.101180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 10/06/2023] [Accepted: 10/31/2023] [Indexed: 09/18/2024] Open
Abstract
The NRF2 pathway is a metabolic- and redox-sensitive signaling axis in which the transcription factor controls the expression of a multitude of genes that enable cells to survive environmental stressors, such as oxidative stress, mainly by inducing the expression of cytoprotective genes. Basal NRF2 levels are maintained under normal physiological conditions, but when exposed to oxidative stress, cells activate the NRF2 pathway, which is crucial for supporting cell survival. Recently, the NRF2 pathway has been found to have novel functions in metabolic regulation and interplay with other signaling pathways, offering novel insights into the treatment of various diseases. Numerous studies have shown that targeting its pathway can effectively investigate the development and progression of age-related musculoskeletal diseases, such as sarcopenia, osteoporosis, osteoarthritis, and intervertebral disc degeneration. Appropriate regulation of the NRF2 pathway flux holds promise as a means to improve musculoskeletal function, thereby providing a new avenue for drug treatment of age-related musculoskeletal diseases in clinical settings. The review summarized an overview of the relationship between NRF2 and cellular processes such as oxidative stress, apoptosis, inflammation, mitochondrial dysfunction, ferroptosis, and autophagy, and explores the potential of targeted NRF2 regulation in the treatment of age-related musculoskeletal diseases.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hengzhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Lin Chen
- Department of Health and Physical Education, Jianghan University, Wuhan, Hubei 430056, China
| | - Yuxiang Wu
- Department of Health and Physical Education, Jianghan University, Wuhan, Hubei 430056, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
4
|
Abdelrahman RS, Abdelmageed ME. Hepatoprotective effects of the xanthine oxidase inhibitor Febuxostat against thioacetamide-induced liver injury in rats: The role of the Nrf2/ HO-1 and TLR4/ NF-κB pathways. Food Chem Toxicol 2024; 194:115087. [PMID: 39489394 DOI: 10.1016/j.fct.2024.115087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/27/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Experimental models of liver injury have been established utilizing thioacetamide (TAA), a classic liver toxic chemical that causes organ damage via oxidative stress and inflammatory induction. This study examined the impact of Febuxostat (a xanthine oxidase inhibitor; Febu, 10-15 mg/kg, orally) against TAA (500 mg/kg, i.p.) -induced liver injury in rats. Febu significantly attenuated TAA-induced alterations in liver function parameters, in addition to promoting hepatic antioxidant effects through a significant elevation of Heme-oxygenase-1(HO-1), nuclear factor erythroid 2-related factor2 (Nrf2), reduced glutathione (GSH) and superoxide dismutase (SOD) levels and reduction in hepatic malondialdehyde (MDA) content. Moreover, Febu improved the hepatic anti-inflammatory status by increasing the anti-inflammatory cytokine Interleukin (IL-10) level and reducing the levels of the pro-inflammatory cytokines (Nuclear factor kappa B (NF-κB), IL-1β, high-mobility group box1 (HMGB1), receptor for advanced glycation end products (RAGE), and toll-like receptor4 (TLR4) levels, in addition to suppressing the increased protein and mRNA expression levels of tumor necrosis factor alpha (TNF-α) and IL-6, hepatic expression of TNF-α and activated mitogen-activated protein kinases (p-JNK/p-p38 MAPK). Histopathologically, Febu markedly normalized TAA-induced alteration in liver sections. In conclusion, Febu, in a dose-dependent fashion, refines TAA-induced hepatotoxicity by enhancing antioxidant capabilities and decreasing inflammatory signals.
Collapse
Affiliation(s)
- Rehab S Abdelrahman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Taibah University, Al-Madina Al-Munawwarah, 30001, Saudi Arabia
| | - Marwa E Abdelmageed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt.
| |
Collapse
|
5
|
Chen H, Weng Z, Kalinowska M, Xiong L, Wang L, Song H, Xiao J, Wang F, Shen X. Anti-osteoporosis effect of bioactives in edible medicinal plants: a comprehensive review. Crit Rev Food Sci Nutr 2024:1-17. [PMID: 39093554 DOI: 10.1080/10408398.2024.2386449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Current treatments for osteoporosis include a calcium-rich diet, adequate exercise, and medication. Many synthetic drugs, although fast-acting, can cause a range of side effects for patients when taken over a long period, such as irritation of the digestive tract and a burden on the kidneys. As the world's population ages, the prevalence of osteoporosis is increasing, and the development of safe and effective treatments is urgently needed. Active compounds in edible and medicinal homologous plants have been used for centuries to improve bone quality. It is possible to employ them as dietary supplements to prevent osteoporosis. In this review, we analyze the influencing factors of osteoporosis and systematically summarize the research progress on the anti-osteoporosis effects of active compounds in edible and medicinal homologous plants. The literature suggests that some naturally occurring active compounds in edible and medicinal homologous plants can inhibit bone loss, prevent the degeneration of bone cell microstructure, and reduce bone fragility through alleviating oxidative stress, regulating autophagy, anti-inflammation, improving gut flora, and regulating estrogen level with little side effects. Our review provides useful guidance for the use of edible and medicinal homologous plants and the development of safer novel anti-osteoporosis dietary supplements.
Collapse
Affiliation(s)
- Huiling Chen
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Zebin Weng
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Monika Kalinowska
- Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Bialystok, Poland
| | - Ling Xiong
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Luanfeng Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Jianbo Xiao
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
- Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA) - CITEXVI, Universidade de Vigo, Nutrition and Bromatology Group, Vigo, Spain
| | - Fang Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
- Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, USA
| | - Xinchun Shen
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| |
Collapse
|
6
|
Dittmar MC, Tohidnezhad M, Fragoulis A, Bücker A, Stein M, Pufe T, Kubo Y. Pharmacological effects of methysticin and L-sulforaphane through the Nrf2/ARE signaling pathway in MLO-Y4 osteocytes: in vitro study. Ann Anat 2024; 254:152260. [PMID: 38521364 DOI: 10.1016/j.aanat.2024.152260] [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: 08/15/2023] [Revised: 02/04/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Oxidative stress plays a crucial role in the pathogenesis of many skeletal diseases by inducing osteocyte death. The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is a master regulator of various antioxidant gene expressions through antioxidant response element (ARE) against cellular oxidative stress and can be induced by various stimulants, including the phytochemicals methysticin (MET) and L-sulforaphane (SFN). This study aimed to establish an osteocyte in vitro model to investigate the pharmacological effects of MET and SFN on the Nrf2/ARE pathway. METHODS MLO-Y4 murine osteocytes and the stably transduced MLO-Y4-SIN-lenti-ARE reporter gene cell line were used. MET and SFN were used as Nrf2 inducers. The cytotoxicity of MET, SFN, and hydrogen peroxide (H2O2) was evaluated using the CytoTox-Glo™ Assay. Time- and dose-dependent ARE induction was examined by Monoluciferase Assay. The mRNA and protein expressions of Nrf2 target markers, such as heme-oxygenase 1 (Ho-1), NADPH quinone dehydrogenase 1 (Nqo1), and thioredoxin reductase 1 (Txnrd1), were detected by RT-qPCR, Western Blot, and immunofluorescence staining, respectively. Osteogenesis markers, osteopontin, and osteocalcin were compared with and without treatment by immunofluorescence staining. RESULTS The experimental data showed that MET and SFN induced ARE activity in a time- and dose-dependent manner and increased the mRNA and protein expression of antioxidant markers compared to vehicle-treated controls. The protein expression of osteopontin and osteocalcin in the samples treated with SFN were significantly higher than without treatment, and the number of cell death treated with SFN was significantly lower than without treatment under H2O2-induced stress conditions. CONCLUSIONS Nrf2 inducers MET and SFN increased the mRNA expression of antioxidant genes through the Nrf2/ARE pathway in osteocytes. Notably, SFN increased the protein expression of osteocyte-associated osteogenic markers and suppressed cell death under H2O2-induced stress condition. Thus, Nrf2 stimulators can exert stress-relieving and osteogenic effects on osteocytes.
Collapse
Affiliation(s)
- Maja Charlotte Dittmar
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, Aachen 52074, Germany
| | - Mersedeh Tohidnezhad
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, Aachen 52074, Germany
| | - Athanassios Fragoulis
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, Aachen 52074, Germany
| | - Annette Bücker
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, Aachen 52074, Germany
| | - Matthias Stein
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, Aachen 52074, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, Aachen 52074, Germany
| | - Yusuke Kubo
- Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, Aachen 52074, Germany; Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| |
Collapse
|
7
|
Bleichman I, Hiram-Bab S, Gabet Y, Savion N. S-Allylmercapto-N-Acetylcysteine (ASSNAC) Attenuates Osteoporosis in Ovariectomized (OVX) Mice. Antioxidants (Basel) 2024; 13:474. [PMID: 38671921 PMCID: PMC11047400 DOI: 10.3390/antiox13040474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/11/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Osteoporosis is a bone-debilitating disease, demonstrating a higher prevalence in post-menopausal women due to estrogen deprivation. One of the main mechanisms underlying menopause-related bone loss is oxidative stress. S-allylmercapto-N-acetylcysteine (ASSNAC) is a nuclear factor erythroid 2-related factor 2 (Nrf2) activator and cysteine supplier, previously shown to have anti-oxidation protective effects in cultured cells and animal models. Here, we studied the therapeutic potential of ASSNAC with and without Alendronate in ovariectomized (OVX) female mice. The experimental outcome included (i) femur and L3 lumbar vertebra morphometry via Micro-Computed Tomography (μCT); (ii) bone remodeling (formation vs. resorption); and (iii) oxidative stress markers in bone marrow (BM) cells. Four weeks after OVX, there was a significant bone loss that remained evident after 8 weeks, as demonstrated via µCT in the femur (cortical and trabecular bone compartments) and vertebra (trabecular bone). ASSNAC at a dose of 50 mg/Kg/day prevented bone loss after the four-week treatment but had no significant effect after 8 weeks, while ASSNAC at a dose of 20 mg/Kg/day significantly protected against bone loss after 8 weeks of treatment. Alendronate prevented ovariectomy-induced bone loss, and combining it with ASSNAC further augmented this effect. OVX mice demonstrated high serum levels of both C-terminal cross-linked telopeptides of type I collagen (CTX) (bone resorption) and procollagen I N-terminal propeptide (P1NP) (bone formation) after 2 weeks, and these returned to control levels after 8 weeks. Alendronate, ASSNAC and their combination decreased CTX and increased P1NP. Alendronate induced oxidative stress as reflected by decreased glutathione and increased malondialdehyde (MDA) levels, and combining it with ASSNAC partially attenuated these changes. These results portray the therapeutic potential of ASSNAC for the management of post-menopausal osteoporosis. Furthermore, ASSNAC ameliorates the Alendronate-associated oxidative stress, suggesting its potential to prevent Alendronate side effects as well as improve its bone-protective effect.
Collapse
Affiliation(s)
- Itay Bleichman
- Department of Human Molecular Genetics and Biochemistry and Goldschleger Eye Research Institute, School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel-Aviv 6997801, Israel;
| | - Sahar Hiram-Bab
- Department of Anatomy and Anthropology, School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel-Aviv 6997801, Israel; (S.H.-B.); (Y.G.)
| | - Yankel Gabet
- Department of Anatomy and Anthropology, School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel-Aviv 6997801, Israel; (S.H.-B.); (Y.G.)
| | - Naphtali Savion
- Department of Human Molecular Genetics and Biochemistry and Goldschleger Eye Research Institute, School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel-Aviv 6997801, Israel;
| |
Collapse
|
8
|
Materozzi M, Resnati M, Facchi C, Trudu M, Orfanelli U, Perini T, Gennari L, Milan E, Cenci S. A novel proteomic signature of osteoclast differentiation unveils the deubiquitinase UCHL1 as a necessary osteoclastogenic driver. Sci Rep 2024; 14:7290. [PMID: 38538704 PMCID: PMC10973525 DOI: 10.1038/s41598-024-57898-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/22/2024] [Indexed: 04/02/2024] Open
Abstract
Bone destruction, a major source of morbidity, is mediated by heightened differentiation and activity of osteoclasts (OC), highly specialized multinucleated myeloid cells endowed with unique bone-resorptive capacity. The molecular mechanisms regulating OC differentiation in the bone marrow are still partly elusive. Here, we aimed to identify new regulatory circuits and actionable targets by comprehensive proteomic characterization of OCgenesis from mouse bone marrow monocytes, adopting two parallel unbiased comparative proteomic approaches. This work disclosed an unanticipated protein signature of OCgenesis, with most gene products currently unannotated in bone-related functions, revealing broad structural and functional cellular reorganization and divergence from macrophagic immune activity. Moreover, we identified the deubiquitinase UCHL1 as the most upregulated cytosolic protein in differentiating OCs. Functional studies proved it essential, as UCHL1 genetic and pharmacologic inhibition potently suppressed OCgenesis. Furthermore, proteomics and mechanistic dissection showed that UCHL1 supports OC differentiation by restricting the anti-OCgenic activity of NRF2, the transcriptional activator of the canonical antioxidant response, through redox-independent stabilization of the NRF2 inhibitor, KEAP1. Besides offering a valuable experimental framework to dissect OC differentiation, our study discloses the essential role of UCHL1, exerted through KEAP1-dependent containment of NRF2 anti-OCgenic activity, yielding a novel potential actionable pathway against bone loss.
Collapse
Affiliation(s)
- Maria Materozzi
- Age Related Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy.
- Università Vita-Salute San Raffaele, Milan, Italy.
| | - Massimo Resnati
- Age Related Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Cecilia Facchi
- Age Related Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Matteo Trudu
- Age Related Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Ugo Orfanelli
- Age Related Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Tommaso Perini
- Age Related Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Luigi Gennari
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Enrico Milan
- Age Related Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy.
- Università Vita-Salute San Raffaele, Milan, Italy.
| | - Simone Cenci
- Age Related Diseases Unit, IRCCS Ospedale San Raffaele, Milan, Italy.
