1
|
Mateos RN, Winardi W, Chiba K, Okada A, Suzuki A, Mitsuishi Y, Shiraishi Y. Splicing junction-based classifier for the detection of abnormal constitutive activation of the KEAP1-NRF2 system. NPJ Syst Biol Appl 2024; 10:147. [PMID: 39643653 PMCID: PMC11624210 DOI: 10.1038/s41540-024-00475-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024] Open
Abstract
The KEAP1-NRF2 system plays a crucial role in responding to oxidative and electrophilic stress. Its dysregulation can cause the overexpression of downstream genes, a known cancer hallmark. Understanding and detecting abnormal KEAP1-NRF2 activity is essential for understanding disease mechanisms and identifying therapeutic targets. This study presents an approach that analyzes splicing patterns by a naive Bayes-based classifier to identify constitutive activation of the KEAP1-NRF2 system, focusing on the higher presence of abnormal splicing junctions as a subproduct of overexpression of downstream genes. Our splicing-based classifier demonstrated robust performance, reliably identifying activation of the KEAP1-NRF2 pathway across extensive datasets, including The Cancer Genome Atlas and the Sequence Read Archive. This shows the classifier's potential to analyze hundreds of thousands of transcriptomes, highlighting its utility in broad-scale genomic studies and provides a new perspective on utilizing splicing aberrations caused by overexpression as diagnostic markers, offering potential improvements in diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Raúl N Mateos
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Wira Winardi
- Division of Respiratory Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Kenichi Chiba
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Ai Okada
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Ayako Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba, 277-8561, Japan
| | - Yoichiro Mitsuishi
- Division of Respiratory Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Yuichi Shiraishi
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| |
Collapse
|
2
|
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
|
3
|
Jin C, Yu XB, Yang J, Lin Z, Ma RX, Lin BH, Zhang HJ, Dai ZH, Xue K, Xie CL, Zheng W, Feng Y, Xiao J, Yang L. Corynoline Suppresses Osteoclastogenesis and Attenuates ROS Activities by Regulating NF-κB/MAPKs and Nrf2 Signaling Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:8149-8166. [PMID: 38551844 DOI: 10.1021/acs.jafc.3c07088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Declining estrogen production in postmenopausal females causes osteoporosis in which the resorption of bone exceeds the increase in bone formation. Although clinical drugs are currently available for the treatment of osteoporosis, sustained medication use is accompanied by serious side effects. Corydalis bungeana Herba, a famous traditional Chinese herb listed in the Chinese Pharmacopoeia Commission, constitutes various traditional Chinese Medicine prescriptions, which date back to thousands of years. One of the primary active components of C. bungeana Turcz. is Corynoline (Cor), a plant isoquinoline alkaloid derived from the Corydalis species, which possesses bone metabolism disease therapeutic potential. The study aimed at exploring the effects as well as mechanisms of Cor on osteoclast formation and bone resorption. TRAcP staining, F-actin belt formation, and pit formation were employed for assessing the osteoclast function. Western blot, qPCR, network pharmacology, and docking analyses were used for analyzing the expression of osteoclast-associated genes and related signaling pathways. The study focused on investigating how Cor affected OVX-induced trabecular bone loss by using a mouse model. Cor could weaken osteoclast formation and function by affecting the biological receptor activators of NF-κB and its ligand at various concentrations. Mechanistically, Cor inhibited the NF-κB activation, and the MAPKs pathway stimulated by RANKL. Besides, Cor enhanced the protein stability of the Nrf2, which effectively abolished the RANKL-stimulated ROS generation. According to an OVX mouse model, Cor functions in restoring bone mass, improving microarchitecture, and reducing the ROS levels in the distal femurs, which corroborated with its in vitro antiosteoclastogenic effect. The present study indicates that Cor may restrain osteoclast formation and bone loss by modulating NF-κB/MAPKs and Nrf2 signaling pathways. Cor was shown to be a potential drug candidate that can be utilized for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Chen Jin
