1
|
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
|
2
|
Sandhu S, Keyworth M, Karimi-Jashni S, Alomar D, Smith BJ, Kozbenko T, Doty S, Hocking R, Hamada N, Reynolds RJ, Scott RT, Costes SV, Beheshti A, Yauk C, Wilkins RC, Chauhan V. AOP Report: Development of an adverse outcome pathway for deposition of energy leading to bone loss. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2024; 65 Suppl 3:85-111. [PMID: 39387375 DOI: 10.1002/em.22631] [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: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 10/15/2024]
Abstract
Bone loss, commonly seen in osteoporosis, is a condition that entails a progressive decline of bone mineral density and microarchitecture, often seen in post-menopausal women. Bone loss has also been widely reported in astronauts exposed to a plethora of stressors and in patients with osteoporosis following radiotherapy for cancer. Studies on mechanisms are well documented but the causal connectivity of events to bone loss development remains incompletely understood. Herein, the adverse outcome pathway (AOP) framework was used to organize data and develop a qualitative AOP beginning from deposition of energy (the molecular initiating event) to bone loss (the adverse outcome). This qualitative AOP was developed in collaboration with bone loss research experts to aggregate relevant findings, supporting ongoing efforts to understand and mitigate human system risks associated with radiation exposures. A literature review was conducted to compile and evaluate the state of knowledge based on the modified Bradford Hill criteria. Following review of 2029 studies, an empirically supported AOP was developed, showing the progression to bone loss through many factors affecting the activities of bone-forming osteoblasts and bone-resorbing osteoclasts. The structural, functional, and quantitative basis of each proposed relationship was defined, for inference of causal changes between key events. Current knowledge and its gaps relating to dose-, time- and incidence-concordance across the key events were identified, as well as modulating factors that influence linkages. The new priorities for research informed by the AOP highlight areas for improvement to enable development of a quantitative AOP used to support risk assessment strategies for space travel or cancer radiotherapy.
Collapse
Affiliation(s)
- Snehpal Sandhu
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Mitchell Keyworth
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Syna Karimi-Jashni
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Dalya Alomar
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Benjamin J Smith
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Tatiana Kozbenko
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Stephen Doty
- Hospital for Special Surgery Research Institute, New York City, New York, USA
| | - Robyn Hocking
- Learning and Knowledge and Library Services, Health Canada, Ottawa, Ontario, Canada
| | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Substantiable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Chiba, Japan
| | | | - Ryan T Scott
- KBR, NASA Ames Research Center, Moffett Field, California, USA
| | - Sylvain V Costes
- NASA Ames Research Center, Space Biosciences Research Branch, Mountain View, California, USA
| | - Afshin Beheshti
- McGowan Institute for Regenerative Medicine - Center for Space Biomedicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Carole Yauk
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Ruth C Wilkins
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Vinita Chauhan
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Liu N, Sun Y. microRNA-148a-3p-targeting p300 protects against osteoblast differentiation and osteoporotic bone reconstruction. Regen Med 2021; 16:435-449. [PMID: 34000812 DOI: 10.2217/rme-2020-0006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: This study sets out to investigate the possible effects of miRNA-148a-3p (miR-148a-3p) on osteoblast differentiation and bone remodeling following osteoporosis. Materials & methods: Expression of miR-148a-3p, p300, Nrf2 and differentiation-related proteins (Runx2, Osteocalcin and Col1a1) was examined in the osteoblast MC3T3-E1 cell line, followed by identification of interaction between miR-148a-3p and p300 and between p300 and Nrf2. After ectopic expression and depletion experiments in MC3T3-E1 cells, cell proliferation, osteogenic mineralization and osteogenic differentiation were measured. Ovariectomy-induced osteoporosis mouse models were established to verify function of miR-148a-3p in vivo. Results: miR-148a-3p expression was restrained and p300 and Nrf2 expression was increased during osteoblast differentiation. miR-148a-3p inhibition or p300 upregulation enhanced proliferation and osteogenic differentiation in MC3T3-E1 cells. p300 was targeted by miR-148a-3p. Additionally, miR-148a-3p reduced BMD, bone volume relative to tissue volume ratio, trabecular bone, trabecular thickness and trabecular spacing in ovariectomy mice. Conclusion: Taken together, miR-148a-3p might prevent the osteoblast differentiation and bone remodeling by disrupting p300-dependent Nrf2 pathway activation.