- Università Vita-Salute San Raffaele, Milan, Italy.
| |
Collapse
|
9
|
Vahidinia Z, Azami Tameh A, Barati S, Izadpanah M, Seyed Hosseini E. Nrf2 activation: a key mechanism in stem cell exosomes-mediated therapies. Cell Mol Biol Lett 2024; 29:30. [PMID: 38431569 PMCID: PMC10909300 DOI: 10.1186/s11658-024-00551-3] [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: 12/13/2023] [Accepted: 02/20/2024] [Indexed: 03/05/2024] Open
Abstract
Exosomes are nano-sized membrane extracellular vesicles which can be released from various types of cells. Exosomes originating from inflammatory or injured cells can have detrimental effects on recipient cells, while exosomes derived from stem cells not only facilitate the repair and regeneration of damaged tissues but also inhibit inflammation and provide protective effects against various diseases, suggesting they may serve as an alternative strategy of stem cells transplantation. Exosomes have a fundamental role in communication between cells, through the transfer of proteins, bioactive lipids and nucleic acids (like miRNAs and mRNAs) between cells. This transfer significantly impacts both the physiological and pathological functions of recipient cells. Nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor, is able to mitigate damage caused by oxidative stress and inflammation through various signaling pathways. The positive effects resulting from the activation of the Nrf2 signaling pathway in different disorders have been documented in various types of literature. Studies have confirmed that exosomes derived from stem cells could act as Nrf2 effective agonists. However, limited studies have explored the Nrf2 role in the therapeutic effects of stem cell-derived exosomes. This review provides a comprehensive overview of the existing knowledge concerning the role of Nrf2 signaling pathways in the impact exerted by stem cell exosomes in some common diseases.
Collapse
Affiliation(s)
- Zeinab Vahidinia
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Abolfazl Azami Tameh
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Shirin Barati
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran
| | - Melika Izadpanah
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elahe Seyed Hosseini
- Gametogenesis Research Center, Institute for Basic Sciences, Kashan University of Medical Science, Kashan, Iran
| |
Collapse
|
10
|
Andrie KM, Palmer DR, Wahl O, Bork S, Campbell M, Walsh MA, Sanford J, Musci RV, Hamilton KL, Santangelo KS, Puttlitz CM. Treatment with PB125 ® Increases Femoral Long Bone Strength in 15-Month-Old Female Hartley Guinea Pigs. Ann Biomed Eng 2024; 52:671-681. [PMID: 38044413 DOI: 10.1007/s10439-023-03415-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023]
Abstract
Nuclear factor-erythroid 2-related factor-2 (Nrf2) is a transcription factor that serves as a master regulator of anti-inflammatory agents, phase I xenobiotic, and phase II antioxidant enzymes, all of which provide a cytoprotective role during disease progression. We hypothesized that oral administration of a purported phytochemical Nrf2-activator, PB125®, would increase long bone strength in aging Hartley guinea pigs, a model prone to musculoskeletal decline. Male (N = 56) and female (N = 56) guinea pigs were randomly assigned to receive daily oral treatment with either PB125® or vehicle control. Animals were treated for a consecutive 3-months (starting at 2-months of age) or 10-months (starting at 5-months of age) and sacrificed at 5-months or 15-months of age, respectively. Outcome measures included: (1) ANY-maze™ enclosure monitoring, (2) quantitative microcomputed tomography, and (3) biomechanical testing. Treatment with PB125® for 10 months resulted in increased long bone strength as determined by ultimate bending stress in female Hartley guinea pigs. In control groups, increasing age resulted in significant effects on geometric and structural properties of long bones, as well as a trending increase in ultimate bending stress. Furthermore, both age and sex had a significant effect on the geometric properties of both cortical and trabecular bone. Collectively, this work suggests that this nutraceutical may serve as a promising target and preventive measure in managing the decline in bone mass and quality documented in aging patients. Auxiliary to this main goal, this work also capitalized upon 5 and 15-month-old male and female animals in the control group to characterize age- and sex-specific differences on long bone geometric, structural, and material properties in this animal model.
Collapse
Affiliation(s)
- K M Andrie
- Department of Microbiology, Immunology & Pathology, Colorado State University, 1619 Campus Delivery, Fort Collins, CO, 80523-1619, USA
| | - D R Palmer
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering, School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - O Wahl
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering, School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - S Bork
- Department of Microbiology, Immunology & Pathology, Colorado State University, 1619 Campus Delivery, Fort Collins, CO, 80523-1619, USA
| | - M Campbell
- Department of Microbiology, Immunology & Pathology, Colorado State University, 1619 Campus Delivery, Fort Collins, CO, 80523-1619, USA
| | - M A Walsh
- Department of Health and Exercise Science, Colorado State University, 1582 Campus Delivery, Fort Collins, CO, 80523-1582, USA
| | - J Sanford
- Department of Microbiology, Immunology & Pathology, Colorado State University, 1619 Campus Delivery, Fort Collins, CO, 80523-1619, USA
| | - R V Musci
- Department of Health and Exercise Science, Colorado State University, 1582 Campus Delivery, Fort Collins, CO, 80523-1582, USA
| | - Karyn L Hamilton
- Department of Health and Exercise Science, Colorado State University, 1582 Campus Delivery, Fort Collins, CO, 80523-1582, USA.
- Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO, USA.
| | - Kelly S Santangelo
- Department of Microbiology, Immunology & Pathology, Colorado State University, 1619 Campus Delivery, Fort Collins, CO, 80523-1619, USA.
- Columbine Health Systems Center for Healthy Aging, Colorado State University, Fort Collins, CO, USA.
| | - Christian M Puttlitz
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering, School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA.
- Department of Mechanical Engineering, Colorado State University, 1374 Campus Delivery, Fort Collins, CO, 80523-1374, USA.
| |
Collapse
|
11
|
Zou J, Chen H, Fan X, Qiu Z, Zhang J, Sun J. Garcinol prevents oxidative stress-induced bone loss and dysfunction of BMSCs through NRF2-antioxidant signaling. Cell Death Discov 2024; 10:82. [PMID: 38365768 PMCID: PMC10873372 DOI: 10.1038/s41420-024-01855-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 02/18/2024] Open
Abstract
There are multiple published data showing that excessive oxidative stress contributes to bone loss and even bone tissue damage, and it is also correlated with the pathophysiology of bone degenerative diseases, including osteoporosis (OP). Garcinol, a polyisoprenylated benzophenone derivative, has been recently established as an anti-oxidant agent. However, it remains elusive whether Garcinol protects bone marrow mesenchymal stem cells (BMSCs) and bone tissue from oxidative stress-induced damage. Here, we explored the potential effects of Garcinol supplementation in ameliorating oxidative stimulation-induced dysfunction of BMSCs and bone loss in osteoporotic mice. In this study, we verified that Garcinol exerted potent protective functions in the hydrogen peroxide (H2O2)-induced excessive oxidative stress and dysfunction of BMSCs. Besides, Garcinol was also identified to improve the reduced bone mass and abnormal lineage commitment of BMSCs in the condition of OP by suppressing the oxidative stimulation. Subsequent analysis revealed that nuclear factor erythroid 2-related factor 2 (NRF2) might be a key regulator in the sheltering effects of Garcinol on the H2O2-regulated oxidative stress, and the protective functions of Garcinol was mediated by NRF2-antioxidant signaling. Collectively, Garcinol prevented oxidative stress-related BMSC damage and bone loss through the NRF2-antioxidant signaling, which suggested the promising therapeutic values of Garcinol in the treatment of oxidative stress-related bone loss. Therefore, Garcinol might contribute to treating OP.
Collapse
Affiliation(s)
- Jilong Zou
- Department of Orthopaedics, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongjun Chen
- Department of Orthopaedics, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinming Fan
- Department of Orthopaedics, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhenrui Qiu
- Department of Orthopaedics, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiale Zhang
- Department of Orthopaedics, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiabing Sun
- Department of Orthopaedics, the First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
12
|
Che J, Yang X, Jin Z, Xu C. Nrf2: A promising therapeutic target in bone-related diseases. Biomed Pharmacother 2023; 168:115748. [PMID: 37865995 DOI: 10.1016/j.biopha.2023.115748] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023] Open
Abstract
Nuclear factor erythroid-2-related factor 2 (Nrf2) plays an important role in maintaining cellular homeostasis, as it suppresses cell damage caused by external stimuli by regulating the transcription of intracellular defense-related genes. Accumulating evidence has highlighted the crucial role of reduction-oxidation (REDOX) imbalance in the development of bone-related diseases. Nrf2, a transcription factor linked to nuclear factor-erythrocyte 2, plays a pivotal role in the regulation of oxidative stress and induction of antioxidant defenses. Therefore, further investigation of the mechanism and function of Nrf2 in bone-related diseases is essential. Considerable evidence suggests that increased nuclear transcription of Nrf2 in response to external stimuli promotes the expression of intracellular antioxidant-related genes, which in turn leads to the inhibition of bone remodeling imbalance, improved fracture recovery, reduced occurrence of osteoarthritis, and greater tumor resistance. Certain natural extracts can selectively target Nrf2, potentially offering therapeutic benefits for osteogenic arthropathy. In this article, the biological characteristics of Nrf2 are reviewed, the intricate interplay between Nrf2-regulated REDOX imbalance and bone-related diseases is explored, and the potential preventive and protective effects of natural products targeting Nrf2 in these diseases are elucidated. A comprehensive understanding of the role of Nrf2 in the development of bone-related diseases provides valuable insights into clinical interventions and can facilitate the discovery of novel Nrf2-targeting drugs.
Collapse
Affiliation(s)
- Jingmin Che
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China; Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China.
| | - Xiaoli Yang
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China; Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Zhankui Jin
- Department of Orthopedics, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China.
| | - Cuixiang Xu
- Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China; Shaanxi Engineering Research Center of Cell Immunology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
13
|
Lin BH, Ma RX, Wu JT, Du SQ, Lv YY, Yu HN, Zhang W, Mao SM, Liu GY, Bu YT, Chen ZH, Jin C, Wu ZY, Yang L. Cinnamaldehyde Alleviates Bone Loss by Targeting Oxidative Stress and Mitochondrial Damage via the Nrf2/HO-1 Pathway in BMSCs and Ovariectomized Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37917162 DOI: 10.1021/acs.jafc.3c03501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Osteoporosis (OP) is typically brought on by disruption of bone homeostasis. Excessive oxidative stress and mitochondrial dysfunction are believed to be the primary mechanisms underlying this disorder. Therefore, in order to restore bone homeostasis effectively, targeted treatment of oxidative stress and mitochondrial dysfunction is necessary. Cinnamaldehyde (CIN), a small molecule that acts as an agonist for the nuclear factor erythroid 2-related factor (Nrf2), has been found to possess antiapoptotic, anti-inflammatory, and antioxidant properties. We found that CIN, while rescuing apoptosis, can also reduce the accumulation of reactive oxygen species (ROS) to improve mitochondrial dysfunction and thus restore the osteogenic differentiation potential of BMSCs disrupted by hydrogen peroxide (H2O2) exposure. The role of CIN was preliminarily considered to be a consequence of Nrf2/HO-1 axis activation. The ovariectomized mice model further demonstrated that CIN treatment ameliorated oxidative stress in vivo, partially reversing OVX-induced bone loss. This improvement was seen in the trabecular microarchitecture and bone biochemical indices. However, when ML385 was concurrently injected with CIN, the positive effects of CIN were largely blocked. In conclusion, this study sheds light on the intrinsic mechanisms by which CIN regulates BMSCs and highlights the potential therapeutic applications of these findings in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Bing-Hao Lin
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou 325000, China
| | - Run-Xun Ma
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou 325000, China
| | - Jing-Tao Wu
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou 325000, China
| | - Shi-Qi Du
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou 325000, China
- Wenzhou Medical University, Wenzhou 325000, China
| | - Yi-Yun Lv
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou 325000, China
- Wenzhou Medical University, Wenzhou 325000, China
| | - Hao-Nan Yu
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou 325000, China
- Wenzhou Medical University, Wenzhou 325000, China
| | - Wei Zhang
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou 325000, China
| | - Shu-Ming Mao
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou 325000, China
| | - Guang-Yao Liu
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou 325000, China
| | - Yi-Tian Bu
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou 325000, China
| | - Zi-Hao Chen
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou 325000, China
| | - Chen Jin
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou 325000, China
| | - Zong-Yi Wu
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou 325000, China
| | - Lei Yang
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopedics of Zhejiang Province, Wenzhou 325000, China
| |
Collapse
|
14
|
Yao H, Du Y, Jiang B, Liao Y, Zhao Y, Yin M, Li T, Sheng Y, Ji Y, Du M. Sulforaphene suppresses RANKL-induced osteoclastogenesis and LPS-induced bone erosion by activating Nrf2 signaling pathway. Free Radic Biol Med 2023; 207:48-62. [PMID: 37423561 DOI: 10.1016/j.freeradbiomed.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND AND PURPOSE Inflammatory disorders have been found to induce bone loss through sustained and persistent activation of osteoclast differentiation, leading to heightened bone resorption. The current pharmacological interventions for combating bone loss to harbor adverse effects or contraindications. There is a pressing need to identify drugs with fewer side effects. EXPERIMENTAL APPROACH The effect and underlying mechanism of sulforaphene (LFS) on osteoclast differentiation were illustrated in vitro and in vivo with RANKL-induced Raw264.7 cell line osteoclastogenesis and lipopolysaccharide (LPS)-induced bone erosion model. KEY RESULTS In this study, LFS has been shown to effectively impede the formation of mature osteoclasts induced from both Raw264.7 cell line and bone marrow macrophages (BMMs), mainly at the early stage. Further mechanistic investigations uncovered that LFS suppressed AKT phosphorylation. SC-79, a potent AKT activator, was found to reverse the inhibitory impact of LFS on osteoclast differentiation. Moreover, transcriptome sequencing analysis revealed that treatment with LFS led to a significant upregulation in the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and antioxidant-related genes. Then it's validated that LFS could promote NRF2 expression and nuclear translocation, as well as effectively resist oxidative stress. NRF2 knockdown reversed the suppression effect of LFS on osteoclast differentiation. In vivo experiments provide convincing evidence that LFS is protective against LPS-induced inflammatory osteolysis. CONCLUSION AND IMPLICATIONS These well-grounded and promising findings suggest LFS as a promising agent to addressing oxidative-stress related diseases and bone loss disorders.