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Xian-Bin Yu
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Jiayi Yang
- Department of Gynaecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Zhen Lin
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Run-Xun Ma
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Bing-Hao Lin
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Hao-Jie Zhang
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Zi-Han Dai
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Kaikai Xue
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
- Department of Burn and Wound Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Cheng-Long Xie
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Wenhao Zheng
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Yongzeng Feng
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| | - Jian Xiao
- Department of Burn and Wound Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Lei Yang
- Department of Orthopaedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, China
| |
Collapse
|
4
|
Saleh SR, Saleh OM, El-Bessoumy AA, Sheta E, Ghareeb DA, Eweda SM. The Therapeutic Potential of Two Egyptian Plant Extracts for Mitigating Dexamethasone-Induced Osteoporosis in Rats: Nrf2/HO-1 and RANK/RANKL/OPG Signals. Antioxidants (Basel) 2024; 13:66. [PMID: 38247490 PMCID: PMC10812806 DOI: 10.3390/antiox13010066] [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: 11/21/2023] [Revised: 12/25/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
The prolonged use of exogenous glucocorticoids, such as dexamethasone (Dex), is the most prevalent secondary cause of osteoporosis, known as glucocorticoid-induced osteoporosis (GIO). The current study examined the preventative and synergistic effect of aqueous chicory extract (ACE) and ethanolic purslane extract (EPE) on GIO compared with Alendronate (ALN). The phytochemical contents, elemental analysis, antioxidant scavenging activity, and ACE and EPE combination index were evaluated. Rats were randomly divided into control, ACE, EPE, and ACE/EPE MIX groups (100 mg/kg orally), Dex group (received 1.5 mg Dex/kg, Sc), and four treated groups received ACE, EPE, ACE/EPE MIX, and ALN with Dex. The bone mineral density and content, bone index, growth, turnover, and oxidative stress were measured. The molecular analysis of RANK/RANKL/OPG and Nrf2/HO-1 pathways were also evaluated. Dex causes osteoporosis by increasing oxidative stress, decreasing antioxidant markers, reducing bone growth markers (OPG and OCN), and increasing bone turnover and resorption markers (NFATc1, RANKL, ACP, ALP, IL-6, and TNF-α). In contrast, ACE, EPE, and ACE/EPE MIX showed a prophylactic effect against Dex-induced osteoporosis by modulating the measured parameters and the histopathological architecture. In conclusion, ACE/EPE MIX exerts a powerful synergistic effect against GIO by a mode of action different from ALN.
Collapse
Affiliation(s)
- Samar R. Saleh
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21515, Egypt; (O.M.S.); (A.A.E.-B.); (D.A.G.); (S.M.E.)
- Bio-Screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21515, Egypt
| | - Omnia M. Saleh
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21515, Egypt; (O.M.S.); (A.A.E.-B.); (D.A.G.); (S.M.E.)
- Bio-Screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21515, Egypt
| | - Ashraf A. El-Bessoumy
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21515, Egypt; (O.M.S.); (A.A.E.-B.); (D.A.G.); (S.M.E.)
| | - Eman Sheta
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria 21515, Egypt;
| | - Doaa A. Ghareeb
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21515, Egypt; (O.M.S.); (A.A.E.-B.); (D.A.G.); (S.M.E.)
- Bio-Screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21515, Egypt
| | - Saber M. Eweda
- Biochemistry Department, Faculty of Science, Alexandria University, Alexandria 21515, Egypt; (O.M.S.); (A.A.E.-B.); (D.A.G.); (S.M.E.)
