1
|
Gong S, Tian A, Lang S, Wang Y, Ma J, Ma X. Paeonol regulates autophagy through the PI3K-AKT-mTOR signaling pathway to inhibit apoptosis of osteocytes. Eur J Pharmacol 2025; 995:177427. [PMID: 39988091 DOI: 10.1016/j.ejphar.2025.177427] [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: 01/16/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 02/25/2025]
Abstract
Osteoporosis is the most common complication of glucocorticoids and predisposes to fractures. Excessive apoptosis of osteocytes is the pathological feature of glucocorticoid-induced osteoporosis. Paeonol, an effective component of Traditional Chinese Medicine Cortex Moutan, known for its anti-inflammatory and analgesic properties, has a long clinical application history. However, the regulatory effect of paeonol on the fate of osteocytes under excessive glucocorticoid remains unclear. The present study aimed to investigate the effect of paeonol against osteocyte death and osteoporosis induced by glucocorticoid and to explore the underlying mechanisms. We found that paeonol not only improved the low proliferation rate of osteocytes induced by dexamethasone but also weakened the dexamethasone-induced apoptosis of osteocytes by stimulating cytoprotective autophagy. Subsequently, proteomic sequencing identified the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) - protein kinase B (AKT) signaling pathway as the potential target of paeonol in attenuating dexamethasone-induced osteocyte injury, and the PI3K activator and inhibitor confirmed this hypothesis. In vivo, paeonol alleviated glucocorticoid-induced osteoporosis, promoted autophagy and inhibited apoptosis of osteocytes by regulating PI3K phosphorylation. In brief, paeonol protects osteocytes from dexamethasone-derived apoptosis by increasing protective autophagy, further inhibiting osteoporosis. Its autophagy-promoting effect was associated with inhibition of PI3K-AKT-mechanistic target of rapamycin (mTOR) of osteocytes.
Collapse
Affiliation(s)
- Shuwei Gong
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Orthopedic Research Institute, Tianjin Hospital, Tianjin, 300050, China; Department of Orthopedics, Tianjin Hospital, Tianjin, 300211, China
| | - Aixian Tian
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Orthopedic Research Institute, Tianjin Hospital, Tianjin, 300050, China
| | - Shuang Lang
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Orthopedic Research Institute, Tianjin Hospital, Tianjin, 300050, China
| | - Yan Wang
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Orthopedic Research Institute, Tianjin Hospital, Tianjin, 300050, China
| | - Jianxiong Ma
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Orthopedic Research Institute, Tianjin Hospital, Tianjin, 300050, China; Department of Orthopedics, Tianjin Hospital, Tianjin, 300211, China.
| | - Xinlong Ma
- Tianjin Key Laboratory of Orthopedic Biomechanics and Medical Engineering, Orthopedic Research Institute, Tianjin Hospital, Tianjin, 300050, China; Department of Orthopedics, Tianjin Hospital, Tianjin, 300211, China.
| |
Collapse
|
2
|
Xiang XN, He HC, He CQ. Advances in mechanism and management of bone homeostasis in osteonecrosis: a review article from basic to clinical applications. Int J Surg 2025; 111:1101-1122. [PMID: 39311934 PMCID: PMC11745759 DOI: 10.1097/js9.0000000000002094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 09/15/2024] [Indexed: 01/23/2025]
Abstract
Osteonecrosis, characterized by bone cell death leading to impaired bone recovery, causes challenges in bone homeostasis maintenance. Bone homeostasis relies on the delicate balance between osteoclasts and osteoblasts, encompassing a series of complex and strictly regulated biological functions. Current treatments, including conservative therapies and surgeries, often fall short of expected outcomes, necessitating a reorientation towards more effective therapeutic strategies according to the pathogenesis. In this review, the authors hierarchically outlined risk factors, emerging mechanisms, and last-decade treatment approaches in osteonecrosis. By connecting mechanisms of bone homeostasis, the authors proposed future research directions should be focused on elucidating risk factors and key molecules, performing high-quality clinical trial, updating practice, and accelerating translational potential.
Collapse
Affiliation(s)
- Xiao-Na Xiang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, P. R. China
| | - Hong-Chen He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, P. R. China
| | - Cheng-Qi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University
- School of Rehabilitation Sciences, West China School of Medicine, Sichuan University
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, P. R. China
| |
Collapse
|
3
|
Li X, Tian F, Liu G, Liu X, Fu M, E Z, Wang C, Gao F. miR-129-5p targets HOXC10 to control BMSC adipogenesis and osteogenesis in a model of steroid-induced osteonecrosis of the femoral head. Am J Transl Res 2024; 16:7374-7384. [PMID: 39822543 PMCID: PMC11733387 DOI: 10.62347/chgj7909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 11/19/2024] [Indexed: 01/19/2025]
Abstract
BACKGROUND Steroid-induced osteonecrosis of the femoral head (SONFH) is a pathological condition primarily driven by an impaired balance in the differentiation of bone marrow mesenchymal stem cells (BMSCs) into adipogenic and osteogenic lineages. This study aimed to explore the role of miR-129-5p as a regulator of SONFH progression and associated mechanisms. METHODS BMSCs were harvested from a rat SONFH model. qPCR was leveraged to assess miR-129-5p levels, while both qPCR and Western immunoblotting were utilized to evaluate HOXC10 expression. CCK8 assay was used to measure the proliferative activity of BMSCs, while their differentiation was analyzed using qPCR and Western blot. RESULTS SONFH was associated with reduced miR-129-5p levels. Downregulation of miR-129-5p promoted BMSC adipogenesis while inhibiting their osteogenic differentiation and enhancing their adipogenesis differentiation. Mechanistically, miR-129-5p was found to regulate these processes by targeting the expression of the HOXC10 gene. CONCLUSIONS MiR-129-5p is downregulated in a rat SONFH model. Reduced miR-129-5p levels facilitated adipogenesis and suppressed osteogenesis in BMSCs through its inhibition of HOXC10. These findings offer novel insights into the prevention and treatment of SONFH.