Collapse
Affiliation(s)
- Hantao Yao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yangge Du
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bulin Jiang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yilin Liao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yaoyu Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Mengjie Yin
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ting Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yue Sheng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yaoting Ji
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Minquan Du
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
15
|
Bu T, Huang J, Yu Y, Sun P, Yang K. Whey Protein Hydrolysate Ameliorated High-Fat-Diet Induced Bone Loss via Suppressing Oxidative Stress and Regulating GSK-3β/Nrf2 Signaling Pathway. Nutrients 2023; 15:2863. [PMID: 37447191 DOI: 10.3390/nu15132863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/11/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Long-term hypercaloric intake such as a high-fat diet (HFD) could act as negative regulators on bone remodeling, thereby inducing bone loss and bone microarchitecture destruction. Currently, food-derived natural compounds represent a promising strategy to attenuate HFD-induced bone loss. We previously prepared a whey protein hydrolysate (WPH) with osteogenic capacity. In this study, we continuously isolated and identified an osteogenic and antioxidant octapeptide TPEVDDA from WPH, which significantly promoted the alkaline phosphatase activities on MC3T3-E1 cells and exerted DPPH radical scavenging capacity. We then established an HFD-fed obese mice model with significantly imbalanced redox status and reduced bone mass and further evaluated the effects of different doses of WPH on ameliorating the HFD-induced bone loss and oxidative damages. Results showed that the administration of 2% and 4% WPH for 12 weeks significantly restored perirenal fat mass, improved serum lipid levels, reduced oxidative stress, and promoted the activity of antioxidant enzymes; meanwhile, WPH significantly preserved bone mass and bone mechanical properties, attenuated the degradation of trabecular microstructure, and regulated serum bone metabolism biomarkers. The protein levels of Runx2, Nrf2, and HO-1, as well as the phosphorylation level of GSK-3β in tibias, were notably activated by WPH. Overall, we found that the potential mechanism of WPH on ameliorating the HFD-induced bone loss mainly through its antioxidant and osteogenic capacity by activating Runx2 and GSK-3β/Nrf2 signaling pathway, demonstrating the potential of WPH to be used as a nutritional strategy for obesity and osteoporosis.
Collapse
Affiliation(s)
- Tingting Bu
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Ju Huang
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yue Yu
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Peilong Sun
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Kai Yang
- Department of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
16
|
Zhang C, Li H, Li J, Hu J, Yang K, Tao L. Oxidative stress: A common pathological state in a high-risk population for osteoporosis. Biomed Pharmacother 2023; 163:114834. [PMID: 37163779 DOI: 10.1016/j.biopha.2023.114834] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/29/2023] [Accepted: 05/01/2023] [Indexed: 05/12/2023] Open
Abstract
Osteoporosis is becoming a major concern in the field of public health. The process of bone loss is insidious and does not directly induce obvious symptoms. Complications indicate an irreversible decrease in bone mass. The high-risk populations of osteoporosis, including postmenopausal women, elderly men, diabetic patients and obese individuals need regular bone mineral density testing and appropriate preventive treatment. However, the primary changes in these populations are different, increasing the difficulty of effective treatment of osteoporosis. Determining the core pathogenesis of osteoporosis helps improve the efficiency and efficacy of treatment among these populations. Oxidative stress is a common pathological state secondary to estrogen deficiency, aging, hyperglycemia and hyperlipemia. In this review, we divided oxidative stress into the direct effect of reactive oxygen species (ROS) and the reduction of antioxidant enzyme activity to discuss their roles in the development of osteoporosis. ROS initiated mitochondrial apoptotic signaling and suppressed osteogenic marker expression to weaken osteogenesis. MAPK and NF-κB signaling pathways mediated the positive effect of ROS on osteoclast differentiation. Antioxidant enzymes not only eliminate the negative effects of ROS, but also directly participate in the regulation of bone metabolism. Additionally, we also described the roles of proinflammatory factors and HIF-1α under the pathophysiological changes of inflammation and hypoxia, which provided a supplement of oxidative stress-induced osteoporosis. In conclusion, our review showed that oxidative stress was a common pathological state in a high-risk population for osteoporosis. Targeted oxidative stress treatment would greatly optimize the therapeutic schedule of various osteoporosis treatments.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China
| | - Hao Li
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China
| | - Jie Li
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China
| | - Jiajin Hu
- Health Sciences Institute, China Medical University, Shenyang 110122, China
| | - Keda Yang
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China.
| | - Lin Tao
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China.
| |
Collapse
|
17
|
Fragoulis A, Tohidnezhad M, Kubo Y, Wruck CJ, Craveiro RB, Bock A, Wolf M, Pufe T, Jahr H, Suhr F. The Contribution of the Nrf2/ARE System to Mechanotransduction in Musculoskeletal and Periodontal Tissues. Int J Mol Sci 2023; 24:ijms24097722. [PMID: 37175428 PMCID: PMC10177782 DOI: 10.3390/ijms24097722] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Mechanosensing plays an essential role in maintaining tissue functions. Across the human body, several tissues (i.e., striated muscles, bones, tendons, ligaments, as well as cartilage) require mechanical loading to exert their physiological functions. Contrary, mechanical unloading triggers pathological remodeling of these tissues and, consequently, human body dysfunctions. At the cellular level, both mechanical loading and unloading regulate a wide spectrum of cellular pathways. Among those, pathways regulated by oxidants such as reactive oxygen species (ROS) represent an essential node critically controlling tissue organization and function. Hence, a sensitive balance between the generation and elimination of oxidants keeps them within a physiological range. Here, the Nuclear Factor-E2-related factor 2/Antioxidant response element (Nrf2/ARE) system plays an essential role as it constitutes the major cellular regulation against exogenous and endogenous oxidative stresses. Dysregulations of this system advance, i.a., liver, neurodegenerative, and cancer diseases. Herein, we extend our comprehension of the Nrf2 system to the aforementioned mechanically sensitive tissues to explore its role in their physiology and pathology. We demonstrate the relevance of it for the tissues' functionality and highlight the imperative to further explore the Nrf2 system to understand the physiology and pathology of mechanically sensitive tissues in the context of redox biology.
Collapse
Affiliation(s)
- Athanassios Fragoulis
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Mersedeh Tohidnezhad
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Yusuke Kubo
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Christoph Jan Wruck
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Rogerio Bastos Craveiro
- Department of Orthodontics, Dental Clinic, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Anna Bock
- Department of Oral and Maxillofacial Surgery, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Michael Wolf
- Department of Orthodontics, Dental Clinic, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Holger Jahr
- Department of Anatomy and Cell Anatomy, Uniklinik RWTH Aachen, RWTH Aachen University, 52074 Aachen, Germany
- Institute of Structural Mechanics and Lightweight Design, RWTH Aachen University, 52062 Aachen, Germany
| | - Frank Suhr
- Division of Molecular Exercise Physiology, Faculty of Life Sciences: Food, Nutrition and Health, University of Bayreuth, 95326 Kulmbach, Germany
| |
Collapse
|
18
|
Exosomes from Adipose-Derived Stem Cells Alleviate Dexamethasone-Induced Bone Loss by Regulating the Nrf2/HO-1 Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:3602962. [PMID: 36778207 PMCID: PMC9908349 DOI: 10.1155/2023/3602962] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 02/04/2023]
Abstract
The widespread use of therapeutic glucocorticoids has increased the incidences of glucocorticoid-induced osteoporosis (GIOP). Oxidative stress and mitochondrial dysfunction are major causes of GIOP; therefore, alleviation of excess oxidative stress in osteoblasts is a potential therapeutic strategy for osteoporosis. Exosomes derived from ADSCs (ADSCs-Exos), as novel cell-free therapeutics, can modulate various biological processes, such as immunomodulation, reduce oxidative damage, and promote tissue repair as well as regeneration. In this study, ADSCs-Exos restored the viability and osteogenic potential of MC3T3-E1 cells by attenuating apoptosis, oxidative damage, intracellular ROS generation, and mitochondrial dysfunction. Moreover, after pretreatment with ADSCs-Exos, Nrf2 expressions were upregulated in Dex-stimulated osteoblasts. Inhibitory assays showed that silencing Nrf2 partially eliminated the protective effects of ADSCs-Exos. The rat model assays confirmed that ADSCs-Exos alleviated the Dex-induced increase in oxidation levels, restored bone mass of the distal femur, and increased the expressions of Nrf2 and osteogenic markers in bone tissues. Thus, ADSCs-Exos alleviated apoptosis and oxidative stress by regulating Nrf2/HO-1 expressions after Dex and prevented the development of GIOP in vivo.
Collapse
|
19
|
Pan Y, Lin H, Jiao H, Zhao J, Wang X. Effects of in ovo feeding of chlorogenic acid on antioxidant capacity of postnatal broilers. Front Physiol 2023; 14:1091520. [PMID: 36726849 PMCID: PMC9885134 DOI: 10.3389/fphys.2023.1091520] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/06/2023] [Indexed: 01/17/2023] Open
Abstract
In this study, chlorogenic acid (CGA) was injected into the amniotic cavity of chicken embryos to study the effects of in ovo feeding of CGA on the antioxidant capacity of postnatal broilers. On the 17th day of embryonic age, a total of 300 healthy broiler fertile eggs with similar weights were randomly subjected to five groups as follows; in ovo injection with 0.5 ml CGA at 4 mg/egg (4CGA) or 7 mg/egg (7CGA) or 10 mg/egg (10CGA), or sham-injection with saline (positive control, PC) or no injection (negative control, NC). Each group had six replicates of ten embryos. Six healthy chicks with similar body weights hatched from each replicate were selected and reared until heat stress treatment (35°C ± 1°C, 8 h/d) at 28-42 days of age. The results showed that there was no significant difference in the hatching rate between the groups (p > 0.05). After heat stress treatment, 4CGA group showed an improved intestinal morphology which was demonstrated by a higher villus height in the duodenum and a higher villus height/crypt depth ratio in the jejunum, compared with the NC group (p < 0.05). The antioxidant capacity of chickens was improved by in ovo feeding of CGA since 4CGA decreased the plasma content of malondialdehyde (MDA) (p < 0.05), whereas, it increased the superoxide dismutase (SOD), glutathione peroxidase (GPX), and catalase (CAT) activities compared with NC group (p < 0.05). Also, the MDA content of the different injection groups had a quadratic effect, with the 4CGA group having the lowest MDA content (P quadratic < 0.05). In the duodenum, 4CGA injection significantly increased the mRNA expressions of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase 1 (H O -1), glutathione synthetase (GSS), and SOD1 compared to the NC and PC groups (p < 0.05). The mRNA expressions of glutathione reductase (GSR) and GPX7 were significantly increased in all CGA-treated groups compared with the PC group (p < 0.05), while the mRNA expression of CAT was significantly increased by 4CGA group than the NC group (p < 0.05). The mRNA expressions of epigenetic-related genes, ten eleven translocation 1 and 2 (Tet1 and Tet2), and DNA-methyltransferase 3 alpha (DNMT3A) in the duodenum of 4CGA injected group was significantly increased compared with the NC and PC groups (p < 0.05). The mRNA expressions of Nrf2, SOD1, and Tet2 showed a significant quadratic effects with the 4CGA group having the highest expression (P quadratic < 0.05). In conclusion, in ovo feeding of CGA alleviated heat stress-induced intestinal oxidative damage. Injection with CGA of 4 mg/egg is considered most effective due to its actions in improving intestinal antioxidant capacity, especially in the duodenum. The antioxidant effects of in ovo CGA on postnatal heat-stressed broilers may be related to its regulation of epigenetic mechanisms. Thus, this study provides technical knowledge to support the in ovo feeding of CGA to alleviate oxidative stress in postnatal heat-stressed broilers.
Collapse
Affiliation(s)
- Yali Pan
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Tai’an, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tai’an, China
| | - Hai Lin
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Tai’an, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tai’an, China
| | - Hongchao Jiao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Tai’an, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tai’an, China
| | - Jingpeng Zhao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Tai’an, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tai’an, China
| | - Xiaojuan Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Technology, Shandong Agricultural University, Tai’an, China,Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Tai’an, China,*Correspondence: Xiaojuan Wang,
| |
Collapse
|
20
|
Qi X, Zhang X, Meng J, Wu J, Cheng W, Huang J, Lin W. Briarane-type diterpenoids, the inhibitors of osteoclast formation by interrupting Keap1-Nrf2 interaction and activating Nrf2 pathway. Eur J Med Chem 2023; 246:114948. [PMID: 36446206 DOI: 10.1016/j.ejmech.2022.114948] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 11/07/2022] [Accepted: 11/19/2022] [Indexed: 11/26/2022]
Abstract
Chemoinformatic and bioassay-guided fractionation of a gorgonian coral Junceella juncea resulted in the isolation of 45 briarane-type diterpenoids, of which 16 new analogues were characterized. Their structures were identified by extensive analyses of the spectroscopic data. Most isolated briaranes showed significant inhibition against the receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclast differentiation in bone marrow-derived macrophages cells (BMMs). Praelolide, one of the active analogues, significantly activates nuclear factor erythroid-2-related factor 2 (Nrf2) nucleus translocation, induces the expression of Nrf2-targeted genes, suppresses reactive oxygen species (ROS) production, abrogates the activation of downstream mitogen-activated protein kinase (MAPK)/nuclear factor-κB (NFκB) signaling, and subsequently attenuates osteoclast differentiation. Mechanically, praelolide interacts with Kelch-like ECH-associated protein 1 (Keap1) protein by non-covalent interaction to interrupt the interaction between Keap1 and Nrf2 and thereby to activate the Nrf2 signaling pathway. In addition, praelolide rescues the bone loss in prednisone-induced zebrafish. The present study provided praelolide as a new natural scaffold to remedy osteoclastogenic bone disease.