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taibah University, Madinah 42353, Saudi Arabia
| |
Collapse
|
5
|
Yiang GT, Su WL, Zheng CM, Liao MT, Cheng TH, Lu CL, Lu KC. The influence of uremic toxins on low bone turnover disease in chronic kidney disease. Tzu Chi Med J 2024; 36:38-45. [PMID: 38406573 PMCID: PMC10887346 DOI: 10.4103/tcmj.tcmj_212_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/11/2023] [Accepted: 09/28/2023] [Indexed: 02/27/2024] Open
Abstract
Uremic toxins play a crucial role in the development of low bone turnover disease in chronic kidney disease (CKD) through the induction of oxidative stress. This oxidative stress disrupts the delicate balance between bone formation and resorption, resulting in a decline in both bone quantity and quality. Reactive oxygen species (ROS) activate nuclear factor kappa-B and mitogen-activated protein kinase signaling pathways, promoting osteoclastogenesis. Conversely, ROS hinder osteoblast differentiation by facilitating the binding of Forkhead box O proteins (FoxOs) to β-catenin, triggering apoptosis through FoxOs-activating kinase phosphorylation. This results in increased osteoblastic receptor activator of nuclear factor kappa-B ligand (RANKL) expression and decreased nuclear factor erythroid 2-related factor 2 levels, compromising antioxidant defenses against oxidative damage. As CKD progresses, the accumulation of protein-bound uremic toxins such as indoxyl sulfate (IS) and p-cresyl sulfate (PCS) intensifies oxidative stress, primarily affecting osteoblasts. IS and PCS directly inhibit osteoblast viability, induce apoptosis, decrease alkaline phosphatase activity, and impair collagen 1 and osteonectin, impeding bone formation. They also reduce cyclic adenosine 3',5'-monophosphate (cAMP) production and lower parathyroid hormone (PTH) receptor expression in osteoblasts, resulting in PTH hyporesponsiveness. In summary, excessive production of ROS by uremic toxins not only reduces the number and function of osteoblasts but also induces PTH hyporesponsiveness, contributing to the initiation and progression of low bone turnover disease in CKD.
Collapse
Affiliation(s)
- Giou-Teng Yiang
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Wen-Lin Su
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| | - Cai-Mei Zheng
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Shuang Ho Hospital, New Taipei, Taiwan
- Taipei Medical University-Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Tong-Hong Cheng
- Department of Internal Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Chien-Lin Lu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
- Division of Nephrology, Department of Medicine, Fu Jen Catholic University Hospital, New Taipei, Taiwan
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei, Taiwan
| |
Collapse
|
6
|
Zhang J, Zhang L, Yao G, Zhao H, Wu S. NRF2 is essential for iron-overload stimulated osteoclast differentiation through regulation of redox and iron homeostasis. Cell Biol Toxicol 2023; 39:3305-3321. [PMID: 37855941 DOI: 10.1007/s10565-023-09834-5] [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: 06/08/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023]
Abstract
Iron overload enhances osteoclastic bone resorption and induces osteoporosis. Excess iron is highly toxic. The modulation of redox and iron homeostasis is critical for osteoclast differentiation under iron-overload condition. Nuclear factor erythroid 2-related factor 2 (NRF2) is a transcription factor that regulates the cellular defense against oxidative stress and iron overload through the expression of genes involved in anti-oxidative processes and iron metabolism. Our studies demonstrated that NRF2 activation was suppressed during osteoclast differentiation. Under iron-overload condition, NRF2 and its mediated antioxidant and iron metabolism genes were activated by reactive oxygen species (ROS), which enhanced antioxidant capability. NRF2 mediated the upregulation of iron exporter ferroportin 1 (FPN1) and iron storage protein ferritin, contributing to decreased levels of intracellular iron. Nfe2l2 knockout induced oxidative stress and promoted osteoclast differentiation under normal condition, but induced ferroptosis under iron-overload condition. Nfe2l2 knockout alleviated iron overload induced bone loss by inhibiting osteoclast differentiation. Our results suggest that NRF2 activation is essential for osteoclast differentiation by enhancing antioxidant capability and reducing intracellular iron under iron-overload condition. Targeting NRF2 to induce ferroptosis could be a potential therapy for the treatment of iron-overload induced osteoporosis.