Collapse
Affiliation(s)
- Ning Liu
- Department of Rehabilitation, The First Affiliated Hospital of China Medical University, Shenyang 110000, PR China
| | - Yongxin Sun
- Department of Rehabilitation, The First Affiliated Hospital of China Medical University, Shenyang 110000, PR China
| |
Collapse
|
6
|
Li R, Yang W, Hu X, Zhou D, Huang K, Wang C, Li Y, Liu B. Effect of autophagy on irradiation‑induced damage in osteoblast‑like MC3T3‑E1 cells. Mol Med Rep 2020; 22:3473-3481. [PMID: 32945432 PMCID: PMC7453677 DOI: 10.3892/mmr.2020.11425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 07/16/2020] [Indexed: 12/05/2022] Open
Abstract
Autophagy is activated under radiation stress, which serves an important role in maintaining bone homeostasis. However, the underlying mechanisms of irradiation-induced autophagy in bone homeostasis is not well understood. The present study aimed to determine the effects of radiation-activated autophagy on pre-osteoblastic MC3T3-E1 cells. X-ray irradiation activated autophagy in a dose-dependent manner, with an increased fluorescence intensity of monodansylcadaverine staining, increased ratio of microtubule-associated protein 1 light chain 3β (LC3)-II/LC3-I, decreased p62 expression, and increased ATG5 and beclin-1 expression levels in MC3T3-E1 cells 72 h after irradiation compared with those in non-irradiated MC3T3-E1 cells. Irradiation reduced colony formation and mineralization in a dose-dependent manner in MC3T3-E1 cells at 2 and 3 weeks after irradiation, respectively. Decreased levels of alkaline phosphatase activity and runt-related transcription factor 2 expression were observed at 72 h post-irradiation. In addition, irradiation-induced apoptosis was accompanied by a decreased ratio of Bcl-2/BAX protein and increased the activity of caspase-3. By contrast, doxycycline (DOX)-inhibited autophagy attenuated the decreased colony formation and mineralization, and aggravated the increased cell apoptosis in irradiated MC3T3-E1 cells. Furthermore, the ratio of phosphorylated P38/P38 was observed to be higher following DOX treatment within 1 week of irradiation, which was reversed 2 weeks post-irradiation. In conclusion, DOX-inhibited autophagy aggravated X-ray irradiation-induced apoptosis at an early stage, but maintained cell proliferation and mineralization at a late stage in irradiated MC3T3-E1 cells.
Collapse
Affiliation(s)
- Rui Li
- School of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Wenke Yang
- School of Basic Medical Science, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xurui Hu
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Dan Zhou
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Ke Huang
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Chenwei Wang
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yi Li
- School of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Bin Liu
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
7
|
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
|
8
|
New Insights into the Nrf-2/HO-1 Signaling Axis and Its Application in Pediatric Respiratory Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3214196. [PMID: 31827672 PMCID: PMC6885770 DOI: 10.1155/2019/3214196] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/19/2019] [Accepted: 11/02/2019] [Indexed: 12/13/2022]
Abstract
Respiratory diseases are one of the most common pediatric diseases in clinical practice. Their pathogenesis, diagnosis, and treatment are thus worthy of further investigation. The nuclear factor erythroid 2-related factor 2/heme oxygenase 1 (Nrf2/HO-1) signaling axis is a multiple organ protection chain that protects against oxidative stress injury. This signaling axis regulates anti-inflammation and antioxidation by regulating calcium ions, mitochondrial oxidative stress, autophagy, ferroptosis, pyroptosis, apoptosis, alkaliptosis, and clockophagy. This review presents an overview of the role of the Nrf2/HO-1 signaling axis in the pathogenesis of pediatric respiratory diseases and the latest research progress on this subject. Overall, the Nrf2/HO-1 signaling axis has an important clinical value in pediatric respiratory diseases, and its protective effect needs further exploration.