Collapse
Affiliation(s)
- Xinyi Qi
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, PR China
| | - Xu Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, PR China
| | - Junjun Meng
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, PR China
| | - Jingshuai Wu
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, PR China
| | - Wei Cheng
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, PR China
| | - Jian Huang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, PR China.
| | - Wenhan Lin
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, PR China; Institute of Ocean Research, Ningbo Institute of Marine Medicine, Peking University, Beijing, 100191, PR China.
| |
Collapse
|
21
|
Kubo Y, Gonzalez JAH, Beckmann R, Weiler M, Pahlavani H, Saldivar MC, Szymanski K, Rosenhain S, Fragoulis A, Leeflang S, Slowik A, Gremse F, Wolf M, Mirzaali MJ, Zadpoor AA, Wruck CJ, Pufe T, Tohidnezhad M, Jahr H. Nuclear factor erythroid 2-related factor 2 (Nrf2) deficiency causes age-dependent progression of female osteoporosis. BMC Musculoskelet Disord 2022; 23:1015. [PMID: 36434613 PMCID: PMC9700883 DOI: 10.1186/s12891-022-05942-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 11/02/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Nuclear factor erythroid 2-related factor 2 (Nrf2) is a crucial transcription factor for cellular redox homeostasis. The association of Nrf2 with elderly female osteoporotic has yet to be fully described. The aim was to elucidate a potential age-dependent Nrf2 contribution to female osteoporosis in mice. METHODS Eighteen female wild type (WT) and 16 Nrf2-knockout (KO) mice were sacrificed at different ages (12 weeks = young mature adult and 90 weeks = old) to analyze their femurs. The morphological properties (trabecular and cortical) were evaluated by micro-computed tomography (μCT) and compared to gold standard histochemistry analysis. The quasi-static compression tests were performed to calculate the mechanical properties of bones. Additionally, the population of bone resorbing cells and aromatase expression by osteocytes was immunohistochemically evaluated and empty osteocyte lacunae was counted in cortical bone. RESULTS Old Nrf2-KO mice revealed a significantly reduced trabecular bone mineral density (BMD), cortical thickness, cortical area, and bone fraction compared to old WT mice, regardless of no significant difference in skeletally mature young adult mice between WT and KO. Specifically, while all old WT mice showed thin metaphyseal trabeculae, trabecular bone was completely absent in 60% of old KO mice. Additionally, old KO mice showed significantly more osteoclast-like cells and fewer aromatase-positive osteocytes than WT mice, whereas the occurrence of empty osteocyte lacunae did not differ between both groups. Nrf2-KO mice further showed an age-dependently reduced fracture resilience compared to age-matched WT mice. CONCLUSION Our results suggest that chronic Nrf2 loss can lead to age-dependent progression of female osteoporosis.
Collapse
Affiliation(s)
- Yusuke Kubo
- grid.412301.50000 0000 8653 1507Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, 52074 Aachen, Germany
| | - Jesus Abraham Herrera Gonzalez
- grid.412301.50000 0000 8653 1507Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, 52074 Aachen, Germany
| | - Rainer Beckmann
- grid.412301.50000 0000 8653 1507Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, 52074 Aachen, Germany
| | - Marek Weiler
- grid.412301.50000 0000 8653 1507Institute for Experimental Molecular Imaging, Helmholtz Institute for Biomedical Engineering, Uniklinik RWTH Aachen, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Helda Pahlavani
- grid.5292.c0000 0001 2097 4740Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Mauricio Cruz Saldivar
- grid.5292.c0000 0001 2097 4740Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Katharina Szymanski
- grid.412301.50000 0000 8653 1507Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, 52074 Aachen, Germany
| | - Stefanie Rosenhain
- grid.412301.50000 0000 8653 1507Institute for Experimental Molecular Imaging, Helmholtz Institute for Biomedical Engineering, Uniklinik RWTH Aachen, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Athanassios Fragoulis
- grid.412301.50000 0000 8653 1507Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, 52074 Aachen, Germany
| | - Sander Leeflang
- grid.5292.c0000 0001 2097 4740Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Alexander Slowik
- grid.412301.50000 0000 8653 1507Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, 52074 Aachen, Germany
| | - Felix Gremse
- grid.412301.50000 0000 8653 1507Institute for Experimental Molecular Imaging, Helmholtz Institute for Biomedical Engineering, Uniklinik RWTH Aachen, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Michael Wolf
- grid.412301.50000 0000 8653 1507Department of Orthodontics, Uniklinik RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Mohammad Javad Mirzaali
- grid.5292.c0000 0001 2097 4740Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Amir Abbas Zadpoor
- grid.5292.c0000 0001 2097 4740Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Mekelweg 2, 2628 CD Delft, The Netherlands
| | - Christoph Jan Wruck
- grid.412301.50000 0000 8653 1507Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, 52074 Aachen, Germany
| | - Thomas Pufe
- grid.412301.50000 0000 8653 1507Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, 52074 Aachen, Germany
| | - Mersedeh Tohidnezhad
- grid.412301.50000 0000 8653 1507Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, 52074 Aachen, Germany
| | - Holger Jahr
- grid.412301.50000 0000 8653 1507Department of Anatomy and Cell Biology, Uniklinik RWTH Aachen, Wendlingweg 2, 52074 Aachen, Germany ,grid.1957.a0000 0001 0728 696XInstitute of Structural Mechanics and Lightweight Design, RWTH Aachen University, Wüllnerstraße 7, 52062 Aachen, Germany
| |
Collapse
|
22
|
Phytol Suppresses Osteoclast Differentiation and Oxidative Stress through Nrf2/HO-1 Regulation in RANKL-Induced RAW264.7 Cells. Cells 2022; 11:cells11223596. [PMID: 36429027 PMCID: PMC9688212 DOI: 10.3390/cells11223596] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Osteoporosis is a systemic skeletal disorder where osteoclasts are prevalent among osteoblasts. Oxidative stress is one of the main causes of osteoporosis, and nuclear factor erythroid-2-related factor 2 (Nrf2) is the master regulator of antioxidant responses. Phytol, a diterpene isolated from Stevia rebaudiana leaves, has many biological effects, including antimicrobial, antioxidant, and anti-inflammatory effects. This study investigated the crosstalk between Nrf2 and osteoclast differentiation in the presence of phytol. Phytol inhibited osteoclast differentiation through TRAP-positive and F-actin formation. The expression of anti-nuclear factor of activated T cells-c1 (NFATc1) and c-Fos was suppressed by phytol, as shown using Western blot and RT-PCR analysis. Phytol inhibited oxidative stress by suppressing reactive oxidant species (ROS) accumulation while recovering antioxidant enzymes, including superoxide dismutase and catalase. Additionally, phytol ameliorated osteoclast-specific differentiation, function, and oxidative stress through Nrf2 regulation by siRNA transfection. In conclusion, these data demonstrate the inhibitory effect of phytol on osteoclast differentiation through Nrf2 regulation, suggesting its potential use in oxidative stress-related osteoporosis and bone diseases.
Collapse
|
23
|
Ginsenoside Rg1 Reduces Oxidative Stress Via Nrf2 Activation to Regulate Age-Related Mesenchymal Stem Cells Fate Switch Between Osteoblasts and Adipocytes. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1411354. [PMID: 36267094 PMCID: PMC9578818 DOI: 10.1155/2022/1411354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/05/2022] [Accepted: 09/19/2022] [Indexed: 11/17/2022]
Abstract
Background An important feature of aging cells is the gradual loss of physiological integrity. As aging progresses, MSCs change preferring to differentiate toward adipocytes rather than osteoblasts. Oxidative stress accumulation is an important factor in age-related bone loss. Many experiments have demonstrated the good therapeutic effect of Ginsenoside (Rg1) on oxidative stress injury. In this study, we investigated the effect of Rg1 on the osteogenic-adipogenic differentiation balance of bone marrow mesenchymal stem cells (BMMSC). Objective To analyze the potential application value of Rg1 in the treatment of senile osteoporosis. Methods BMMSCs were isolated from healthy donors of different ages and identified based on isotype and by multi-differentiation induction. Rg1 was used to treat BMMSCs, The differentiation propensity was analyzed by induction of differentiation assay. Antioxidant capacity of BMMSCs as measured by oxidative stress product assay Related mechanism studies were confirmed by quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR), immunofluorescence, western blotting, and inhibitor treatment. Moreover, Observation of the effects of Rg1 on aging BMMSCs under in vivo conditions by treatment of aged mice with Rg1 injections. Results Rg1 treatment rescued age-induced switch of BMMSCs differentiation fate in vitro. In elderly people, Rg1 markedly increased osteogenic differentiation of BMMSCs by decreasing oxidative stress, while inhibiting adipogenic differentiation. However, this effect was abolished in BMMSCs by an Nrf2-inhibitor. Notably, aging mice showed a reduction in adipocyte distribution in the bone marrow and a decrease in oxidative stress products after a 3-month period of Rg1 treatment. Conclusion We have uncovered a novel function for Rg1 that involves attenuating bone loss via Nrf2 antioxidant signaling, which in turn may potentially be utilized as a therapeutic agent for improving osteogenic differentiation in aging BMMSCs.
Collapse
|
24
|
Abu-Kheit R, Kotev-Emeth S, Hiram-Bab S, Gabet Y, Savion N. S-allylmercapto- N-acetylcysteine protects bone cells from oxidation and improves femur microarchitecture in healthy and diabetic mice. Exp Biol Med (Maywood) 2022; 247:1489-1500. [PMID: 35658550 PMCID: PMC9493761 DOI: 10.1177/15353702221095047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Oxidative stress is involved in the deterioration of bone quality and mechanical strength in both diabetic and aging adults. Therefore, we studied the ability of the antioxidant compound, S-allylmercapto-N-acetylcysteine (ASSNAC) to protect bone marrow stromal cells (BMSCs) from advanced glycation end-products (AGEs) cytotoxicity and improve bone microarchitecture of adult healthy and obese/diabetic (db/db) female mice. ASSNAC effect on AGEs-treated cultured rat BMSCs was evaluated by Neutral Red and XTT cell survival and reactive oxygen species (ROS) level assays. Its effect on healthy (C57BL/6) and obese/diabetic (C57BLKS/J Leprdb+/+; db/db) female mice femur parameters, such as (1) number of adherent BMSCs, (2) percentage of CD73+/CD45- cells in bone marrow (BM), (3) glutathione level in BM cells, and (4) femur microarchitecture parameters by microcomputed tomography, was studied. ASSNAC treatment protected BMSCs by significantly decreasing AGEs-induced ROS production and increasing their cellular resistance to the cytotoxic effect of AGEs. ASSNAC treatment of healthy female mice (50 mg/kg/day; i.p.; age 12-20 weeks) significantly increased the number of BMSCs (+60%), CD73+/CD45- cells (+134%), and glutathione level (+110%) in the femur bone marrow. Furthermore, it increased the femur length (+3%), cortical diameter (+3%), and cortical areal moment of inertia (Ct.MOI; +10%) a surrogate for biomechanical strength. In db/db mice that demonstrated a compromised trabecular bone and growth plate microarchitecture, ASSNAC treatment restored the trabecular number (Tb.N, +29%), bone volume fraction (Tb.BV/TV, +130%), and growth plate primary spongiosa volumetric bone mineral density (PS-vBMD, +7%) and thickness (PS-Th, +18%). In conclusion, this study demonstrates that ASSNAC protects bone marrow cells from oxidative stress and may improve bone microarchitecture in adult healthy and diabetic female mice.
Collapse
Affiliation(s)
- Reem Abu-Kheit
- Department of Human Molecular Genetics and
Biochemistry and Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv
University, Tel Aviv 6997801, Israel
| | - Shlomo Kotev-Emeth
- Department of Human Molecular Genetics and
Biochemistry and Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv
University, Tel Aviv 6997801, Israel
| | - Sahar Hiram-Bab
- Department of Anatomy and Anthropology,
Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Yankel Gabet
- Department of Anatomy and Anthropology,
Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Naphtali Savion
- Department of Human Molecular Genetics and
Biochemistry and Goldschleger Eye Research Institute, Sackler Faculty of Medicine, Tel Aviv
University, Tel Aviv 6997801, Israel;,Naphtali Savion.
| |
Collapse
|
25
|
Liu Q, Liu Z, Wang C, Gao X, Li C, Wang M, Wang Q, Cai JP. Increased production of 8-oxo-7,8-dihydroguanine in human urine, a novel biomarker of osteoporosis. Free Radic Res 2022; 56:358-365. [PMID: 35880390 DOI: 10.1080/10715762.2022.2106224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Osteoporosis is a worldwide disease that seriously affects the quality of life and survival rate of the elderly. The detection of bone biomarkers will provide supplementary information of bone mineral density, contributing to the accurate diagnosis of osteoporosis and better health care for prevention. This study aimed to investigate the efficacy of oxidative stress markers-8-oxo-7,8-dihydroguanine (8-oxoGsn) and 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodGsn) in the assessment of osteoporosis. We conducted a cross-sectional study among menopausal women with a mean (standard deviation) age of 62.967 (7.798) years old (n = 151). Participants were recruited for the bone mineral density (BMD) assessment, blood and urinary samples. Urinary 8-oxo-7,8-dihydro-2'-deoxyguanosine and 8-oxo-7,8-dihydro-guanine concentrations were measured by ultra performance liquid chromatography and tandem mass spectrometry (UPLC-MS/MS). The urinary 8-oxoGsn/Cre value differed significantly between normal and osteoporotic participants (p < 0.001), while the 8-oxodGsn/Cre value did not (p = 0.720). Even after adjusting for the age and body mass index, the BMD was still associated with urinary 8-oxoGsn/Cre value. ROC analysis showed that 8-oxoGsn has a strong diagnostic value for osteoporosis (AUC =0.744). The results show for the first time that 8-oxoGsn may be a biomarker for the future diagnosis of osteoporosis in women.