Collapse
Affiliation(s)
- Jian Zhang
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Huaxi University Town, Guiyang, 550025, Guizhou, China.
| | - Lingyan Zhang
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Huaxi University Town, Guiyang, 550025, Guizhou, China
| | - Gang Yao
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Huaxi University Town, Guiyang, 550025, Guizhou, China
| | - Hai Zhao
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Huaxi University Town, Guiyang, 550025, Guizhou, China
| | - Shuguang Wu
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Huaxi University Town, Guiyang, 550025, Guizhou, China
| |
Collapse
|
7
|
Fu F, Luo H, Du Y, Chen Y, Tian K, Pan J, Li J, Wang N, Bao R, Jin H, Tong P, Ruan H, Wu C. AR/PCC herb pair inhibits osteoblast pyroptosis to alleviate diabetes-related osteoporosis by activating Nrf2/Keap1 pathway. J Cell Mol Med 2023; 27:3601-3613. [PMID: 37621124 PMCID: PMC10660633 DOI: 10.1111/jcmm.17928] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/28/2023] [Accepted: 08/04/2023] [Indexed: 08/26/2023] Open
Abstract
Osteoporosis is a prevalent complication of diabetes, characterized by systemic metabolic impairment of bone mass and microarchitecture, particularly in the spine. Anemarrhenae Rhizoma/Phellodendri Chinensis Cortex (AR/PCC) herb pair has been extensively employed in Traditional Chinese Medicine to manage diabetes; however, its potential to ameliorate diabetic osteoporosis (DOP) has remained obscure. Herein, we explored the protective efficacy of AR/PCC herb pair against DOP using a streptozotocin (STZ)-induced rat diabetic model. Our data showed that AR/PCC could effectively reduce the elevated fasting blood glucose and reverse the osteoporotic phenotype of diabetic rats, resulting in significant improvements in vertebral trabecular area percentage, trabecular thickness and trabecular number, while reducing trabecular separation. Specifically, AR/PCC herb pair improved impaired osteogenesis, nerve ingrowth and angiogenesis. More importantly, it could mitigate the aberrant activation of osteoblast pyroptosis in the vertebral bodies of diabetic rats by reducing increased expressions of Nlrp3, Asc, Caspase1, Gsdmd and IL-1β. Mechanistically, AR/PCC activated antioxidant pathway through the upregulation of the antioxidant response protein Nrf2, while concurrently decreasing its negative feedback regulator Keap1. Collectively, our in vivo findings demonstrate that AR/PCC can inhibit osteoblast pyroptosis and alleviate STZ-induced rat DOP, suggesting its potential as a therapeutic agent for mitigating DOP.
Collapse
Affiliation(s)
- Fangda Fu
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Huan Luo
- Department of Pharmacy, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Yu Du
- The First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Yuying Chen
- The Fourth Clinical Medical College of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Kun Tian
- Department of OrthopaedicsThe First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Jin Pan
- Department of Architecture, School of ArchitectureChina Academy of ArtHangzhouChina
| | - Jian Li
- Department of OrthopaedicsHangzhou Ninth People's HospitalHangzhouChina
| | - Nani Wang
- Department of MedicineZhejiang Academy of Traditional Chinese MedicineHangzhouChina
| | - Ronghua Bao
- Hangzhou Fuyang Hospital of TCM Orthopedics and TraumatologyHangzhouChina
| | - Hongting Jin
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Peijian Tong
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Hongfeng Ruan
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| | - Chengliang Wu
- Institute of Orthopaedics and TraumatologyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine)HangzhouChina
| |
Collapse
|
8
|
Awad K, Ahuja N, Yacoub AS, Brotto L, Young S, Mikos A, Aswath P, Varanasi V. Revolutionizing bone regeneration: advanced biomaterials for healing compromised bone defects. FRONTIERS IN AGING 2023; 4:1217054. [PMID: 37520216 PMCID: PMC10376722 DOI: 10.3389/fragi.2023.1217054] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/06/2023] [Indexed: 08/01/2023]
Abstract
In this review, we explore the application of novel biomaterial-based therapies specifically targeted towards craniofacial bone defects. The repair and regeneration of critical sized bone defects in the craniofacial region requires the use of bioactive materials to stabilize and expedite the healing process. However, the existing clinical approaches face challenges in effectively treating complex craniofacial bone defects, including issues such as oxidative stress, inflammation, and soft tissue loss. Given that a significant portion of individuals affected by traumatic bone defects in the craniofacial area belong to the aging population, there is an urgent need for innovative biomaterials to address the declining rate of new bone formation associated with age-related changes in the skeletal system. This article emphasizes the importance of semiconductor industry-derived materials as a potential solution to combat oxidative stress and address the challenges associated with aging bone. Furthermore, we discuss various material and autologous treatment approaches, as well as in vitro and in vivo models used to investigate new therapeutic strategies in the context of craniofacial bone repair. By focusing on these aspects, we aim to shed light on the potential of advanced biomaterials to overcome the limitations of current treatments and pave the way for more effective and efficient therapeutic interventions for craniofacial bone defects.