Collapse
Affiliation(s)
- Xuezhao Li
- Department of Orthopedics, Heilongjiang Provincial HospitalHarbin 150036, Heilongjiang, China
| | - Fangqiu Tian
- Department of Orthopedics, Heilongjiang Provincial HospitalHarbin 150036, Heilongjiang, China
| | - Guohua Liu
- Department of Orthopedics, Heilongjiang Provincial HospitalHarbin 150036, Heilongjiang, China
| | - Xiang Liu
- Department of Orthopedics, Heilongjiang Provincial HospitalHarbin 150036, Heilongjiang, China
| | - Ming Fu
- Department of Orthopedics, Heilongjiang Provincial HospitalHarbin 150036, Heilongjiang, China
| | - Zhiyin E
- Department of Orthopedics, Heilongjiang Provincial HospitalHarbin 150036, Heilongjiang, China
| | - Cong Wang
- Department of Orthopedics, Heilongjiang Provincial HospitalHarbin 150036, Heilongjiang, China
| | - Fapeng Gao
- Department of Orthopedics, Huai’an Hospital of Huai’an CityHuai’an 223200, Jiangsu, China
| |
Collapse
|
4
|
Xu H, Wang L, Zhu X, Zhang H, Chen H, Zhang H. Jintiange capsule ameliorates glucocorticoid-induced osteonecrosis of the femoral head in rats by regulating the activity and differentiation of BMSCs. J Tradit Complement Med 2024; 14:568-580. [PMID: 39262662 PMCID: PMC11384076 DOI: 10.1016/j.jtcme.2024.03.013] [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: 09/12/2023] [Revised: 02/26/2024] [Accepted: 03/05/2024] [Indexed: 09/13/2024] Open
Abstract
Background and aim A surplus of glucocorticoids (GC) is a main cause of non-traumatic osteonecrosis of the femoral head (ONFH), and Jintiange (JTG), as one of the traditional Chinese medicines (TCM), also plays an instrumental role in the alleviation of bone loss simultaneously. Therefore, JTG was thought to be able to reverse GC-induced ONFH (GC-ONFH) to a certain extent. Experimental procedure In vivo, the effect of JTG on trabeculae in the subchondral bone of the femoral head was investigated using micro-computed tomography (micro-CT), TdT-mediated dUTP nick end labeling (TUNEL) and histological staining; in vitro, proliferation, viability, apoptosis, and senescence of purified bone mesenchymal stem cells (BMSCs) were examined to demonstrate the direct impact of JTG on these cells. Meanwhile after using a series of interventions, the function of JTG on BMSC differentiation could be assessed by measuring of osteogenic and adipogenic markers at levels of protein and mRNA. Results Our final results demonstrated that with the involvement of Wnt/β-catenin pathway, JTG was able to significantly promote osteogenesis, restrain adipogenesis, delay senescence in BMSCs, reduce osteoclast number, weaken apoptosis, and enhance proliferation of osteocytes, all of which could mitigate the progression of subchondral osteonecrosis. Conclusion According to the results of experiments in vitro and vivo, JTG was deemed to relieve the early GC-ONFH using the prevention of destruction of subchondral bone, which was contributed to regulating the differentiation of BMSCs and the number of osteoclasts.