Collapse
Affiliation(s)
- Qian Liu
- Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing, P.R. China
| | - Zhen Liu
- Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing, P.R. China
| | - Chenchen Wang
- Department of Pharmacy, Quzhou KeCheng People's Hospital, Quzhou, Zhejiang, China
| | - Xin Gao
- Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing, P.R. China
| | - Chuanbao Li
- Department of Laboratory Medicine, Beijing Hospital, National Center of Gerontology, Beijing, P.R. China
| | - Meng Wang
- Department of Laboratory Medicine, Beijing Hospital, National Center of Gerontology, Beijing, P.R. China
| | - Qiang Wang
- Department of Orthopedics, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jian Ping Cai
- Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing, P.R. China
| |
Collapse
|
26
|
Anti-Osteoporotic Effect of Viscozyme-Assisted Polysaccharide Extracts from Portulaca oleracea L. on H2O2-Treated MC3T3-E1 Cells and Zebrafish. SEPARATIONS 2022. [DOI: 10.3390/separations9050128] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
This study aims to screen and characterize the protective effect of polysaccharides from Portulaca oleracea L. (POP) against H2O2-stimulated osteoblast apoptosis in vivo and in vitro. The enzymes viscozyme, celluclast, α-amylase, and β-glucanase were used to extract POPs. Among all enzyme-assisted POPs, the first participating fraction of viscozyme extract POP (VPOP1) exhibited the highest antioxidant activity. Hoechst 33342 and acridine orange/ethidium bromide staining and flow cytometry of MC3T3 cells revealed that VPOP1 inhibited apoptosis in a dose-dependent manner. Moreover, VPOP1 increased the expression levels of heme oxygenase-1 (HO-1) and NADPH quinine oxidoreductase 1 (NQO1) and decreased the expression levels of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and Kelch-like ECH-associated protein 1 (Keap1) in H2O2-induced cells compared with their controls. The results of an in vivo experiment show that VPOP1 significantly reduced reactive oxygen species generation and lipid peroxidation in zebrafish at 72 h post-fertilization and promoted bone growth at 9 days post-fertilization. Furthermore, VPOP1 was identified via 1-phenyl-3-methyl-5-pyrazolone derivatization as an acidic heteropolysaccharide comprising mannose and possessing a molecular weight of approximately 7.6 kDa. Collectively, VPOP1 was selected as a potential anti-osteoporotic functional food because of its protective activity against H2O2-induced damage in vitro and in vivo.
Collapse
|
27
|
Wang YF, Chang YY, Zhang XM, Gao MT, Zhang QL, Li X, Zhang L, Yao WF. Salidroside protects against osteoporosis in ovariectomized rats by inhibiting oxidative stress and promoting osteogenesis via Nrf2 activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:154020. [PMID: 35278902 DOI: 10.1016/j.phymed.2022.154020] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/26/2022] [Accepted: 02/25/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Osteoporosis (OP) is characterized as low bone mass, bone microarchitecture breakdown and bone fragility. The increase of oxidative stress could lead to breakdown in the balance of bone formation and resorption which gives rise to OP. Nrf2 is a transcription factor which takes part in oxidative stress and recently was reported that it can regulate the occurrence of OP. Salidroside (SAL) with the efficacies of anti-oxidation, anti-aging and bone-protection is one of the active ingredients in Ligustri Lucidi Fructus, a traditional Chinese medicinal herb. Nevertheless, few studies have explored the potential mechanism of SAL preventing OP development from the perspective of oxidative stress intervention. PURPOSE This study aimed to investigate the pharmacological effect and molecular mechanisms of SAL on OP. STUDY DESIGNS AND METHODS A tert-butyl hydroperoxide (t-BHP)-induced oxidative stress model was applied for investigating the effects of SAL in vitro, and an ovariectomized (OVX) model was used for in vivo study on the effect of SAL for OP. Related pharmacodynamic actions and molecular mechanisms of SAL were explored in both rat osteoblasts (ROBs) and OVX rats. Network biology and cell metabolomics were performed for further investigating the correlation and association among potential biomarkers, targets and pathways. RESULTS SAL reduced levels of ROS and lipid peroxidation (LPO), increased activities of antioxidant enzymes like GPx and SOD, and enhanced osteogenic differentiation in t-BHP-induced ROBs and OVX rats. Mechanistic studies showed SAL prevented OP development and reduced oxidative damage in ROBs and OVX rats through up-regulating Nrf2 expression and facilitating its nuclear translocation. The joint analysis of network biology and cell metabolomics revealed that galactose metabolism and fatty acid metabolism could be the major influenced pathways following treatment with SAL. CONCLUSION SAL could protect against OP by inhibiting oxidative stress, promoting osteogenesis through the up-regulation of Nrf2 and intervening galactose metabolism and fatty acid metabolism. Our study implied that SAL may be a potential drug to treat OP.
Collapse
Affiliation(s)
- Yi-Fei Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Yue-Yue Chang
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Xue-Meng Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Meng-Ting Gao
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Qiu-Lan Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Xin Li
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Li Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wei-Feng Yao
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
28
|
Sheppard AJ, Barfield AM, Barton S, Dong Y. Understanding Reactive Oxygen Species in Bone Regeneration: A Glance at Potential Therapeutics and Bioengineering Applications. Front Bioeng Biotechnol 2022; 10:836764. [PMID: 35198545 PMCID: PMC8859442 DOI: 10.3389/fbioe.2022.836764] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/19/2022] [Indexed: 01/24/2023] Open
Abstract
Although the complex mechanism by which skeletal tissue heals has been well described, the role of reactive oxygen species (ROS) in skeletal tissue regeneration is less understood. It has been widely recognized that a high level of ROS is cytotoxic and inhibits normal cellular processes. However, with more recent discoveries, it is evident that ROS also play an important, positive role in skeletal tissue repair, specifically fracture healing. Thus, dampening ROS levels can potentially inhibit normal healing. On the same note, pathologically high levels of ROS cause a sharp decline in osteogenesis and promote nonunion in fracture repair. This delicate balance complicates the efforts of therapeutic and engineering approaches that aim to modulate ROS for improved tissue healing. The physiologic role of ROS is dependent on a multitude of factors, and it is important for future efforts to consider these complexities. This review first discusses how ROS influences vital signaling pathways involved in the fracture healing response, including how they affect angiogenesis and osteogenic differentiation. The latter half glances at the current approaches to control ROS for improved skeletal tissue healing, including medicinal approaches, cellular engineering, and enhanced tissue scaffolds. This review aims to provide a nuanced view of the effects of ROS on bone fracture healing which will inspire novel techniques to optimize the redox environment for skeletal tissue regeneration.
Collapse
Affiliation(s)
- Aaron J. Sheppard
- Department of Orthopaedic Surgery, Louisiana State University Health Shreveport, Shreveport, LA, United States
- School of Medicine, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Ann Marie Barfield
- Department of Orthopaedic Surgery, Louisiana State University Health Shreveport, Shreveport, LA, United States
- School of Medicine, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Shane Barton
- Department of Orthopaedic Surgery, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Yufeng Dong
- Department of Orthopaedic Surgery, Louisiana State University Health Shreveport, Shreveport, LA, United States
| |
Collapse
|
29
|
Xi X, Li Z, Liu H, Chen S, Liu D. Nrf2 Activation Is Involved in Cyclic Mechanical Stress-Stimulated Osteogenic Differentiation in Periodontal Ligament Stem Cells via PI3K/Akt Signaling and HO1-SOD2 Interaction. Front Cell Dev Biol 2022; 9:816000. [PMID: 35071244 PMCID: PMC8770743 DOI: 10.3389/fcell.2021.816000] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/15/2021] [Indexed: 12/22/2022] Open
Abstract
Nuclear factor erythroid-2-related factor-2 (Nrf2), the major transcriptional regulator in antioxidant response and cellular defense, had the vital effect on regulating osteogenic differentiation. Our previous study revealed that Nrf2 activation was involved in cyclic mechanical stress-stimulated osteogenic differentiation in the human periodontal ligament stem cells (PDLSCs). However, the mechanisms of Nrf2 underlying this process remained unclear. The goal of the study was to explore the mechanisms of Nrf2 in PDLSCs during cyclic mechanical stress-stimulated osteogenic differentiation via the tandem mass tag (TMT)-based liquid chromatography tandem-mass spectrometry (LC-MS/MS) analysis. And we applied tert-Butylhydroquinone (t-BHQ), the Nrf2 activator, to the orthodontic rats and detected the expression levels of the osteogenesis markers by immunohistochemistry (IHC) staining. Our results showed that Nrf2 activation in PDLSCs was involved in cyclic mechanical stress-stimulated osteogenic differentiation via phosphoinositide 3 kinase (PI3K)/protein kinase B (Akt) pathway. The protein-protein interaction between Akt and Nrf2 was detected. And the protein-protein interaction between heme oxygenase 1 (HO1) and superoxide dismutase 2 (SOD2), the downstream antioxidants of Nrf2, was associated with cyclic mechanical stress-stimulated osteogenic differentiation. T-BHQ enhanced the expression levels of the osteogenesis markers in orthodontic rats. Nrf2 might possess the potential to be a feasible molecular target in orthodontics.
Collapse
Affiliation(s)
- Xun Xi
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Zixuan Li
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Hong Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Shuai Chen
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Dongxu Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| |
Collapse
|
30
|
Han J, Yang K, An J, Jiang N, Fu S, Tang X. The Role of NRF2 in Bone Metabolism - Friend or Foe? Front Endocrinol (Lausanne) 2022; 13:813057. [PMID: 35282459 PMCID: PMC8906930 DOI: 10.3389/fendo.2022.813057] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/05/2022] [Indexed: 12/14/2022] Open
Abstract
Bone metabolism is closely related to oxidative stress. As one of the core regulatory factors of oxidative stress, NRF2 itself and its regulation of oxidative stress are both involved in bone metabolism. NRF2 plays an important and controversial role in the regulation of bone homeostasis in osteoblasts, osteoclasts and other bone cells. The role of NRF2 in bone is complex and affected by several factors, such as its expression levels, age, sex, the presence of various physiological and pathological conditions, as well as its interaction with certains transcription factors that maintain the normal physiological function of the bone tissue. The properties of NRF2 agonists have protective effects on the survival of osteogenic cells, including osteoblasts, osteocytes and stem cells. Activation of NRF2 directly inhibits osteoclast differentiation by resisting oxidative stress. The effects of NRF2 inhibition and hyperactivation on animal skeleton are still controversial, the majority of the studies suggest that the presence of NRF2 is indispensable for the acquisition and maintenance of bone mass, as well as the protection of bone mass under various stress conditions. More studies show that hyperactivation of NRF2 may cause damage to bone formation, while moderate activation of NRF2 promotes increased bone mass. In addition, the effects of NRF2 on the bone phenotype are characterized by sexual dimorphism. The efficacy of NRF2-activated drugs for bone protection and maintenance has been verified in a large number of in vivo and in vitro studies. Additional research on the role of NRF2 in bone metabolism will provide novel targets for the etiology and treatment of osteoporosis.
Collapse
Affiliation(s)
- Jie Han
- The First Clinical College of Lanzhou University, Lanzhou, China
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Kuan Yang
- The First Clinical College of Lanzhou University, Lanzhou, China
| | - Jinyang An
- The First Clinical College of Lanzhou University, Lanzhou, China
| | - Na Jiang
- The First Clinical College of Lanzhou University, Lanzhou, China
| | - Songbo Fu
- The First Clinical College of Lanzhou University, Lanzhou, China
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xulei Tang
- The First Clinical College of Lanzhou University, Lanzhou, China
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Xulei Tang,
| |
Collapse
|
31
|
Dexamethasone Administration in Mice Leads to Less Body Weight Gain over Time, Lower Serum Glucose, and Higher Insulin Levels Independently of NRF2. Antioxidants (Basel) 2021; 11:antiox11010004. [PMID: 35052508 PMCID: PMC8773000 DOI: 10.3390/antiox11010004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/17/2021] [Accepted: 12/19/2021] [Indexed: 12/17/2022] Open
Abstract
Glucocorticoids are used widely on a long-term basis in autoimmune and inflammatory diseases. Their adverse effects include the development of hyperglycemia and osteoporosis, whose molecular mechanisms have been only partially studied in preclinical models. Both these glucocorticoid-induced pathologies have been shown to be mediated at least in part by oxidative stress. The transcription factor nuclear erythroid factor 2-like 2 (NRF2) is a central regulator of antioxidant and cytoprotective responses. Thus, we hypothesized that NRF2 may play a role in glucocorticoid-induced metabolic disease and osteoporosis. To this end, WT and Nrf2 knockout (Nrf2KO) mice of both genders were treated with 2 mg/kg dexamethasone or vehicle 3 times per week for 13 weeks. Dexamethasone treatment led to less weight gain during the treatment period without affecting food consumption, as well as to lower glucose levels and high insulin levels compared to vehicle-treated mice. Dexamethasone also reduced cortical bone volume and density. All these effects of dexamethasone were similar between male and female mice, as well as between WT and Nrf2KO mice. Hepatic NRF2 signaling and gluconeogenic gene expression were not affected by dexamethasone. A 2-day dexamethasone treatment was also sufficient to increase insulin levels without affecting body weight and glucose levels. Hence, dexamethasone induces hyperinsulinemia, which potentially leads to decreased glucose levels, as well as osteoporosis, both independently of NRF2.
Collapse
|
32
|
Zhou X, Yuan W, Xiong X, Zhang Z, Liu J, Zheng Y, Wang J, Liu J. HO-1 in Bone Biology: Potential Therapeutic Strategies for Osteoporosis. Front Cell Dev Biol 2021; 9:791585. [PMID: 34917622 PMCID: PMC8669958 DOI: 10.3389/fcell.2021.791585] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/12/2021] [Indexed: 02/05/2023] Open
Abstract
Osteoporosis is a prevalent bone disorder characterized by bone mass reduction and deterioration of bone microarchitecture leading to bone fragility and fracture risk. In recent decades, knowledge regarding the etiological mechanisms emphasizes that inflammation, oxidative stress and senescence of bone cells contribute to the development of osteoporosis. Studies have demonstrated that heme oxygenase 1 (HO-1), an inducible enzyme catalyzing heme degradation, exhibits anti-inflammatory, anti-oxidative stress and anti-apoptosis properties. Emerging evidence has revealed that HO-1 is critical in the maintenance of bone homeostasis, making HO-1 a potential target for osteoporosis treatment. In this Review, we aim to provide an introduction to current knowledge of HO-1 biology and its regulation, focusing specifically on its roles in bone homeostasis and osteoporosis. We also examine the potential of HO-1-based pharmacological therapeutics for osteoporosis and issues faced during clinical translation.