Collapse
Affiliation(s)
- Kamal Awad
- Bone Muscle Research Center, College of Nursing and Health Innovations, University of Texas at Arlington, Arlington, TX, United States
- Department of Materials Science and Engineering, College of Engineering, The University of Texas at Arlington, Arlington, TX, United States
| | - Neelam Ahuja
- Bone Muscle Research Center, College of Nursing and Health Innovations, University of Texas at Arlington, Arlington, TX, United States
| | - Ahmed S. Yacoub
- Bone Muscle Research Center, College of Nursing and Health Innovations, University of Texas at Arlington, Arlington, TX, United States
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt
| | - Leticia Brotto
- Bone Muscle Research Center, College of Nursing and Health Innovations, University of Texas at Arlington, Arlington, TX, United States
| | - Simon Young
- Katz Department of Oral and Maxillofacial Surgery, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Antonios Mikos
- Center for Engineering Complex Tissues, Center for Excellence in Tissue Engineering, J.W. Cox Laboratory for Biomedical Engineering, Rice University, Houston, TX, United States
| | - Pranesh Aswath
- Department of Materials Science and Engineering, College of Engineering, The University of Texas at Arlington, Arlington, TX, United States
| | - Venu Varanasi
- Bone Muscle Research Center, College of Nursing and Health Innovations, University of Texas at Arlington, Arlington, TX, United States
- Department of Materials Science and Engineering, College of Engineering, The University of Texas at Arlington, Arlington, TX, United States
| |
Collapse
|
9
|
Dương TB, Dwivedi R, Bain LJ. 2,4-di-tert-butylphenol exposure impairs osteogenic differentiation. Toxicol Appl Pharmacol 2023; 461:116386. [PMID: 36682590 PMCID: PMC9974311 DOI: 10.1016/j.taap.2023.116386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023]
Abstract
2,4-di-tert-butylphenol (2,4-DTBP) is a synthetic antioxidant used in polyethylene crosspolymer (PEX) water distribution pipes and food-related plastics. 2,4-DTBP can leach from plastic materials and has been found in breast milk, cord blood, and placental tissue, giving rise to the concern that this compound may interfere with fetal development. The objective of this study is to assess the impacts of 2,4-DTBP on cellular differentiation. Human induced pluripotent stem (HiPS) cells were differentiated into osteoblasts or myoblasts over 40 days, and analyzed for markers of somite, dermomyotome, sclerotome, myoblast, and osteoblast development. When cultured as stem cells, 2,4-DTBP did not alter cell viability and expression of markers (NANOG, OCT4). However, upon differentiation into somite-like cells, 2,4-DTBP had reduced levels of MEOX1 and TBX6 transcripts, while NANOG and OCT4 were in turn upregulated in a dose-dependent manner. At the sclerotome-like stage, PAX9 mRNA decreased by 2-fold in the 0.5 μM and 1.0 μM 2,4-DTBP exposure groups. After 40 days of differentiation into an osteoblast-like lineage, exposure to 2,4-DTBP significantly reduced expression of the osteogenesis transcripts RUNX2 and OSX in a dose-dependent manner. Further, Alizarin Red staining of calcium deposits was decreased in the 0.5 μM and 1.0 μM treatment groups. In contrast, myogenesis was not affected by 2,4-DTBP exposure. Interestingly, KEAP1 expression was significantly increased in the sclerotomal-like cells, but decreased in the dermomytomal-like cells, which may suggest a mechanism of action. Overall, this study shows that 2,4-DTBP can delay key processes during sclerotome and osteoblast development, leading to a potential for bone developmental issues in exposed individuals.