Collapse
|
9
|
Li G, Jiang X, Liu L, Liu X, Liu H, Zhang Z. Effect of estradiol on high glucose‑induced osteoblast injury. Mol Med Rep 2019; 20:3019-3026. [PMID: 31432111 PMCID: PMC6755179 DOI: 10.3892/mmr.2019.10552] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 04/12/2019] [Indexed: 01/06/2023] Open
Abstract
Estradiol (E2) serves an important role in the changes of postmenopausal bone turnover rate and the development of osteoporosis. The present study aimed to investigate the effects of E2 on high glucose (HG)‑induced osteoblast injury. Cell Counting Kit‑8 was used to determine cell viability. Reverse transcription‑quantitative PCR (RT‑qPCR) and western blotting was used to analyze the mRNA and protein expression levels of osteocalcin, Runt‑related transcription factor 2 (Runx2), nuclear factor E2‑related factor 2 (Nrf2) and heme oxygenase‑1 (HO1). Flow cytometry was performed to analyze apoptosis. The results revealed that cell viability was lower in cells treated with HG (100, 200 or 300 mg/dl) compared with the control group. Cell viability was decreased in cells treated with 200 mg/dl HG on days 3, 5 and 7. In addition, cell viability was increased by 0.1 µM E2. E2 with HG co‑treatment increased cell viability, osteocalcin and Runx2 mRNA expression levels and nuclear Nrf2 and HO1 protein expression levels compared with the HG‑only group. All these changes, with the exception of Runx2, were reversed by silencing Nrf2 expression using small interfering (si)RNA (siNrf2). Additionally, apoptosis was reduced by E2 in HG‑treated cells, which was reversed by siNrf2 transfection. These results demonstrated that E2 may prevent HG‑induced osteoblast injury by activating Nrf2/HO1 signaling pathways.
Collapse
Affiliation(s)
- Guangrun Li
- Department of Spinal Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Xiaofeng Jiang
- Department of Joint Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Liping Liu
- Department of Allergy, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Xiaoyang Liu
- Department of Spinal Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Hongtao Liu
- Department of Spinal Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Zuofu Zhang
- Department of Joint Surgery, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
10
|
Yang J, Zhang G, Dong D, Shang P. Effects of Iron Overload and Oxidative Damage on the Musculoskeletal System in the Space Environment: Data from Spaceflights and Ground-Based Simulation Models. Int J Mol Sci 2018; 19:E2608. [PMID: 30177626 PMCID: PMC6163331 DOI: 10.3390/ijms19092608] [Citation(s) in RCA: 23] [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: 08/15/2018] [Revised: 08/29/2018] [Accepted: 09/01/2018] [Indexed: 12/15/2022] Open
Abstract
The space environment chiefly includes microgravity and radiation, which seriously threatens the health of astronauts. Bone loss and muscle atrophy are the two most significant changes in mammals after long-term residency in space. In this review, we summarized current understanding of the effects of microgravity and radiation on the musculoskeletal system and discussed the corresponding mechanisms that are related to iron overload and oxidative damage. Furthermore, we enumerated some countermeasures that have a therapeutic potential for bone loss and muscle atrophy through using iron chelators and antioxidants. Future studies for better understanding the mechanism of iron and redox homeostasis imbalance induced by the space environment and developing the countermeasures against iron overload and oxidative damage consequently may facilitate human to travel more safely in space.
Collapse
Affiliation(s)
- Jiancheng Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Gejing Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Dandan Dong
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China.
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Peng Shang
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an 710072, China.
- Research & Development Institute in Shenzhen, Northwestern Polytechnical University, Shenzhen 518057, China.
| |
Collapse
|
11
|
The Impact of Oxidative Stress on the Bone System in Response to the Space Special Environment. Int J Mol Sci 2017; 18:ijms18102132. [PMID: 29023398 PMCID: PMC5666814 DOI: 10.3390/ijms18102132] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/28/2017] [Accepted: 10/09/2017] [Indexed: 12/25/2022] Open
Abstract
The space special environment mainly includes microgravity, radiation, vacuum and extreme temperature, which seriously threatens an astronaut’s health. Bone loss is one of the most significant alterations in mammalians after long-duration habitation in space. In this review, we summarize the crucial roles of major factors—namely radiation and microgravity—in space in oxidative stress generation in living organisms, and the inhibitory effect of oxidative stress on bone formation. We discussed the possible mechanisms of oxidative stress-induced skeletal involution, and listed some countermeasures that have therapeutic potentials for bone loss via oxidative stress antagonism. Future research for better understanding the oxidative stress caused by space environment and the development of countermeasures against oxidative damage accordingly may facilitate human beings to live more safely in space and explore deeper into the universe.