Collapse
Affiliation(s)
- Hui Xu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lin Wang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xunpeng Zhu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Haigang Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hongwei Chen
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
5
|
Aravena-Canales D, Valenzuela-Muñoz V, Gallardo-Escarate C, Molina A, Valdés JA. Transcriptomic and Epigenomic Responses to Cortisol-Mediated Stress in Rainbow Trout ( Oncorhynchus mykiss) Skeletal Muscle. Int J Mol Sci 2024; 25:7586. [PMID: 39062828 PMCID: PMC11276852 DOI: 10.3390/ijms25147586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
The production and release of cortisol during stress responses are key regulators of growth in teleosts. Understanding the molecular responses to cortisol is crucial for the sustainable farming of rainbow trout (Oncorhynchus mykiss) and other salmonid species. While several studies have explored the genomic and non-genomic impacts of cortisol on fish growth and skeletal muscle development, the long-term effects driven by epigenetic mechanisms, such as cortisol-induced DNA methylation, remain unexplored. In this study, we analyzed the transcriptome and genome-wide DNA methylation in the skeletal muscle of rainbow trout seven days after cortisol administration. We identified 550 differentially expressed genes (DEGs) by RNA-seq and 9059 differentially methylated genes (DMGs) via whole-genome bisulfite sequencing (WGBS) analysis. KEGG enrichment analysis showed that cortisol modulates the differential expression of genes associated with nucleotide metabolism, ECM-receptor interaction, and the regulation of actin cytoskeleton pathways. Similarly, cortisol induced the differential methylation of genes associated with focal adhesion, adrenergic signaling in cardiomyocytes, and Wnt signaling. Through integrative analyses, we determined that 126 genes showed a negative correlation between up-regulated expression and down-regulated methylation. KEGG enrichment analysis of these genes indicated participation in ECM-receptor interaction, regulation of actin cytoskeleton, and focal adhesion. Using RT-qPCR, we confirmed the differential expression of lamb3, itga6, limk2, itgb4, capn2, and thbs1. This study revealed for the first time the molecular responses of skeletal muscle to cortisol at the transcriptomic and whole-genome DNA methylation levels in rainbow trout.
Collapse
Affiliation(s)
- Daniela Aravena-Canales
- Laboratorio de Biotecnología Molecular, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago 8370035, Chile; (D.A.-C.); (A.M.)
- Interdisciplinary Center for Aquaculture Research (INCAR), University of Concepción, Concepcion 4030000, Chile; (V.V.-M.); (C.G.-E.)
| | - Valentina Valenzuela-Muñoz
- Interdisciplinary Center for Aquaculture Research (INCAR), University of Concepción, Concepcion 4030000, Chile; (V.V.-M.); (C.G.-E.)
- Escuela de Medicina Veterinaria, Facultad de Ciencias de la Naturaleza, Universidad San Sebastián, Concepción 4030000, Chile
- Laboratory of Biotechnology and Aquatic Genomics, Department of Oceanography, University of Concepción, Concepcion 4030000, Chile
| | - Cristian Gallardo-Escarate
- Interdisciplinary Center for Aquaculture Research (INCAR), University of Concepción, Concepcion 4030000, Chile; (V.V.-M.); (C.G.-E.)
- Laboratory of Biotechnology and Aquatic Genomics, Department of Oceanography, University of Concepción, Concepcion 4030000, Chile
| | - Alfredo Molina
- Laboratorio de Biotecnología Molecular, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago 8370035, Chile; (D.A.-C.); (A.M.)
- Interdisciplinary Center for Aquaculture Research (INCAR), University of Concepción, Concepcion 4030000, Chile; (V.V.-M.); (C.G.-E.)
- Centro de Investigación Marina Quintay (CIMARQ), Universidad Andres Bello, Quintay 2340000, Chile
| | - Juan Antonio Valdés
- Laboratorio de Biotecnología Molecular, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago 8370035, Chile; (D.A.-C.); (A.M.)
- Interdisciplinary Center for Aquaculture Research (INCAR), University of Concepción, Concepcion 4030000, Chile; (V.V.-M.); (C.G.-E.)
- Centro de Investigación Marina Quintay (CIMARQ), Universidad Andres Bello, Quintay 2340000, Chile
| |
Collapse
|
6
|
Paes T, Feelders RA, Hofland LJ. Epigenetic Mechanisms Modulated by Glucocorticoids With a Focus on Cushing Syndrome. J Clin Endocrinol Metab 2024; 109:e1424-e1433. [PMID: 38517306 PMCID: PMC11099489 DOI: 10.1210/clinem/dgae151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 02/08/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024]
Abstract
In Cushing syndrome (CS), prolonged exposure to high cortisol levels results in a wide range of devastating effects causing multisystem morbidity. Despite the efficacy of treatment leading to disease remission and clinical improvement, hypercortisolism-induced complications may persist. Since glucocorticoids use the epigenetic machinery as a mechanism of action to modulate gene expression, the persistence of some comorbidities may be mediated by hypercortisolism-induced long-lasting epigenetic changes. Additionally, glucocorticoids influence microRNA expression, which is an important epigenetic regulator as it modulates gene expression without changing the DNA sequence. Evidence suggests that chronically elevated glucocorticoid levels may induce aberrant microRNA expression which may impact several cellular processes resulting in cardiometabolic disorders. The present article reviews the evidence on epigenetic changes induced by (long-term) glucocorticoid exposure. Key aspects of some glucocorticoid-target genes and their implications in the context of CS are described. Lastly, the effects of epigenetic drugs influencing glucocorticoid effects are discussed for their ability to be potentially used as adjunctive therapy in CS.