Collapse
Affiliation(s)
- Thanh-Bình Dương
- Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, SC 29634, USA
| | - Raj Dwivedi
- Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, SC 29634, USA
| | - Lisa J Bain
- Department of Biological Sciences, Clemson University, 132 Long Hall, Clemson, SC 29634, USA.
| |
Collapse
|
10
|
Nichenametla SN, Mattocks DAL, Cooke D, Midya V, Malloy VL, Mansilla W, Øvrebø B, Turner C, Bastani N, Sokolová J, Pavlíková M, Richie JP, Shoveller A, Refsum H, Olsen T, Vinknes KJ, Kožich V, Ables GP. Cysteine restriction-specific effects of sulfur amino acid restriction on lipid metabolism. Aging Cell 2022; 21:e13739. [PMID: 36403077 PMCID: PMC9741510 DOI: 10.1111/acel.13739] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 10/12/2022] [Accepted: 10/21/2022] [Indexed: 11/21/2022] Open
Abstract
Decreasing the dietary intake of methionine exerts robust anti-adiposity effects in rodents but modest effects in humans. Since cysteine can be synthesized from methionine, animal diets are formulated by decreasing methionine and eliminating cysteine. Such diets exert both methionine restriction (MR) and cysteine restriction (CR), that is, sulfur amino acid restriction (SAAR). Contrarily, SAAR diets formulated for human consumption included cysteine, and thus might have exerted only MR. Epidemiological studies positively correlate body adiposity with plasma cysteine but not methionine, suggesting that CR, but not MR, is responsible for the anti-adiposity effects of SAAR. Whether this is true, and, if so, the underlying mechanisms are unknown. Using methionine- and cysteine-titrated diets, we demonstrate that the anti-adiposity effects of SAAR are due to CR. Data indicate that CR increases serinogenesis (serine biosynthesis from non-glucose substrates) by diverting substrates from glyceroneogenesis, which is essential for fatty acid reesterification and triglyceride synthesis. Molecular data suggest that CR depletes hepatic glutathione and induces Nrf2 and its downstream targets Phgdh (the serine biosynthetic enzyme) and Pepck-M. In mice, the magnitude of SAAR-induced changes in molecular markers depended on dietary fat concentration (60% fat >10% fat), sex (males > females), and age-at-onset (young > adult). Our findings are translationally relevant as we found negative and positive correlations of plasma serine and cysteine, respectively, with triglycerides and metabolic syndrome criteria in a cross-sectional epidemiological study. Controlled feeding of low-SAA, high-polyunsaturated fatty acid diets increased plasma serine in humans. Serinogenesis might be a target for treating hypertriglyceridemia.
Collapse
Affiliation(s)
- Sailendra N. Nichenametla
- Animal Science LaboratoryOrentreich Foundation for the Advancement of ScienceCold Spring‐on‐HudsonNew YorkUSA
| | - Dwight A. L. Mattocks
- Animal Science LaboratoryOrentreich Foundation for the Advancement of ScienceCold Spring‐on‐HudsonNew YorkUSA
| | - Diana Cooke
- Animal Science LaboratoryOrentreich Foundation for the Advancement of ScienceCold Spring‐on‐HudsonNew YorkUSA
| | - Vishal Midya
- Department of Environmental Medicine and Public HealthIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Virginia L. Malloy
- Animal Science LaboratoryOrentreich Foundation for the Advancement of ScienceCold Spring‐on‐HudsonNew YorkUSA
| | - Wilfredo Mansilla
- Department of Animal BioscienceUniversity of GuelphGuelphOntarioCanada
| | - Bente Øvrebø
- Department of Nutrition, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Cheryl Turner
- Department of PharmacologyUniversity of OxfordOxfordUK
| | - Nasser E. Bastani
- Department of Nutrition, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Jitka Sokolová
- Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital in PragueCharles University‐First Faculty of MedicinePragueCzech Republic
| | - Markéta Pavlíková
- Department of Probability and Mathematical StatisticsCharles University ‐ Faculty of Mathematics and PhysicsPragueCzech Republic
| | - John P. Richie
- Departments of Public Health Sciences and PharmacologyPenn State University College of MedicineHersheyPennsylvaniaUSA
| | - Anna K. Shoveller
- Department of Animal BioscienceUniversity of GuelphGuelphOntarioCanada
| | - Helga Refsum
- Department of Nutrition, Institute of Basic Medical SciencesUniversity of OsloOsloNorway,Department of PharmacologyUniversity of OxfordOxfordUK
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Kathrine J. Vinknes
- Department of Nutrition, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Viktor Kožich
- Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital in PragueCharles University‐First Faculty of MedicinePragueCzech Republic
| | - Gene P. Ables
- Animal Science LaboratoryOrentreich Foundation for the Advancement of ScienceCold Spring‐on‐HudsonNew YorkUSA
| |
Collapse
|
11
|
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: 0] [Impact Index Per Article: 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
|
12
|
Okagu IU, Ezeorba TPC, Aguchem RN, Ohanenye IC, Aham EC, Okafor SN, Bollati C, Lammi C. A Review on the Molecular Mechanisms of Action of Natural Products in Preventing Bone Diseases. Int J Mol Sci 2022; 23:ijms23158468. [PMID: 35955603 PMCID: PMC9368769 DOI: 10.3390/ijms23158468] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
The drugs used for treating bone diseases (BDs), at present, elicit hazardous side effects that include certain types of cancers and strokes, hence the ongoing quest for the discovery of alternatives with little or no side effects. Natural products (NPs), mainly of plant origin, have shown compelling promise in the treatments of BDs, with little or no side effects. However, the paucity in knowledge of the mechanisms behind their activities on bone remodeling has remained a hindrance to NPs’ adoption. This review discusses the pathological development of some BDs, the NP-targeted components, and the actions exerted on bone remodeling signaling pathways (e.g., Receptor Activator of Nuclear Factor κ B-ligand (RANKL)/monocyte/macrophage colony-stimulating factor (M-CSF)/osteoprotegerin (OPG), mitogen-activated protein kinase (MAPK)s/c-Jun N-terminal kinase (JNK)/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), Kelch-like ECH-associated protein 1 (Keap-1)/nuclear factor erythroid 2–related factor 2 (Nrf2)/Heme Oxygenase-1 (HO-1), Bone Morphogenetic Protein 2 (BMP2)-Wnt/β-catenin, PhosphatidylInositol 3-Kinase (PI3K)/protein kinase B (Akt)/Glycogen Synthase Kinase 3 Beta (GSK3β), and other signaling pathways). Although majority of the studies on the osteoprotective properties of NPs against BDs were conducted ex vivo and mostly on animals, the use of NPs for treating human BDs and the prospects for future development remain promising.
Collapse
Affiliation(s)
- Innocent U. Okagu
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001, Nigeria; (I.U.O.); (T.P.C.E.); (R.N.A.); (E.C.A.)
| | - Timothy P. C. Ezeorba
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001, Nigeria; (I.U.O.); (T.P.C.E.); (R.N.A.); (E.C.A.)
| | - Rita N. Aguchem
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001, Nigeria; (I.U.O.); (T.P.C.E.); (R.N.A.); (E.C.A.)
| | - Ikenna C. Ohanenye
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8M5, Canada;
| | - Emmanuel C. Aham
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka 410001, Nigeria; (I.U.O.); (T.P.C.E.); (R.N.A.); (E.C.A.)