Collapse
Affiliation(s)
- Xueman Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Wenxiu Yuan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Xiong
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhenzhen Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaqi Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yingcheng Zheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jin Liu
- Lab for Aging Research, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
33
|
Ibrahim N'I, Mohd Noor H'I, Shuid AN, Mohamad S, Abdul Malik MM, Jayusman PA, Shuid AN, Naina Mohamed I. Osteoprotective Effects in Postmenopausal Osteoporosis Rat Model: Oral Tocotrienol vs. Intraosseous Injection of Tocotrienol-Poly Lactic-Co-Glycolic Acid Combination. Front Pharmacol 2021; 12:706747. [PMID: 34867320 PMCID: PMC8637158 DOI: 10.3389/fphar.2021.706747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 10/19/2021] [Indexed: 11/13/2022] Open
Abstract
Osteoporosis, the most common bone disease, is associated with compromised bone strength and increased risk of fracture. Previous studies have shown that oxidative stress contributes to the progression of osteoporosis. Specifically, for postmenopausal osteoporosis, the reduction in estrogen levels leads to increased oxidative stress in bone remodeling. Tocotrienol, a member of vitamin E that exhibits antioxidant activities, has shown potential as an agent for the treatment of osteoporosis. Most studies on the osteoprotective effects of tocotrienols had used the oral form of tocotrienols, despite their low bioavailability due the lack of transfer proteins and high metabolism in the liver. Several bone studies have utilized tocotrienol combined with a nanocarrier to produce a controlled release of tocotrienol particles into the system. However, the potential of delivering tocotrienol-nanocarrier combination through the intraosseous route has never been explored. In this study, tocotrienol was combined with a nanocarrier, poly lactic-co-glycolic acid (PLGA), and injected intraosseously into the bones of ovariectomized rats to produce targeted and controlled delivery of tocotrienol into the bone microenvironment. This new form of tocotrienol delivery was compared with the conventional oral delivery in terms of their effects on bone parameters. Forty Sprague-Dawley rats were divided into five groups. The first group was sham operated, while other groups were ovariectomized (OVX). Following 2 months, the right tibiae of all the rats were drilled at the metaphysis region to provide access for intraosseous injection. The estrogen group (OVX + ESTO) and tocotrienol group (OVX + TTO) were given daily oral gavages of Premarin (64.5 mg/kg) and annatto-tocotrienol (60 mg/kg), respectively. The locally administered tocotrienol group (OVX + TTL) was given a single intraosseous injection of tocotrienol-PLGA combination. After 8 weeks of treatment, both OVX + TTO and OVX + TTL groups have significantly lower bone markers and higher bone mineral content than the OVX group. In terms of bone microarchitecture, both groups demonstrated significantly higher trabecular separation and connectivity density than the OVX group (p < 0.05). Both groups also showed improvement in bone strength by the significantly higher stress, strain, stiffness, and Young's modulus parameters. In conclusion, daily oral tocotrienol and one-time intraosseous injection of tocotrienol-PLGA combination were equally effective in offering protection against ovariectomy-induced bone changes.
Collapse
Affiliation(s)
- Nurul 'Izzah Ibrahim
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Hasnul 'Iffah Mohd Noor
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Ahmad Naqib Shuid
- Advanced Medical & Dental Institute (AMDI), Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Sharlina Mohamad
- Advanced Medical & Dental Institute (AMDI), Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Mohd Maaruf Abdul Malik
- Centre of Preclinical Science Studies, Faculty of Dentistry, Universiti Teknologi MARA, Sungai Buloh Campus, Jalan Hospital, Sungai Buloh, Malaysia
| | - Putri Ayu Jayusman
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Ahmad Nazrun Shuid
- Department of Pharmacology, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Jalan Hospital, Sungai Buloh, Malaysia
| | - Isa Naina Mohamed
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
34
|
Zhang Y, Liu X, Li Y, Song M, Li Y, Yang A, Zhang Y, Wang D, Hu M. Aucubin slows the development of osteoporosis by inhibiting osteoclast differentiation via the nuclear factor erythroid 2-related factor 2-mediated antioxidation pathway. PHARMACEUTICAL BIOLOGY 2021; 59:1556-1565. [PMID: 34757891 PMCID: PMC8583775 DOI: 10.1080/13880209.2021.1996614] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
CONTEXT Osteoporosis (OP) is a metabolic disease. We have previously demonstrated that aucubin (AU) has anti-OP effects that are due to its promotion of the formation of osteoblasts. OBJECTIVES To investigate the mechanisms of anti-OP effects of AU. MATERIALS AND METHODS C57BL/6 mice were randomly divided into control group, 30 mg/kg Dex-induced OP group (OP model group, 15 μg/kg oestradiol-treated positive control group, 5 or 45 mg/kg AU-treated group), and 45 mg/kg AU-alone-treated group. The administration lasted for 7 weeks. Subsequently, 1, 2.5 and 5 µM AU were incubated with 50 ng/mL RANKL-induced RAW264.7 cells for 7 days to observe osteoclast differentiation. The effect of AU was evaluated by analysing tissue lesions, biochemical factor and protein expression. RESULTS The LD50 of AU was greater than 45 mg/kg. AU increased the number of trabeculae and reduced the loss of chondrocytes in OP mice. Compared to OP mice, AU-treated mice exhibited decreased serum concentrations of TRAP5b (19.6% to 28.4%), IL-1 (12.2% to 12.6%), IL-6 (12.1%) and ROS (5.9% to 10.7%) and increased serum concentrations of SOD (14.6% to 19.4%) and CAT (17.2% to 27.4%). AU treatment of RANKL-exposed RAW264.7 cells decreased the numbers of multi-nuclear TRAP-positive cells, reversed the over-expression of TRAP5, NFATc1 and CTSK. Furthermore, AU increased the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream proteins in RANKL-exposed RAW264.7 cells. CONCLUSIONS AU slows the development of OP via Nrf2-mediated antioxidant pathways, indicating the potential use of AU in OP therapy and other types of OP research.
Collapse
Affiliation(s)
- Yongfeng Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun, China
- School of Life Sciences, Jilin University, Changchun, China
| | - Xin Liu
- School of Life Sciences, Jilin University, Changchun, China
| | - Yangyang Li
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Minkai Song
- School of Life Sciences, Jilin University, Changchun, China
| | - Yutong Li
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Anhui Yang
- School of Life Sciences, Jilin University, Changchun, China
| | - Yaqin Zhang
- School of Life Sciences, Jilin University, Changchun, China
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun, China
- CONTACT Di Wang School of Life Sciences, Jilin University, Qianjin Street 2699, Changchun, Jilin, P. R. China
| | - Min Hu
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun, China
- Min Hu Department of Orthodontics, College of Stomatology, Jilin University, No. 1500, Qinghua Road, Changchun, Jilin, P. R. China
| |
Collapse
|
35
|
Priddy C, Li J. The role of the Nrf2/Keap1 signaling cascade in mechanobiology and bone health. Bone Rep 2021; 15:101149. [PMID: 34869801 PMCID: PMC8626578 DOI: 10.1016/j.bonr.2021.101149] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/06/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
In conjunction with advancements in modern medicine, bone health is becoming an increasingly prevalent concern among a global population with an ever-growing life expectancy. Countless factors contribute to declining bone strength, and age exacerbates nearly all of them. The detrimental effects of bone loss have a profound impact on quality of life. As such, there is a great need for full exploration of potential therapeutic targets that may provide antiaging benefits and increase the life and strength of bone tissues. The Keap1-Nrf2 pathway is a promising avenue of this research. The cytoprotective and antioxidant functions of this pathway have been shown to mitigate the deleterious effects of oxidative stress on bone tissues, but the exact cellular and molecular mechanisms by which this occurs are not yet fully understood. Presently, refined animal and loading models are allowing exploration into the effect of the Keap1-Nrf2 pathway in a tissue-specific or even cell-specific manner. In addition, Nrf2 activators currently undergoing clinical trials can be utilized to investigate the particular cellular mechanisms at work in this cytoprotective cascade. Although the timing and dosing of treatment with Nrf2 activators need to be further investigated, these activators have great potential to be used clinically to prevent and treat osteoporosis.
Collapse
Affiliation(s)
- Carlie Priddy
- Department of Biology, Indiana University – Purdue University Indianapolis, Indianapolis, IN, USA
| | - Jiliang Li
- Department of Biology, Indiana University – Purdue University Indianapolis, Indianapolis, IN, USA
| |
Collapse
|
36
|
Bhattarai G, So HS, Kieu TTT, Kook SH, Lee JC, Jeon YM. Astaxanthin Inhibits Diabetes-Triggered Periodontal Destruction, Ameliorates Oxidative Complications in STZ-Injected Mice, and Recovers Nrf2-Dependent Antioxidant System. Nutrients 2021; 13:3575. [PMID: 34684576 PMCID: PMC8537008 DOI: 10.3390/nu13103575] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 09/29/2021] [Accepted: 10/07/2021] [Indexed: 12/12/2022] Open
Abstract
Numerous studies highlight that astaxanthin (ASTX) ameliorates hyperglycemic condition and hyperglycemia-associated chronic complications. While periodontitis and periodontic tissue degradation are also triggered under chronic hyperglycemia, the roles of ASTX on diabetes-associated periodontal destruction and the related mechanisms therein are not yet fully understood. Here, we explored the impacts of supplemental ASTX on periodontal destruction and systemic complications in type I diabetic mice. To induce diabetes, C57BL/6 mice received a single intraperitoneal injection of streptozotocin (STZ; 150 mg/kg), and the hyperglycemic mice were orally administered with ASTX (12.5 mg/kg) (STZ+ASTX group) or vehicle only (STZ group) daily for 60 days. Supplemental ASTX did not improve hyperglycemic condition, but ameliorated excessive water and feed consumptions and lethality in STZ-induced diabetic mice. Compared with the non-diabetic and STZ+ASTX groups, the STZ group exhibited severe periodontal destruction. Oral gavage with ASTX inhibited osteoclastic formation and the expression of receptor activator of nuclear factor (NF)-κB ligand, 8-OHdG, γ-H2AX, cyclooxygenase 2, and interleukin-1β in the periodontium of STZ-injected mice. Supplemental ASTX not only increased the levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and osteogenic transcription factors in the periodontium, but also recovered circulating lymphocytes and endogenous antioxidant enzyme activity in the blood of STZ-injected mice. Furthermore, the addition of ASTX blocked advanced glycation end products-induced oxidative stress and growth inhibition in human-derived periodontal ligament cells by upregulating the Nrf2 pathway. Together, our results suggest that ASTX does not directly improve hyperglycemia, but ameliorates hyperglycemia-triggered periodontal destruction and oxidative systemic complications in type I diabetes.
Collapse
Affiliation(s)
- Govinda Bhattarai
- Cluster for Craniofacial Development and Regeneration Research, School of Dentistry, Jeonbuk National University, Jeonju 54896, Korea;
- Institute of Oral Biosciences, School of Dentistry, Jeonbuk National University, Jeonju 54896, Korea
| | - Han-Sol So
- Research Center of Bioactive Materials, Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju 54896, Korea; (H.-S.S.); (T.T.T.K.)
| | - Thi Thu Trang Kieu
- Research Center of Bioactive Materials, Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju 54896, Korea; (H.-S.S.); (T.T.T.K.)
| | - Sung-Ho Kook
- Cluster for Craniofacial Development and Regeneration Research, School of Dentistry, Jeonbuk National University, Jeonju 54896, Korea;
- Research Center of Bioactive Materials, Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju 54896, Korea; (H.-S.S.); (T.T.T.K.)
| | - Jeong-Chae Lee
- Cluster for Craniofacial Development and Regeneration Research, School of Dentistry, Jeonbuk National University, Jeonju 54896, Korea;
- Institute of Oral Biosciences, School of Dentistry, Jeonbuk National University, Jeonju 54896, Korea
- Research Center of Bioactive Materials, Department of Bioactive Material Sciences, Jeonbuk National University, Jeonju 54896, Korea; (H.-S.S.); (T.T.T.K.)
| | - Young-Mi Jeon
- Cluster for Craniofacial Development and Regeneration Research, School of Dentistry, Jeonbuk National University, Jeonju 54896, Korea;
- Institute of Oral Biosciences, School of Dentistry, Jeonbuk National University, Jeonju 54896, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University, Jeonju 54907, Korea
| |
Collapse
|
37
|
Karim K, Giribabu N, Salleh N. Marantodes pumilum Var Alata (Kacip Fatimah) ameliorates derangement in RANK/RANKL/OPG pathway and reduces inflammation and oxidative stress in the bone of estrogen-deficient female rats with type-2 diabetes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153677. [PMID: 34333329 DOI: 10.1016/j.phymed.2021.153677] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND M. pumilum has been claimed to protect the bone against the adverse effect of estrogen deficiency. Additionally, it also exhibits anti-diabetic activity. In view of these, this study aims to identify the mechanisms underlying the bone protective effect of M. pumilum in the presence of both estrogen deficiency and diabetes mellitus (DM). METHODS Ovariectomized, diabetic female rats were given M. pumilum leave aqueous extract (MPLA) (50 and 100 mg/kg/day), estrogen, glibenclamide and estrogen plus glibenclamide for 28 consecutive days. At the end of the treatment, fasting blood glucose (FBG), serum insulin, Ca2+, PO43- and bone alkaline phosphatase (BALP) levels were measured. Rats were sacrificed and femur bones were harvested for determination of expression level and distribution of RANK, RANKL, OPG and oxidative stress and inflammatory proteins by molecular biological techniques. RESULTS 100 mg/kg/day MPLA treatment decreased the FBG and BALP levels but increased the serum insulin, Ca2+ and PO43- levels in estrogen deficient, diabetic rats. Expression and distribution of RANKL, NF-κB p65, IKKβ, IL-6, IL-1β and Keap-1 decreased however expression and distribution of RANK, OPG, BMP-2, Type-1 collagen, Runx2, TRAF6, Nrf2, NQO-1, HO-1, SOD and CAT increased in the bone of estrogen deficient, diabetic rats which received 100 mg/kg/day MPLA with greater effects than estrogen-only, glibenclamide-only and estrogen plus glibenclamide treatments. CONCLUSION MPLA helps to overcome the adverse effect of estrogen deficiency and DM on the bone and thus this herb could potentially be used for the treatment and prevention of osteoporosis in postmenopausal women with diabetes.