Collapse
Affiliation(s)
- Ticiana Paes
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston 02115, MA, USA
| | - Richard A Feelders
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Leo J Hofland
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
7
|
Yan Y, Yan H, Qiu Z, Long L. Mechanism of KDM5A-mediated H3K4me3 modification in the osteogenic differentiation of mesenchymal stem cells in steroid-induced osteonecrosis of the femoral head. Int J Rheum Dis 2024; 27:e15090. [PMID: 38443978 DOI: 10.1111/1756-185x.15090] [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: 11/08/2023] [Revised: 01/26/2024] [Accepted: 02/06/2024] [Indexed: 03/07/2024]
Abstract
OBJECTIVES Steroid-induced osteonecrosis of the femoral head (SONFH) is characterized by impaired osteogenesis in bone marrow mesenchymal stem cells (BMSCs). This study investigates the role of lysine-specific demethylase 5A (KDM5A) in SONFH to identify potential therapeutic targets. METHODS Human BMSCs were isolated and characterized for cell surface markers and differentiation capacity. A SONFH cell model was established using dexamethasone treatment. BMSCs were transfected with KDM5A overexpression vectors or si-KDM5A, and the expression of KDM5A, miR-107, runt-related transcription factor 2 (RUNX2), osteocalcin (OCN), and osteopontin (OPN) was assessed. Alizarin red staining was used to observe mineralization nodules, while alkaline phosphatase activity and cell viability were measured. The enrichment of KDM5A and histone 3 lysine 4 trimethylation (H3K4me3) on the promoters of RUNX2, OCN, and OPN was analyzed. The binding between miR-107 and KDM5A 3'UTR was validated, and the combined effect of miR-107 overexpression and KDM5A overexpression on BMSC osteogenic differentiation was evaluated. RESULTS KDM5A was upregulated in BMSCs from SONFH. Inhibition of KDM5A promoted osteogenic differentiation of BMSCs, associated with increased RUNX2, OCN, and OPN promoters. KDM5A bound to the promoters of RUNX2, OCN, and OPN, leading to reduced H3K4me3 levels and downregulation of their expression. Overexpression of miR-107 inhibited KDM5A and enhanced BMSC osteogenic differentiation. CONCLUSION KDM5A negatively regulates BMSC osteogenic differentiation by modulating H3K4me3 levels on the promoters of key osteogenic genes. miR-107 overexpression counteracts the inhibitory effect of KDM5A on osteogenic differentiation. These findings highlight the potential of targeting the KDM5A/miR-107 axis for SONFH therapy.
Collapse
Affiliation(s)
- Yi Yan
- Department of Orthopedics, Xiangtan Central Hospital, Xiangtan, China
| | - Hao Yan
- Department of Adult Emergency, Boai Hospital, Zhongshan, China
| | - Zhilong Qiu
- Department of Orthopedics, Xiangtan Central Hospital, Xiangtan, China
| | - Liang Long
- Department of Clinical Pharmacy, Xiangtan Central Hospital, Xiangtan, China
| |
Collapse
|
8
|
Nie J, Ma S, Zhang Y, Yu S, Yang J, Li A, Pei D. COPI Vesicle Disruption Inhibits Mineralization via mTORC1-Mediated Autophagy. Int J Mol Sci 2023; 25:339. [PMID: 38203512 PMCID: PMC10779376 DOI: 10.3390/ijms25010339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Bone mineralization is a sophisticated regulated process composed of crystalline calcium phosphate and collagen fibril. Autophagy, an evolutionarily conserved degradation system, whereby double-membrane vesicles deliver intracellular macromolecules and organelles to lysosomes for degradation, has recently been shown to play an essential role in mineralization. However, the formation of autophagosomes in mineralization remains to be determined. Here, we show that Coat Protein Complex I (COPI), responsible for Golgi-to-ER transport, plays a pivotal role in autophagosome formation in mineralization. COPI vesicles were increased after osteoinduction, and COPI vesicle disruption impaired osteogenesis. Mechanistically, COPI regulates autophagy activity via the mTOR complex 1 (mTORC1) pathway, a key regulator of autophagy. Inhibition of mTOR1 rescues the impaired osteogenesis by activating autophagy. Collectively, our study highlights the functional importance of COPI in mineralization and identifies COPI as a potential therapeutic target for treating bone-related diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dandan Pei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi’an Jiaotong University, Xi’an 710004, China
| |
Collapse
|
9
|
Huang C, Qing L, Xiao Y, Tang J, Wu P. Insight into Steroid-Induced ONFH: The Molecular Mechanism and Function of Epigenetic Modification in Mesenchymal Stem Cells. Biomolecules 2023; 14:4. [PMID: 38275745 PMCID: PMC10813482 DOI: 10.3390/biom14010004] [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: 09/19/2023] [Revised: 11/15/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a common refractory orthopedic disease, which is one of the common causes of hip pain and dysfunction. ONFH has a very high disability rate, which is associated with a heavy burden to patients, families, and society. The pathogenesis of ONFH is not completely clear. At present, it is believed that it mainly includes coagulation dysfunction, abnormal lipid metabolism, an imbalance of osteogenic/adipogenic differentiation, and poor vascularization repair. The prevention and treatment of ONFH has always been a great challenge for clinical orthopedic surgeons. However, recent studies have emphasized that the use of mesenchymal stem cells (MSCs) to treat steroid-induced ONFH (SONFH) is a promising therapy. This review focuses on the role and molecular mechanism of epigenetic regulation in the progress of MSCs in the treatment of SONFH, and discusses the significance of the latest research in the treatment of SONFH from the perspective of epigenetics.