- Natural Science Unit, School of General Studies, University of Nigeria, Nsukka 410001, Nigeria
- School of Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Sunday N. Okafor
- Department of Pharmaceutical and Medicinal Chemistry, University of Nigeria, Nsukka 410001, Nigeria;
| | - Carlotta Bollati
- Department of Pharmaceutical Sciences, University of Milan, via Mangiagalli 25, 20133 Milano, Italy;
| | - Carmen Lammi
- Department of Pharmaceutical Sciences, University of Milan, via Mangiagalli 25, 20133 Milano, Italy;
- Correspondence: ; Tel.: +39-02-5031-9372
| |
Collapse
|
13
|
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
|
14
|
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: 28] [Impact Index Per Article: 9.3] [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
|
15
|
Sun Y, Li J, Xie X, Gu F, Sui Z, Zhang K, Yu T. Recent Advances in Osteoclast Biological Behavior. Front Cell Dev Biol 2021; 9:788680. [PMID: 34957116 PMCID: PMC8694526 DOI: 10.3389/fcell.2021.788680] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/15/2021] [Indexed: 12/19/2022] Open
Abstract
With the progress of the aging population, bone-related diseases such as osteoporosis and osteoarthritis have become urgent problems. Recent studies have demonstrated the importance of osteoclasts in bone homeostasis, implying these will be an important mediator in the treatment of bone-related diseases. Up to now, several reviews have been performed on part of osteoclast biological behaviors such as differentiation, function, or apoptosis. However, few reviews have shown the complete osteoclast biology and research advances in recent years. Therefore, in this review, we focus on the origin, differentiation, apoptosis, behavior changes and coupling signals with osteoblasts, providing a simple but comprehensive overview of osteoclasts for subsequent studies.
Collapse
Affiliation(s)
- Yang Sun
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Jiangbi Li
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Xiaoping Xie
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Feng Gu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Zhenjiang Sui
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Ke Zhang
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Tiecheng Yu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
16
|
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
|
17
|
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: 12] [Impact Index Per Article: 3.0] [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
|
18
|
Sakai E, Sato M, Memtily N, Tsukuba T, Sato C. Liquid-phase ASEM imaging of cellular and structural details in cartilage and bone formed during endochondral ossification: Keap1-deficient osteomalacia. Sci Rep 2021; 11:5722. [PMID: 33707458 PMCID: PMC7952587 DOI: 10.1038/s41598-021-84202-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 02/03/2021] [Indexed: 11/09/2022] Open
Abstract
Chondrogenesis and angiogenesis drive endochondral ossification. Using the atmospheric scanning electron microscopy (ASEM) without decalcification and dehydration, we directly imaged angiogenesis-driven ossification at different developmental stages shortly after aldehyde fixation, using aqueous radical scavenger glucose solution to preserve water-rich structures. An embryonic day 15.5 mouse femur was fixed and stained with phosphotungstic acid (PTA), and blood vessel penetration into the hypertrophic chondrocyte zone was visualised. We observed a novel envelope between the perichondrium and proliferating chondrocytes, which was lined with spindle-shaped cells that could be borderline chondrocytes. At postnatal day (P)1, trabecular and cortical bone mineralisation was imaged without staining. Additional PTA staining visualised surrounding soft tissues; filamentous connections between osteoblast-like cells and osteocytes in cortical bone were interpreted as the osteocytic lacunar-canalicular system. By P10, resorption pits had formed on the tibial trabecular bone surface. The applicability of ASEM for pathological analysis was addressed using knockout mice of Keap1, an oxidative-stress sensor. In Keap1-/- femurs, we observed impaired calcification and angiogenesis of epiphyseal cartilage, suggesting impaired bone development. Overall, the quick ASEM method we developed revealed mineralisation and new structures in wet bone tissue at EM resolution and can be used to study mineralisation-associated phenomena of any hydrated tissue.
Collapse
Affiliation(s)
- Eiko Sakai
- Division of Dental Pharmacology, Department of Developmental and Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan.
| | - Mari Sato
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, Higashi 1-1-1, Tsukuba, Ibaraki, 305-8566, Japan
| | - Nassirhadjy Memtily
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, Higashi 1-1-1, Tsukuba, Ibaraki, 305-8566, Japan
- Traditional Uyghur Medicine Institute of Xinjiang Medical University, 393 Xinyi Rd, Urumqi, 830011, Xinjiang Uyghur Autonomous Region, China
| | - Takayuki Tsukuba
- Division of Dental Pharmacology, Department of Developmental and Reconstructive Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Chikara Sato
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, Higashi 1-1-1, Tsukuba, Ibaraki, 305-8566, Japan
| |
Collapse
|
19
|
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: 12] [Impact Index Per Article: 2.4] [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
|