Collapse
Affiliation(s)
- Kamarulzaman Karim
- Department of Physiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Nelli Giribabu
- Department of Physiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Naguib Salleh
- Department of Physiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
38
|
Mei L, Zheng Y, Ma T, Xia B, Gao X, Hao Y, Luo Z, Huang J. The Novel Antioxidant Compound JSH-23 Prevents Osteolysis by Scavenging ROS During Both Osteoclastogenesis and Osteoblastogenesis. Front Pharmacol 2021; 12:734774. [PMID: 34566656 PMCID: PMC8458573 DOI: 10.3389/fphar.2021.734774] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/27/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammatory osteolysis is a pathological skeletal disease associated with not only the production of inflammatory cytokines but also local oxidative status. Excessive reactive oxygen species (ROS) promote bone resorption by osteoclasts and induce the apoptosis of osteoblasts. In consideration of the lack of effective preventive or treatments options against osteolysis, the exploitation of novel pharmacological compounds/agents is critically required. In our study, we found that a novel antioxidant compound, JSH-23, plays a role in restoring bone homeostasis by scavenging intracellular ROS during both osteoclastogenesis and osteoblastogenesis. Mechanically, JSH-23 suppressed RANKL-induced osteoclastogenesis, bone resorption and the expression of specific genes (including NFATc1, c-Fos, TRAP, CTSK and DC-STAMP) via inhibition of the NF-κB signaling pathway. Meanwhile, JSH-23 suppressed RANKL-induced ROS generation via the TRAF6/Rac1/NOX1 pathway and the enhanced expression of Nrf2/HO-1. In addition, JSH-23 attenuated H2O2-induced apoptosis and mineralization reduction in osteoblasts by reducing ROS production and enhancing Nrf2/HO-1 expression. Our in vivo results further revealed that JSH-23 exerts its protective effects on bone mass through its antioxidant activity. In conclusion, our results show that the application of JSH-23 might be a novel and plausible strategy for the treatment of osteolysis-related disease.
Collapse
Affiliation(s)
- Liangwei Mei
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Yi Zheng
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Teng Ma
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Bing Xia
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Xue Gao
- Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Yiming Hao
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Zhuojing Luo
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| | - Jinghui Huang
- Department of Orthopaedics, Xijing Hospital, the Fourth Military Medical University, Xi'an, China
| |
Collapse
|
39
|
Kubo Y, Drescher W, Fragoulis A, Tohidnezhad M, Jahr H, Gatz M, Driessen A, Eschweiler J, Tingart M, Wruck CJ, Pufe T. Adverse Effects of Oxidative Stress on Bone and Vasculature in Corticosteroid-Associated Osteonecrosis: Potential Role of Nuclear Factor Erythroid 2-Related Factor 2 in Cytoprotection. Antioxid Redox Signal 2021; 35:357-376. [PMID: 33678001 DOI: 10.1089/ars.2020.8163] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Osteonecrosis (ON) is characterized by bone tissue death due to disturbance of the nutrient artery. The detailed process leading to the necrotic changes has not been fully elucidated. Clinically, high-dose corticosteroid therapy is one of the main culprits behind osteonecrosis of the femoral head (ONFH). Recent Advances: Numerous studies have proposed that such ischemia concerns various intravascular mechanisms. Of all reported risk factors, the involvement of oxidative stress in the irreversible damage suffered by bone-related and vascular endothelial cells during ischemia simply cannot be overlooked. Several articles also have sought to elucidate oxidative stress in relation to ON using animal models or in vitro cell cultures. Critical Issues: However, as far as we know, antioxidant monotherapy has still not succeeded in preventing ONFH in humans. To provide this desideratum, we herein summarize the current knowledge about the influence of oxidative stress on ON, together with data about the preventive effects of administering antioxidants in corticosteroid-induced ON animal models. Moreover, oxidative stress is counteracted by nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent cytoprotective network through regulating antioxidant expressions. Therefore, we also describe Nrf2 regulation and highlight its role in the pathology of ON. Future Directions: This is a review of all available literature to date aimed at developing a deeper understanding of the pathological mechanism behind ON from the perspective of oxidative stress. It may be hoped that this synthesis will spark the development of a prophylactic strategy to benefit corticosteroid-associated ONFH patients. Antioxid. Redox Signal. 35, 357-376.
Collapse
Affiliation(s)
- Yusuke Kubo
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Wolf Drescher
- Department of Orthopaedic Surgery, RWTH Aachen University, Aachen, Germany.,Department of Orthopaedics and Traumatology, Rummelsberg Hospital, Schwarzenbruck, Germany
| | | | | | - Holger Jahr
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Matthias Gatz
- Department of Orthopaedic Surgery, RWTH Aachen University, Aachen, Germany
| | - Arne Driessen
- Department of Orthopaedic Surgery, RWTH Aachen University, Aachen, Germany
| | - Jörg Eschweiler
- Department of Orthopaedic Surgery, RWTH Aachen University, Aachen, Germany
| | - Markus Tingart
- Department of Orthopaedic Surgery, RWTH Aachen University, Aachen, Germany
| | - Christoph Jan Wruck
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
40
|
Hussein RM, Sawy DM, Kandeil MA, Farghaly HS. Chlorogenic acid, quercetin, coenzyme Q10 and silymarin modulate Keap1-Nrf2/heme oxygenase-1 signaling in thioacetamide-induced acute liver toxicity. Life Sci 2021; 277:119460. [PMID: 33811899 DOI: 10.1016/j.lfs.2021.119460] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS The normal functioning of Kelch-like ECH-associated protein-1 (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2) complex is necessary for the cellular protection against oxidative stress. We investigated the effect of chlorogenic acid (CGA), quercetin (Qt), coenzyme Q10 (Q10) and silymarin on the expression of Keap1/Nrf2 complex and its downstream target; heme oxygenase-1 (HO-1) as well as inflammation and apoptosis in an acute liver toxicity model induced by thioacetamide (TAA). MAIN METHODS Wistar rats were divided into 13 groups: Control, silymarin, CGA, Qt, Q10, TAA (single dose 50 mg/kg, i.p.), TAA + silymarin (400 mg/kg, p.o.), TAA + CGA (100 & 200 mg/kg, p.o.), TAA + Qt (200 &300 mg/kg, p.o.) and TAA+ Q10 (30&50 mg/kg, p.o.) and treated for 8 days. KEY FINDINGS The results showed improved liver functions and hepatic tissue integrity in all tested doses of TAA + silymarin, TAA + CGA, TAA + Qt and TAA + Q10 groups compared to the TAA group. Furthermore, these groups showed significantly lower ROS, malondialdehyde and nitric oxide levels but higher glutathione content and superoxide dismutase activity compared to the TAA group, p < 0.05. In these groups, Keap1 expression was significantly decreased while Nrf2 expression and HO-1 activity were increased. In addition, the number of apoptotic cells and the expression level of TNF-α in the liver tissues were significantly decreased compared to the TAA group. SIGNIFICANCE CGA, Qt, Q10 and silymarin protect against TAA-induced acute liver toxicity via antioxidant, anti-inflammatory, anti-apoptotic activities and regulating Keap1-Nrf2/HO-1 expression.
Collapse
Affiliation(s)
- Rasha M Hussein
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Mutah University, 61710 Al-Karak, Jordan; Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, 62514 Beni-Suef, Egypt.
| | - Doaa M Sawy
- Department of Biochemistry, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| | - Mohamed A Kandeil
- Department of Biochemistry, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Hatem S Farghaly
- Department of Biochemistry, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| |
Collapse
|
41
|
Targeting reactive oxygen species in stem cells for bone therapy. Drug Discov Today 2021; 26:1226-1244. [PMID: 33684524 DOI: 10.1016/j.drudis.2021.03.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 12/04/2020] [Accepted: 03/02/2021] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) have emerged as key players in regulating the fate and function of stem cells from both non-hematopoietic and hematopoietic lineages in bone marrow, and thus affect the osteoblastogenesis-osteoclastogenesis balance and bone homeostasis. Accumulating evidence has linked ROS and associated oxidative stress with the progression of bone disorders, and ROS-based therapeutic strategies have appeared to achieve favorable outcomes in bone. We review current knowledge of the multifactorial roles and mechanisms of ROS as a target in bone pathology. In addition, we discuss emerging ROS-based therapeutic strategies that show potential for bone therapy. Finally, we highlight the opportunities and challenges facing ROS-targeted stem cell therapeutics for improving bone health.
Collapse
|
42
|
Nrf2 epigenetic derepression induced by running exercise protects against osteoporosis. Bone Res 2021; 9:15. [PMID: 33637693 PMCID: PMC7910611 DOI: 10.1038/s41413-020-00128-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/06/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022] Open
Abstract
Osteoporosis (OP) is a common skeletal disease involving low bone mineral density (BMD) that often leads to fragility fracture, and its development is affected by multiple cellular pathologies and associated with marked epigenetic alterations of osteogenic genes. Proper physical exercise is beneficial for bone health and OP and reportedly possesses epigenetic modulating capacities; however, whether the protective effects of exercise on OP involve epigenetic mechanisms is unclear. Here, we report that epigenetic derepression of nuclear factor erythroid derived 2-related factor-2 (Nrf2), a master regulator of oxidative stress critically involved in the pathogenesis of OP, mediates the significant osteoprotective effects of running exercise (RE) in a mouse model of OP induced by ovariectomy. We showed that Nrf2 gene knockout (Nfe2l2-/-) ovariectomized mice displayed a worse BMD reduction than the controls, identifying Nrf2 as a critical antiosteoporotic factor. Further, femoral Nrf2 was markedly repressed with concomitant DNA methyltransferase (Dnmt) 1/Dnmt3a/Dnmt3b elevations and Nrf2 promoter hypermethylation in both patients with OP and ovariectomized mice. However, daily 1-h treadmill RE significantly corrected epigenetic alterations, recovered Nrf2 loss and improved the femur bone mass and trabecular microstructure. Consistently, RE also normalized the adverse expression of major osteogenic factors, including osteoblast/osteoclast markers, Nrf2 downstream antioxidant enzymes and proinflammatory cytokines. More importantly, the RE-conferred osteoprotective effects observed in the wild-type control mice were largely abolished in the Nfe2l2-/- mice. Thus, Nrf2 repression due to aberrant Dnmt elevation and subsequent Nrf2 promoter hypermethylation is likely an important epigenetic feature of the pathogenesis of OP, and Nrf2 derepression is essential for the antiosteoporotic effects of RE.
Collapse
|
43
|
Majidi Z, Ansari M, Maghbooli Z, Ghasemi A, Ebrahimi SSS, Hossein-Nezhad A, Emamgholipour S. Oligopin® Supplementation Mitigates Oxidative Stress in Postmenopausal Women with Osteopenia: A Randomized, Double-blind, Placebo-Controlled Trial. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 81:153417. [PMID: 33250314 DOI: 10.1016/j.phymed.2020.153417] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/14/2020] [Accepted: 11/09/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Evidence indicates a close association between oxidative stress and the etiopathogenesis of osteopenia. In vitro and animal studies report that Oligopin®, an extract of French maritime pine bark extract, has beneficial effects on oxidative stress. PURPOSE Here, we aimed to determine whether supplementation with Oligopin® affects bone turnover markers, antioxidant enzymes, and oxidative stress markers in these patients. METHODS Forty-three postmenopausal women with osteopenia were randomized in a placebo-controlled, double-blind clinical trial to receive either 150 mg/day Oligopin® (n = 22) or placebo (n = 21) for 12 weeks. Plasma levels of bone turnover markers; osteocalcin (OC), type I collagen cross-linked C-telopeptide (CTX-1), OC/CTX1 ratio along with total antioxidant capacity(TAC), malondialdehyde (MDA) concentration, protein carbonyl, and total thiol contents in plasma, activities of manganese superoxide dismutase (MnSOD) and catalase in both peripheral blood mononuclear cells (PBMCs) and plasma as well as mRNA expression of MnSOD, catalase, and Nrf2 in PBMCs were measured at the baseline and the end of the intervention. RESULTS Oligopin® supplementation significantly increased OC levels and the ratio of OC to CTX1 in women with osteopenia compared to placebo intervention after 12 weeks. Oligopin® significantly decreased plasma protein carbonyl content in postmenopausal women compared with the after placebo treatment. Moreover, Oligopin® intervention significantly increased plasma total thiol content, TAC, plasma activity of both MnSOD and catalase, and the transcript level of Nrf2, MnSOD, and catalase in comparison with the placebo group. CONCLUSION Supplementation with 150 mg/day Oligopin® for 12 weeks exerts beneficial effects in postmenopausal osteopenia through improving the antioxidant defense system in the plasma and PBMCs that was accompanied by an increase in indicators of bone turnover.
Collapse
Affiliation(s)
- Ziba Majidi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ansari
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zhila Maghbooli
- MS Research Center, Neurosciences Institute of Tehran University of Medical Sciences, Tehran, Iran
| | - Afsaneh Ghasemi
- Department of Obstetrics and Gynecology, Akbar Abadi Teaching Hospital, Iran University of Medical Sciences and Health Services, Tehran, Iran
| | - Shadi Sadat Seyyed Ebrahimi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Hossein-Nezhad
- Department of Medicine, Section of Endocrinology, Nutrition, and Diabetes, Vitamin D, Skin and Bone Research Laboratory, Boston University Medical Center, Boston, Massachusetts, United States of America.
| | - Solaleh Emamgholipour
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
44
|
Sánchez-de-Diego C, Pedrazza L, Pimenta-Lopes C, Martinez-Martinez A, Dahdah N, Valer JA, Garcia-Roves P, Rosa JL, Ventura F. NRF2 function in osteocytes is required for bone homeostasis and drives osteocytic gene expression. Redox Biol 2020; 40:101845. [PMID: 33373776 PMCID: PMC7773566 DOI: 10.1016/j.redox.2020.101845] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 12/21/2020] [Indexed: 12/17/2022] Open
Abstract
Osteocytes, the most abundant bone cell type, are derived from osteoblasts through a process in which they are embedded in an osteoid. We previously showed that nutrient restriction promotes the osteocyte transcriptional program and is associated with increased mitochondrial biogenesis. Here, we show that increased mitochondrial biogenesis increase reactive oxygen species (ROS) levels and consequently, NRF2 activity during osteocytogenesis. NRF2 activity promotes osteocyte-specific expression of Dmp1, Mepe, and Sost in IDG-SW3 cells, primary osteocytes, and osteoblasts, and in murine models with Nfe2l2 deficiency in osteocytes or osteoblasts. Moreover, ablation of Nfe2l2 in osteocytes or osteoblasts generates osteopenia and increases osteoclast numbers with marked sexual dimorphism. Finally, treatment with dimethyl fumarate prevented the deleterious effects of ovariectomy in trabecular bone masses of mice and restored osteocytic gene expression. Altogether, we uncovered the role of NRF2 activity in osteocytes during the regulation of osteocyte gene expression and maintenance of bone homeostasis. ROS levels and NRF2 activity are increased during osteocytogenesis. NRF2 drives osteocyte specification and activate the transcription of osteocyte-specific genes. NRF2 in osteocytes has a fundamental role in bone homeostasis and its deletion induces osteopenia. Activation of NRF2 with dimethyl fumarate prevents osteopenia induced by ovariectomy.