Collapse
Affiliation(s)
| | | | | | - Juyu Tang
- Department of Orthopedics, Hand and Microsurgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital of Central South University, Changsha 410008, China; (C.H.); (L.Q.); (Y.X.)
| | - Panfeng Wu
- Department of Orthopedics, Hand and Microsurgery, National Clinical Research Center of Geriatric Disorders, Xiangya Hospital of Central South University, Changsha 410008, China; (C.H.); (L.Q.); (Y.X.)
| |
Collapse
|
10
|
Meng F, Yu Y, Tian Y, Deng M, Zheng K, Guo X, Zeng B, Li J, Qian A, Yin C. A potential therapeutic drug for osteoporosis: prospect for osteogenic LncRNAs. Front Endocrinol (Lausanne) 2023; 14:1219433. [PMID: 37600711 PMCID: PMC10435887 DOI: 10.3389/fendo.2023.1219433] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Long non-coding RNAs (LncRNAs) play essential roles in multiple physiological processes including bone formation. Investigators have revealed that LncRNAs regulated bone formation through various signaling pathways and micro RNAs (miRNAs). However, several problems exist in current research studies on osteogenic LncRNAs, including sophisticated techniques, high cost for in vivo experiment, as well as low homology of LncRNAs between animal model and human, which hindered translational medicine research. Moreover, compared with gene editing, LncRNAs would only lead to inhibition of target genes rather than completely knocking them out. As the studies on osteogenic LncRNA gradually proceed, some of these problems have turned osteogenic LncRNA research studies into slump. This review described some new techniques and innovative ideas to address these problems. Although investigations on osteogenic LncRNAs still have obtacles to overcome, LncRNA will work as a promising therapeutic drug for osteoporosis in the near future.
Collapse
Affiliation(s)
- Fanjin Meng
- Department of Clinical Laboratory, Department of Oncology, Department of Rehabilitation Medicine, Ministry of Science and Technology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, China
| | - Yang Yu
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Ye Tian
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Meng Deng
- Department of Clinical Laboratory, Department of Oncology, Department of Rehabilitation Medicine, Ministry of Science and Technology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, China
| | - Kaiyuan Zheng
- Department of Clinical Laboratory, Department of Oncology, Department of Rehabilitation Medicine, Ministry of Science and Technology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, China
| | - Xiaolan Guo
- Department of Clinical Laboratory, Department of Oncology, Department of Rehabilitation Medicine, Ministry of Science and Technology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, China
| | - Beilei Zeng
- Department of Clinical Laboratory, Department of Oncology, Department of Rehabilitation Medicine, Ministry of Science and Technology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jingjia Li
- Department of Clinical Laboratory, Department of Oncology, Department of Rehabilitation Medicine, Ministry of Science and Technology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Airong Qian
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Chong Yin
- Department of Clinical Laboratory, Department of Oncology, Department of Rehabilitation Medicine, Ministry of Science and Technology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, China
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| |
Collapse
|
11
|
Duan P, Wang H, Yi X, Zhang H, Chen H, Pan Z. C/EBPα regulates the fate of bone marrow mesenchymal stem cells and steroid-induced avascular necrosis of the femoral head by targeting the PPARγ signalling pathway. Stem Cell Res Ther 2022; 13:342. [PMID: 35883192 PMCID: PMC9327281 DOI: 10.1186/s13287-022-03027-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 07/02/2022] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND The imbalance of osteogenic/adipogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) is closely related to steroid-induced avascular necrosis of the femoral head (SANFH). We aimed to investigate the epigenetic mechanism of intramedullary fat accumulation and continuous osteonecrosis after glucocorticoid (GC) withdrawal in SANFH. METHODS An SANFH model was established in SD rats, which received an intermittent high GC dose for the first 4 weeks followed by an additional 4 weeks without GC. We explored the synergistic effects and mechanisms of C/EBPα and PPARγ on the differentiation of BMSCs by lentivirus-mediated gene knockdown and overexpression assays. A chromatin immunoprecipitation assay was performed to identify epigenetic modification sites on PPARγ in vivo and in vitro. RESULTS In the SANFH model, intramedullary fat was significantly increased, and the transcription factors C/EBPα and PPARγ were upregulated simultaneously in the femoral head. In vitro, C/EBPα promoted adipogenic differentiation of BMSCs by targeting the PPARγ signalling pathway, while overexpression of C/EBPα significantly impaired osteogenic differentiation. Further studies demonstrated that histone H3K27 acetylation of PPARγ played an important role in the epigenetic mechanism underlying SANFH. C/EBPα upregulates the histone H3K27 acetylation level in the PPARγ promoter region by inhibiting HDAC1. Additionally, inhibiting the histone acetylation level of PPARγ effectively prevented adipogenic differentiation, thus slowing the progression of SANFH. CONCLUSIONS Our results demonstrate the molecular mechanism by which C/EBPα regulates PPARγ expression by acetylating histones and revealed the epigenetic phenomenon in SANFH for the first time.