Collapse
|
12
|
Alwood JS, Tran LH, Schreurs AS, Shirazi-Fard Y, Kumar A, Hilton D, Tahimic CGT, Globus RK. Dose- and Ion-Dependent Effects in the Oxidative Stress Response to Space-Like Radiation Exposure in the Skeletal System. Int J Mol Sci 2017; 18:ijms18102117. [PMID: 28994728 PMCID: PMC5666799 DOI: 10.3390/ijms18102117] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 09/29/2017] [Accepted: 09/30/2017] [Indexed: 12/12/2022] Open
Abstract
Space radiation may pose a risk to skeletal health during subsequent aging. Irradiation acutely stimulates bone remodeling in mice, although the long-term influence of space radiation on bone-forming potential (osteoblastogenesis) and possible adaptive mechanisms are not well understood. We hypothesized that ionizing radiation impairs osteoblastogenesis in an ion-type specific manner, with low doses capable of modulating expression of redox-related genes. 16-weeks old, male, C57BL6/J mice were exposed to low linear-energy-transfer (LET) protons (150 MeV/n) or high-LET 56Fe ions (600 MeV/n) using either low (5 or 10 cGy) or high (50 or 200 cGy) doses at NASA's Space Radiation Lab. Five weeks or one year after irradiation, tissues were harvested and analyzed by microcomputed tomography for cancellous microarchitecture and cortical geometry. Marrow-derived, adherent cells were grown under osteoblastogenic culture conditions. Cell lysates were analyzed by RT-PCR during the proliferative or mineralizing phase of growth, and differentiation was analyzed by imaging mineralized nodules. As expected, a high dose (200 cGy), but not lower doses, of either 56Fe or protons caused a loss of cancellous bone volume/total volume. Marrow cells produced mineralized nodules ex vivo regardless of radiation type or dose; 56Fe (200 cGy) inhibited osteoblastogenesis by more than 90% (5 weeks and 1 year post-IR). After 5 weeks, irradiation (protons or 56Fe) caused few changes in gene expression levels during osteoblastogenesis, although a high dose 56Fe (200 cGy) increased Catalase and Gadd45. The addition of exogenous superoxide dismutase (SOD) protected marrow-derived osteoprogenitors from the damaging effects of exposure to low-LET (137Cs γ) when irradiated in vitro, but had limited protective effects on high-LET 56Fe-exposed cells. In sum, either protons or 56Fe at a relatively high dose (200 cGy) caused persistent bone loss, whereas only high-LET 56Fe increased redox-related gene expression, albeit to a limited extent, and inhibited osteoblastogenesis. Doses below 50 cGy did not elicit widespread responses in any parameter measured. We conclude that high-LET irradiation at 200 cGy impaired osteoblastogenesis and regulated steady-state gene expression of select redox-related genes during osteoblastogenesis, which may contribute to persistent bone loss.
Collapse
Affiliation(s)
- Joshua S Alwood
- Bone and Signaling Laboratory, Space BioSciences Division, NASA Ames Research Center, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
| | - Luan H Tran
- Bone and Signaling Laboratory, Space BioSciences Division, NASA Ames Research Center, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
| | - Ann-Sofie Schreurs
- Bone and Signaling Laboratory, Space BioSciences Division, NASA Ames Research Center, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
| | - Yasaman Shirazi-Fard
- Bone and Signaling Laboratory, Space BioSciences Division, NASA Ames Research Center, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
| | - Akhilesh Kumar
- Bone and Signaling Laboratory, Space BioSciences Division, NASA Ames Research Center, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
| | - Diane Hilton
- Bone and Signaling Laboratory, Space BioSciences Division, NASA Ames Research Center, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
| | - Candice G T Tahimic
- Bone and Signaling Laboratory, Space BioSciences Division, NASA Ames Research Center, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
- Wyle Laboratories, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
| | - Ruth K Globus
- Bone and Signaling Laboratory, Space BioSciences Division, NASA Ames Research Center, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
| |
Collapse
|
13
|
Marcoux S, Drouin S, Laverdière C, Alos N, Andelfinger GU, Bertout L, Curnier D, Friedrich MG, Kritikou EA, Lefebvre G, Levy E, Lippé S, Marcil V, Raboisson MJ, Rauch F, Robaey P, Samoilenko M, Séguin C, Sultan S, Krajinovic M, Sinnett D. The PETALE study: Late adverse effects and biomarkers in childhood acute lymphoblastic leukemia survivors. Pediatr Blood Cancer 2017; 64. [PMID: 27917589 DOI: 10.1002/pbc.26361] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/27/2016] [Accepted: 10/17/2016] [Indexed: 02/02/2023]
Abstract