Collapse
Affiliation(s)
- Cristina Sánchez-de-Diego
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Leonardo Pedrazza
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Carolina Pimenta-Lopes
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Arturo Martinez-Martinez
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Norma Dahdah
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - José Antonio Valer
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Pablo Garcia-Roves
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Jose Luis Rosa
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Francesc Ventura
- Departament de Ciències Fisiològiques, Universitat de Barcelona, IDIBELL, L'Hospitalet de Llobregat, Spain.
| |
Collapse
|
45
|
He C, He W, Hou J, Chen K, Huang M, Yang M, Luo X, Li C. Bone and Muscle Crosstalk in Aging. Front Cell Dev Biol 2020; 8:585644. [PMID: 33363144 PMCID: PMC7758235 DOI: 10.3389/fcell.2020.585644] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoporosis and sarcopenia are two age-related diseases that affect the quality of life in the elderly. Initially, they were thought to be two independent diseases; however, recently, increasing basic and clinical data suggest that skeletal muscle and bone are both spatially and metabolically connected. The term "osteosarcopenia" is used to define a condition of synergy of low bone mineral density with muscle atrophy and hypofunction. Bone and muscle cells secrete several factors, such as cytokines, myokines, and osteokines, into the circulation to influence the biological and pathological activities in local and distant organs and cells. Recent studies reveal that extracellular vesicles containing microRNAs derived from senescent skeletal muscle and bone cells can also be transported and aid in regulating bone-muscle crosstalk. In this review, we summarize the age-related changes in the secretome and extracellular vesicle-microRNAs secreted by the muscle and bone, and discuss their interactions between muscle and bone cells during aging.
Collapse
Affiliation(s)
- Chen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Wenzhen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Jing Hou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Kaixuan Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Mei Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Mi Yang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| | - Changjun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| |
Collapse
|
46
|
Proanthocyanidins-Mediated Nrf2 Activation Ameliorates Glucocorticoid-Induced Oxidative Stress and Mitochondrial Dysfunction in Osteoblasts. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9102012. [PMID: 33062149 PMCID: PMC7533007 DOI: 10.1155/2020/9102012] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/22/2020] [Accepted: 07/17/2020] [Indexed: 12/25/2022]
Abstract
The widespread use of therapeutic glucocorticoids has increased the frequency of glucocorticoid-induced osteoporosis (GIOP). One of the potential pathological processes of GIOP is an increased level of oxidative stress and mitochondrial dysfunction, which eventually leads to osteoblast apoptosis. Proanthocyanidins (PAC) are plant-derived antioxidants that have therapeutic potential against GIOP. In our study, a low dose of PAC was nontoxic to healthy osteoblasts and restored osteogenic function in dexamethasone- (Dex-) treated osteoblasts by suppressing oxidative stress, mitochondrial dysfunction, and apoptosis. Mechanistically, PAC neutralized Dex-induced damage in the osteoblasts by activating the Nrf2 pathway, since silencing Nrf2 partly eliminated the protective effects of PAC. Furthermore, PAC injection restored bone mass and promoted the expression of Nrf2 in the distal femur of Dex-treated osteoporotic rats. In summary, PAC protect osteoblasts against Dex-induced oxidative stress and mitochondrial dysfunction via the Nrf2 pathway activation and may be a promising drug for treating GIOP.
Collapse
|
47
|
Park PSU, Mun SH, Zeng SL, Kim H, Bae S, Park-Min KH. NRF2 Is an Upstream Regulator of MYC-Mediated Osteoclastogenesis and Pathological Bone Erosion. Cells 2020; 9:E2133. [PMID: 32967239 PMCID: PMC7564846 DOI: 10.3390/cells9092133] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/07/2020] [Accepted: 09/17/2020] [Indexed: 12/30/2022] Open
Abstract
Osteoclasts are the sole bone-resorbing cells that play an essential role in homeostatic bone remodeling and pathogenic bone destruction such as inflammatory arthritis. Pharmacologically targeting osteoclasts has been a promising approach to alleviating bone disease, but there remains room for improvement in mitigating drug side effects and enhancing cell specificity. Recently, we demonstrated the crucial role of MYC and its downstream effectors in driving osteoclast differentiation. Despite these advances, upstream regulators of MYC have not been well defined. In this study, we identify nuclear factor erythroid 2-related factor 2 (NRF2), a transcription factor known to regulate the expression of phase II antioxidant enzymes, as a novel upstream regulator of MYC. NRF2 negatively regulates receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclastogenesis through the ERK and p38 signaling-mediated suppression of MYC transcription. Furthermore, the ablation of MYC in osteoclasts reverses the enhanced osteoclast differentiation and activity in NRF2 deficiency in vivo and in vitro in addition to protecting NRF2-deficient mice from pathological bone loss in a murine model of inflammatory arthritis. Our findings indicate that this novel NRF2-MYC axis could be instrumental for the fine-tuning of osteoclast formation and provides additional ways in which osteoclasts could be therapeutically targeted to prevent pathological bone erosion.
Collapse
Affiliation(s)
- Peter Sang Uk Park
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA; (P.S.U.P.); (S.H.M.); (S.L.Z.); (H.K.)
| | - Se Hwan Mun
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA; (P.S.U.P.); (S.H.M.); (S.L.Z.); (H.K.)
| | - Steven L. Zeng
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA; (P.S.U.P.); (S.H.M.); (S.L.Z.); (H.K.)
| | - Haemin Kim
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA; (P.S.U.P.); (S.H.M.); (S.L.Z.); (H.K.)
- Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Seyeon Bae
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA; (P.S.U.P.); (S.H.M.); (S.L.Z.); (H.K.)
- Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Kyung-Hyun Park-Min
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA; (P.S.U.P.); (S.H.M.); (S.L.Z.); (H.K.)
- Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
- BCMB Allied Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10021, USA
| |
Collapse
|
48
|
Chen R, Liu G, Sun X, Cao X, He W, Lin X, Liu Q, Zhao J, Pang Y, Li B, Qin A. Chitosan derived nitrogen-doped carbon dots suppress osteoclastic osteolysis via downregulating ROS. NANOSCALE 2020; 12:16229-16244. [PMID: 32706362 DOI: 10.1039/d0nr02848g] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Osteoclasts are the main cells involved in normal bone remodeling and pathological bone destruction in vivo. Overactivation of osteoclasts can lead to osteolytic diseases, including breast cancer, bone tumors, arthritis, the aseptic loosening of orthopedic implants, and Paget's disease. Excessive reactive oxygen species are the main cause of osteoclast overactivation. We have synthesized chitosan derived nitrogen-doped carbon dots (N-CDs) with a high synthetic yield and the ability to scavenge reactive oxygen species (ROS). N-CDs effectively abrogated RANKL-induced elevation in ROS generation and therefore impaired the activation of NF-κB and MAPK pathways. Osteoclastogenesis and bone resorption was effectively attenuated in vitro. Furthermore, the in vivo administration of N-CDs in mice protected them against lipopolysaccharide (LPS)-induced calvarial bone destruction and breast cancer cell-induced tibial bone loss. Based on the good biocompatibility of N-CDs and the ability to efficiently remove ROS, a nanomaterial treatment scheme was provided for the first time for the clinical treatment of osteolytic diseases.
Collapse
Affiliation(s)
- Runfeng Chen
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Guangxi, 530021, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Park C, Lee H, Han MH, Jeong JW, Kim SO, Jeong SJ, Lee BJ, Kim GY, Park EK, Jeon YJ, Choi YH. Cytoprotective effects of fermented oyster extracts against oxidative stress-induced DNA damage and apoptosis through activation of the Nrf2/HO-1 signaling pathway in MC3T3-E1 osteoblasts. EXCLI JOURNAL 2020; 19:1102-1119. [PMID: 33013267 PMCID: PMC7527492 DOI: 10.17179/excli2020-2376] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022]
Abstract
Osteoblast damage by oxidative stress has been recognized as a cause of bone-related disease, including osteoporosis. Recently, we reported that fermented Pacific oyster (Crassostrea gigas) extracts (FO) inhibited osteoclastogenesis and osteoporosis, while promoting osteogenesis. However, since the beneficial potential of FO on osteoblasts is not well known, in the present study, we investigated the cytoprotective effect of FO against oxidative stress in MC3T3-E1 osteoblasts. Our results demonstrated that FO inhibited hydrogen peroxide (H2O2)-induced DNA damage and cytotoxicity through the rescue of mitochondrial function by blocking abnormal ROS accumulation. FO also prevented apoptosis by suppressing loss of mitochondrial membrane potential and cytosolic release of cytochrome c, decreasing the rate of Bax/Bcl-2 expression and reducing the activity of caspase-9 and caspase-3 in H2O2-stimulated MC3T3-E1 osteoblasts, suggesting that FO protected MC3T3-E1 osteoblasts from the induction of caspase dependent- and mitochondria-mediated apoptosis by oxidative stress. In addition, FO markedly promoted the activation of nuclear factor-erythroid-2-related factor 2 (Nrf2), which was associated with the enhanced expression of heme oxygenase-1 (HO-1). However, inhibiting the expression of HO-1 by artificially blocking the expression of Nrf2 using siRNA significantly eliminated the protective effect of FO, indicating that FO activates the Nrf2/HO-1 signaling pathway in MC3T3-E1 osteoblasts to protect against oxidative stress. Based on the present data, FO is thought to be useful as a potential therapeutic agent for the inhibition of oxidative stress in osteoblasts.
Collapse
Affiliation(s)
- Cheol Park
- Division of Basic Sciences, College of Liberal Studies, Dong?eui University, Busan, Republic of Korea
| | - Hyesook Lee
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan, Republic of Korea
| | - Min Ho Han
- National Marine Biodiversity Institute of Korea, Seocheon, Republic of Korea
| | - Jin-Woo Jeong
- Freshwater Bioresources Utilization Bureau, Nakdonggang National Institute of Biological Resources, Sangju, Republic of Korea
| | - Sung Ok Kim
- Department of Food Science and Biotechnology, College of Engineering, Kyungsung University, Busan, Republic of Korea
| | - Soon-Jeong Jeong
- Department of Dental Hygiene, College of Health Science, Youngsan University, Yangsan, Republic of Korea
| | - Bae-Jin Lee
- Ocean Fisheries & Biology Center, Marine Bioprocess Co., Ltd., Busan, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju, Republic of Korea
| | - Eui Kyun Park
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - You-Jin Jeon
- Department of Marine Life Science, Jeju National University, Jeju, Republic of Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan, Republic of Korea
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan, Republic of Korea
| |
Collapse
|
50
|
Li X, Chen Y, Mao Y, Dai P, Sun X, Zhang X, Cheng H, Wang Y, Banda I, Wu G, Ma J, Huang S, Forouzanfar T. Curcumin Protects Osteoblasts From Oxidative Stress-Induced Dysfunction via GSK3β-Nrf2 Signaling Pathway. Front Bioeng Biotechnol 2020; 8:625. [PMID: 32612986 PMCID: PMC7308455 DOI: 10.3389/fbioe.2020.00625] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 05/21/2020] [Indexed: 12/14/2022] Open
Abstract
Osteoblasts dysfunction, induced by oxidative stress (OS), is one of major pathological mechanisms for osteoporosis. Curcumin (Cur), a bioactive antioxidant compound, isolated from Curcumin longa L, was regarded as a strong reactive oxygen species (ROS) scavenger. However, it remains unveiled whether Cur can prevent osteoblasts from OS-induced dysfunction. To approach this question, we adopted a well-established OS model to investigate the preventive effect of Cur on osteoblasts dysfunction by measuring intracellular ROS production, cell viability, apoptosis rate and osteoblastogenesis markers. We showed that the pretreatment of Cur could significantly antagonize OS so as to suppress endogenous ROS production, maintain osteoblasts viability and promote osteoblastogenesis. Inhibiting Glycogen synthase kinase (GSK3β) and activating nuclear factor erythroid 2 related factor 2 (Nrf2) could significantly antagonize the destructive effects of OS, which indicated the critical role of GSK3β-Nrf2 signaling. Furthermore, Cur also abolished the suppressive effects of OS on GSK3β-Nrf2 signaling pathway. Our findings demonstrated that Cur could protect osteoblasts against OS-induced dysfunction via GSK3β-Nrf2 signaling and provide a promising way for osteoporosis treatment.
Collapse
Affiliation(s)
- Xumin Li
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam, MOVE Research Institute, University of Amsterdam and Vrije University Amsterdam, Amsterdam, Netherlands.,Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Department of Oral and Maxillofacial Surgary/Pathology, Amsterdam UMC and Academic Centre for Dentistry Amsterdam, Amsterdam Movement Science, Vrije Universitetit Amsterdam, Amsterdam, Netherlands
| | - Yang Chen
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yixin Mao
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Laboratory for Myology, Amsterdam Movement Sciences, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Panpan Dai
- Department of Stomatology, Taizhou Hospital, Wenzhou Medical University, Linhai, China
| | - Xiaoyu Sun
- Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam, MOVE Research Institute, University of Amsterdam and Vrije University Amsterdam, Amsterdam, Netherlands.,Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Department of Periodontology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Xiaorong Zhang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Haoran Cheng
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yingting Wang
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Isaac Banda
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Gang Wu
- Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam, MOVE Research Institute, University of Amsterdam and Vrije University Amsterdam, Amsterdam, Netherlands
| | - Jianfeng Ma
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Shengbin Huang
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam, MOVE Research Institute, University of Amsterdam and Vrije University Amsterdam, Amsterdam, Netherlands.,Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Tim Forouzanfar
- Department of Oral and Maxillofacial Surgary/Pathology, Amsterdam UMC and Academic Centre for Dentistry Amsterdam, Amsterdam Movement Science, Vrije Universitetit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|