Collapse
Affiliation(s)
- Ping Duan
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hanyu Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xinzeyu Yi
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hao Zhang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hui Chen
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhenyu Pan
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
12
|
Pan J, Wang J, Fang K, Hou W, Li B, Zhao J, Ma X. RNA m 6A Alterations Induced by Biomineralization Nanoparticles: A Proof-of-Concept Study of Epitranscriptomics for Nanotoxicity Evaluation. NANOSCALE RESEARCH LETTERS 2022; 17:23. [PMID: 35122526 PMCID: PMC8817964 DOI: 10.1186/s11671-022-03663-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/26/2022] [Indexed: 06/14/2023]
Abstract
Although various strategies have been included in nanotoxicity evaluation, epitranscriptomics has rarely been integrated into this field. In this proof-of-concept study, N6-methyladenosine (m6A) changes of mRNA in HEK293T cells induced by three bovine serum albumin (BSA)-templated Au, CuS and Gd2O3 nanoparticles are systematically explored, and their possible biological mechanisms are preliminarily investigated. It has been found that all the three BSA-templated nanoparticles can reduce m6A levels, and the genes with reduced m6A are enriched for TGF-beta signaling, which is critical for cell proliferation, differentiation and apoptosis. Further results indicate that abnormal aggregation of m6A-related enzymes at least partly account for the nanoparticle-induced epitranscriptomic changes. These findings demonstrate that epitranscriptomics analysis can provide an unprecedented landscape of the biological effect induced by nanomaterials, which should be involved in the nanotoxicity evaluation to promote the potential clinical translation of nanomaterials.
Collapse
Affiliation(s)
- Jinbin Pan
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Jiaojiao Wang
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Kun Fang
- Department of Radiology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Wenjing Hou
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Bing Li
- Department of Orthopedics, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Jie Zhao
- Department of Orthopedics, Tianjin Hospital, Tianjin University, Tianjin, 300211, China.
| | - Xinlong Ma
- Department of Orthopedics, Tianjin Hospital, Tianjin University, Tianjin, 300211, China.
| |
Collapse
|
13
|
Chi J, Zhang W, Li Y, Zhao J, Zheng X, Gao M. TET3 Mediates 5hmC Level and Promotes Tumorigenesis by Activating AMPK Pathway in Papillary Thyroid Cancer. Int J Endocrinol 2022; 2022:2658727. [PMID: 35755313 PMCID: PMC9217609 DOI: 10.1155/2022/2658727] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/18/2022] [Accepted: 05/05/2022] [Indexed: 11/18/2022] Open
Abstract
Thyroid cancer is the most common endocrine malignant tumor. The accurate risk stratification and prognosis assessment is particularly important for patients with thyroid cancer, which can reduce the tumor recurrence rate, morbidity, and mortality effectively. DNA methylation is one of the most widely studied epigenetic modifications. Many studies have shown that 5hmC-mediated demethylation played an important role in tumors. The hydroxylation of 5mC is catalyzed by ten-eleven translocation dioxygenase (TET). In this study, we first found that the abnormal expression of 5hmC was closely related to microcarcinoma, multifocal, extraglandular invasion and lymph node metastasis of thyroid carcinoma. Then, we identified TET3 was differentially expressed in thyroid cancers and normal tissues from the TET family. TET3 can promote the proliferation, migration, and invasion of thyroid cancer. TET3-mediated 5hmC can regulate the transcription of AMPK pathway-related genes to activate the AMPK pathway and autophagy and therefore promote PTC proliferation. These findings provide a preclinical rationale for the design of novel therapeutic strategies for this target to improve the clinical outcome of patients with PTC.