BACKGROUND Childhood cancer survivorship issues represent an established public health challenge. Most late adverse effects (LAEs) have been demonstrated to be time and treatment dependent. The PETALE study is a multidisciplinary research project aiming to comprehensively characterize LAEs and identify associated predictive biomarkers in childhood acute lymphoblastic leukemia (cALL) survivors. METHODS cALL survivors treated at Sainte-Justine University Health Center with Dana-Farber Cancer Institution-ALL protocols 87-01 through 2005-01 were eligible. During Phase I of the study, the participants underwent comprehensive clinical, biologic, and psychosocial investigation targeting metabolic syndrome, cardiotoxicity, bone morbidity, neurocognitive problems, and quality of life issues. Whole-exome sequencing was performed for all participants. Subjects identified with an extreme phenotype during Phase I were recalled for additional testing (Phase II). RESULTS Phase I included 246 survivors (recall rate 71.9%). Of those, 85 participants completed Phase II (recall rate 88.5%). Survivors agreeing to participate in Phase I (n = 251) were similar to those who refused (n = 31) in terms of relapse risk profile, radiotherapy exposure, and age at the time of study. Participants, however, tended to be slightly older at diagnosis (6.1 vs. 4.7 years old, P = 0.08), with a higher proportion of female agreeing to participate compared with males (93.2 vs. 86.5%, P = 0.07). CONCLUSION The PETALE study will contribute to comprehensively characterize clinical, psychosocial, biologic, and genomic features of cALL survivors using an integrated approach. Expected outcomes include LAE early detection biomarkers, long-term follow-up guidelines, and recommendations for physicians and health professionals.
Collapse
Affiliation(s)
- Sophie Marcoux
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Simon Drouin
- Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Caroline Laverdière
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Nathalie Alos
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Gregor U Andelfinger
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Laurence Bertout
- Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Daniel Curnier
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | | | | | | | - Emile Levy
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Sarah Lippé
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Valérie Marcil
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Marie-Josée Raboisson
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Frank Rauch
- Shriners Hospitals for Children, Montréal, Québec, Canada
| | - Philippe Robaey
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada.,Children's Hospital of Eastern Ontario (CHEO), Ottawa, Ontario, Canada
| | | | - Chantal Séguin
- McGill University Health Center Research Institute, Montréal, Québec, Canada
| | - Serge Sultan
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Maja Krajinovic
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| | - Daniel Sinnett
- Université de Montréal, Montréal, Québec, Canada.,Sainte-Justine University Health Center (SJUHC), Montréal, Québec, Canada
| |
Collapse
|
14
|
Gong W, Li J, Chen Z, Huang J, Chen Q, Cai W, Liu P, Huang H. Polydatin promotes Nrf2-ARE anti-oxidative pathway through activating CKIP-1 to resist HG-induced up-regulation of FN and ICAM-1 in GMCs and diabetic mice kidneys. Free Radic Biol Med 2017; 106:393-405. [PMID: 28286065 DOI: 10.1016/j.freeradbiomed.2017.03.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 03/01/2017] [Accepted: 03/01/2017] [Indexed: 12/11/2022]
Abstract
Our previous study indicated that Casein kinase 2 interacting protein-1 (CKIP-1) could promote the activation of the nuclear factor E2-related factor 2 (Nrf2)/ antioxidant response element (ARE) pathway, playing a significant role in inhibiting the fibrosis of diabetic nephropathy (DN). Polydatin (PD) has been shown to possess strong resistance effects on renal fibrosis which is closely related to activating the Nrf2/ARE pathway, too. Whereas, whether PD could resist DN through regulating CKIP-1 and consequently promoting the activation of Nrf2-ARE pathway needs further investigation. Here, we found that PD significantly reversed the down-regulation of CKIP-1 and attenuated fibronectin (FN) and intercellular cell adhesion molecule-1 (ICAM-1) in glomerular mesangial cells (GMCs) exposed to high glucose (HG). Moreover, PD could decrease Keap1 expression and promote the nuclear content, ARE-binding ability, and transcriptional activity of Nrf2. The activation of Nrf2-ARE pathway by PD eventually led to the quenching of hydrogen peroxide (H2O2) and superoxide overproduction boosted by HG. Depletion of CKIP-1 blocked the Nrf2-ARE pathway activation and reversed FN and ICAM-1 down-regulation induced by PD in GMCs challenged with HG. PD increased CKIP-1 and Nrf2 levels in the kidney tissues as well as improved the anti-oxidative effect and renal dysfunction of diabetic mice, which eventually reversed the up-regulation of FN and ICAM-1. Experiments above suggested that PD could increase the CKIP-1-Nrf2-ARE pathway activation to prevent the OSS-induced insult in GMCs and diabetic mice which effectively postpone the diabetic renal fibrosis and the up-regulation of CKIP-1 is probably a novel mechanism in this process.
Collapse
Affiliation(s)
- Wenyan Gong
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jie Li
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhiquan Chen
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Junying Huang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Qiuhong Chen
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Weibin Cai
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Sun Yat-sen University, Guangzhou 510006, China
| | - Peiqing Liu
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Heqing Huang
- Laboratory of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Guangdong Engineering & Technology Research Center for Disease-Model Animals, Sun Yat-sen University, Guangzhou 510006, China; National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
15
|
Wagner PJ, Park HR, Wang Z, Kirchner R, Wei Y, Su L, Stanfield K, Guilarte TR, Wright RO, Christiani DC, Lu Q. In Vitro Effects of Lead on Gene Expression in Neural Stem Cells and Associations between Up-regulated Genes and Cognitive Scores in Children. ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:721-729. [PMID: 27562236 PMCID: PMC5381979 DOI: 10.1289/ehp265] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 07/26/2016] [Accepted: 07/26/2016] [Indexed: 05/21/2023]
Abstract
BACKGROUND Lead (Pb) adversely affects neurodevelopment in children. Neural stem cells (NSCs) play an essential role in shaping the developing brain, yet little is known about how Pb perturbs NSC functions and whether such perturbation contributes to impaired neurodevelopment. OBJECTIVES We aimed to identify Pb-induced transcriptomic changes in NSCs and to link these changes to neurodevelopmental outcomes in children who were exposed to Pb. METHODS We performed RNA-seq-based transcriptomic profiling in human NSCs treated with 1 μM Pb. We used qRT-PCR, Western blotting, ELISA, and ChIP (chromatin immunoprecipitation) to characterize Pb-induced gene up-regulation. Through interrogation of a genome-wide association study, we examined the association of gene variants with neurodevelopment outcomes in the ELEMENT birth cohort. RESULTS We identified 19 genes with significantly altered expression, including many known targets of NRF2-the master transcriptional factor for the oxidative stress response. Pb induced the expression of SPP1 (secreted phosphoprotein 1), which has known neuroprotective effects. We demonstrated that SPP1 is a novel direct NRF2 target gene. Single nucleotide polymorphisms (SNPs) (rs12641001) in the regulatory region of SPP1 exhibited a statistically significant association (p = 0.005) with the Cognitive Development Index (CDI). CONCLUSION Our findings revealed that Pb induces an NRF2-dependent transcriptional response in neural stem cells and identified SPP1 up-regulation as a potential novel mechanism linking Pb exposure with neural stem cell function and neurodevelopment in children.
Collapse
Affiliation(s)
- Peter J. Wagner
- Department of Environmental Health,
- Program in Molecular and Integrative Physiological Sciences, and
| | - Hae-Ryung Park
- Department of Environmental Health,
- Program in Molecular and Integrative Physiological Sciences, and
| | | | - Rory Kirchner
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | | | - Li Su
- Department of Environmental Health,
| | - Kirstie Stanfield
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York City, New York, USA
| | - Tomas R. Guilarte
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York City, New York, USA
| | - Robert O. Wright
- Department of Preventative Medicine, Mount Sinai School of Medicine, New York City, New York, USA
| | | | - Quan Lu
- Department of Environmental Health,
- Program in Molecular and Integrative Physiological Sciences, and
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Address correspondence to Q. Lu, Harvard School of Public Health, 665 Huntington Ave., Boston, MA 02215 USA. Telephone: (617) 432-7145. E-mail:
| |
Collapse
|
16
|
Glucose oxidase facilitates osteogenic differentiation and mineralization of embryonic stem cells through the activation of Nrf2 and ERK signal transduction pathways. Mol Cell Biochem 2016; 419:157-63. [DOI: 10.1007/s11010-016-2760-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 07/07/2016] [Indexed: 10/21/2022]
|