Collapse
Affiliation(s)
- Jiadong Chi
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Wei Zhang
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- School of Medicine, Nankai University, Tianjin 300071, China
- Department of Breast and Thyroid Surgery, Tianjin Union Medical Center, No. 190 Jieyuan Road, Hongqiao District, Tianjin 300121, China
| | - Yigong Li
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jie Zhao
- Department of Orthopedics, Tianjin Hospital, Tianjin University, Tianjin 300211, China
| | - Xiangqian Zheng
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Ming Gao
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Department of Breast and Thyroid Surgery, Tianjin Union Medical Center, No. 190 Jieyuan Road, Hongqiao District, Tianjin 300121, China
| |
Collapse
|
14
|
Wang T, Wu H, Sun M, Liu T, An F, Dong Q, Wang J. Association between Genetic Polymorphisms of MIR3142HG and the Risk of Steroid-Induced Osteonecrosis of the Femoral Head in the Population of Northern China. Public Health Genomics 2021; 25:1-9. [PMID: 34788764 DOI: 10.1159/000519577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 09/09/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Steroid-induced osteonecrosis of the femoral head (ONFH) is aseptic necrosis of the femoral head caused by glucocorticoid use. Once necrotic femoral head necrosis occurs, it irreversibly affects the quality of life seriously. Studies have shown that the susceptibility to steroid-induced ONFH is likely to be related to the variation of miRNA-coding genes. Therefore, this study aimed was to investigate the effect of MIR3142HG on steroid-induced ONFH. METHODS Agena MassARRAY was used to genotype MIR3142HG gene rs1582417, rs2431689, rs7727155, and rs17057846 in 199 patients and 725 healthy people. A genetic model and haplotype analysis were used to evaluate the relationship between the MIR3142HG polymorphism and the risk of steroid-induced ONFH. The odds ratio and 95% confidence intervals were obtained through logistic regression to assess the influence of gene polymorphisms on the occurrence of steroid-induced ONFH. RESULTS The consequences show that rs7727115 is a protective factor, it could reduce the risk of steroid-induced ONFH, and rs1582417 could increase the risk of steroid-induced ONFH. In the genetic model, rs1582417 was associated with increased risk of alcohol-induced ONFH in dominant model and log-additive model. rs7727115 showed a decreased risk in codominant model, dominant model, and log-additive model. In addition, rs2431689 is related to HDL-C (p = 0.012) and ApoA1 (p = 0.010) levels, and rs17057846 (p = 0.024) is related to ApoB levels. Thelinkage analysis indicated 3 single-nucleotide polymorphisms (rs2431689, rs7727115, and rs17057846) in MIR3142HG with significant chain imbalance. In addition, haplotype "GGG" of MIR3142HG was found out and is harmful for steroid-induced ONFH. CONCLUSION Our results first confirm that the genetic polymorphism of MIR3142HG is associated with steroid-induced ONFH susceptibility in Chinese Han population.
Collapse
Affiliation(s)
- Tiantian Wang
- Inner Mongolia Medical University, Hohhot, China
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Huiqiang Wu
- Rehabilitation Division, Inner Mongolia Autonomous Region Hospital of Traditional Chinese Medicine, Hohhot, China
| | - Menghu Sun
- Inner Mongolia Medical University, Hohhot, China
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Tingting Liu
- Inner Mongolia Medical University, Hohhot, China
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Feimeng An
- Rehabilitation Division, Inner Mongolia Autonomous Region Hospital of Traditional Chinese Medicine, Hohhot, China
| | - Qiumei Dong
- College of Traditional Chinese Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Jianzhong Wang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
15
|
Fan ZQ, Bai SC, Xu Q, Li ZJ, Cui WH, Li H, Li XH, Zhang HF. Oxidative Stress Induced Osteocyte Apoptosis in Steroid-Induced Femoral Head Necrosis. Orthop Surg 2021; 13:2145-2152. [PMID: 34559465 PMCID: PMC8528976 DOI: 10.1111/os.13127] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/05/2021] [Accepted: 06/05/2021] [Indexed: 12/20/2022] Open
Abstract
Objective To investigate the effect and mechanism of Glucocorticoids (GCs) induced oxidative stress and apoptosis on necrosis of the femoral head in patients and rats. Methods Eight patients with steroid‐induced avascular necrosis of the femoral head (SINFH) and eight patients with developmental dysplasia of the hips (DDH) were enrolled in our study. In animal model, twenty male Sprague‐Dawley rats were randomly divided into two groups (SINFH group and NS group). The SINFH model group received the methylprednisolone (MPS) injection, while control group was injected with normal saline (NS). MRI was used to confirm SINFH rat model was established successfully. Then, the rats were sacrificed 4 weeks later and femoral head samples were harvested. Histopathological staining was preformed to evaluate osteonecrosis. TUNEL staining was performed with 8‐OHdG and DAPI immunofluorescence staining to evaluate oxidative injury and osteocyte apoptosis. Immunohistochemistry staining was used to detect Nox1, Nox2, and Nox4 protein expression. Results MRI showed signs of typical osteonecrosis of femoral head in SIHFH patients. Histopathological staining showed that the rate of empty lacunae in SINFH patients was significantly higher (56.88% ± 9.72% vs 19.92% ± 4.18%, T = −11.04, P < 0.001) than that in DDH patients. The immunofluorescence staining indicated that the TUNEL‐positive cell and 8‐OHdG‐positve cell in SINFH patients were significantly higher (49.32% ± 12.95% vs 8.00% ± 2.11%, T = −7.04, P = 0.002, 54.6% ± 23.8% vs 9.75% ± 3.31%, T = −4.17, P = 0.003) compared to the DDH patients. The immunohistochemistry staining showed that the protein expression of NOX1, NOX2 and NOX4 in SINFH patients were significantly increased (64.50% ± 7.57% vs 37.58% ± 9.23%, T = −3.88, P = 0.018, 90.84% ± 2.93% vs 49.56% ± 16.47%, T = −5.46, P = 0.001, 85.46% ± 9.3% vs 40.69% ± 6.77%, T = −8.03, P = 0.001) compared to the DDH patients. In animal model, MRI showed signs of edema of femoral head in MPS group, which represents SINFH rat model was established successfully. Histological evaluation showed the rate of empty lacunae in MPS group was significantly higher (25.85% ± 4.68% vs 9.35% ± 1.99%, T = −7.96, P < 0.001) than that in NS group. The immunofluorescence staining indicated that the TUNEL‐positive cell and 8‐OHdG‐positve cell (in MPS group were significantly increased (31.93% ± 1.01% vs 11.73% ± 1.16%, T = −32.26, P < 0.001, 47.59% ± 1.39% vs 22.07% ± 2.45%, T = −22.18, P < 0.001) compared to the NS group. The immunohistochemistry staining showed that the expression of NOX2 in MPS group was significantly increased (76.77% ± 8.34% vs 50.32% ± 10.84%, T = −4.74, P = 0.001) compare with NS group. Conclusion Our findings indicated that GC‐induced NOXs expression may be an important source of oxidative stress, which could lead to osteocyte apoptosis in the process of SINFH
Collapse
Affiliation(s)
- Zhen-Qi Fan
- The Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Shu-Cai Bai
- Department of Orthopedics, Tianjin Hospital, Tianjin, China
| | - Qian Xu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhi-Jun Li
- The Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wen-Hao Cui
- Department of Endocrinology, The Shenzhen Second People's Hospital, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.,Department of pharmacology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hui Li
- The Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiao-Hui Li
- Department of Orthopedics, Tianjin Hospital, Tianjin, China
| | - Hua-Feng Zhang
- The Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
16
|
Medication-Related Osteonecrosis of the Jaw: A Critical Narrative Review. J Clin Med 2021; 10:jcm10194367. [PMID: 34640383 PMCID: PMC8509366 DOI: 10.3390/jcm10194367] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/15/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Nearly two decades have passed since a paradoxical reaction in the orofacial region to some bone modifying agents and other drugs was recognized, namely medication-related osteonecrosis of the jaw (MRONJ). Purpose: The aim of this manuscript was to critically review published data on MRONJ to provide an update on key terminology, concepts, and current trends in terms of prevention and diagnosis. In addition, our objective was to examine and evaluate the therapeutic options available for MRONJ. Methods: The authors perused the most relevant literature relating to MRONJ through a search in textbooks and published articles included in several databases for the years 2003–2021. Results and conclusions: A comprehensive update of the current understanding of these matters was elaborated, addressing these topics and identifying relevant gaps of knowledge. This review describes our updated view of the previous thematic blocks, highlights our current clinical directions, and emphasizes controversial aspects and barriers that may lead to extending the accumulating body of evidence related to this severe treatment sequela.
Collapse
|
17
|
DNA 5-hydroxymethylcytosine in pediatric central nervous system tumors may impact tumor classification and is a positive prognostic marker. Clin Epigenetics 2021; 13:176. [PMID: 34538273 PMCID: PMC8451154 DOI: 10.1186/s13148-021-01156-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 08/18/2021] [Indexed: 01/05/2023] Open
Abstract
Background Nucleotide-specific 5-hydroxymethylcytosine (5hmC) remains understudied in pediatric central nervous system (CNS) tumors. 5hmC is abundant in the brain, and alterations to 5hmC in adult CNS tumors have been reported. However, traditional approaches to measure DNA methylation do not distinguish between 5-methylcytosine (5mC) and its oxidized counterpart 5hmC, including those used to build CNS tumor DNA methylation classification systems. We measured 5hmC and 5mC epigenome-wide at nucleotide resolution in glioma, ependymoma, and embryonal tumors from children, as well as control pediatric brain tissues using tandem bisulfite and oxidative bisulfite treatments followed by hybridization to the Illumina Methylation EPIC Array that interrogates over 860,000 CpG loci.
Results Linear mixed effects models adjusted for age and sex tested the CpG-specific differences in 5hmC between tumor and non-tumor samples, as well as between tumor subtypes. Results from model-based clustering of tumors was used to test the relation of cluster membership with patient survival through multivariable Cox proportional hazards regression. We also assessed the robustness of multiple epigenetic CNS tumor classification methods to 5mC-specific data in both pediatric and adult CNS tumors. Compared to non-tumor samples, tumors were hypohydroxymethylated across the epigenome and tumor 5hmC localized to regulatory elements crucial to cell identity, including transcription factor binding sites and super-enhancers. Differentially hydroxymethylated loci among tumor subtypes tended to be hypermethylated and disproportionally found in CTCF binding sites and genes related to posttranscriptional RNA regulation, such as DICER1. Model-based clustering results indicated that patients with low 5hmC patterns have poorer overall survival and increased risk of recurrence. Our results suggest 5mC-specific data from OxBS-treated samples impacts methylation-based tumor classification systems giving new opportunities for further refinement of classifiers for both pediatric and adult tumors. Conclusions We identified that 5hmC localizes to super-enhancers, and genes commonly implicated in pediatric CNS tumors were differentially hypohydroxymethylated. We demonstrated that distinguishing methylation and hydroxymethylation is critical in identifying tumor-related epigenetic changes. These results have implications for patient prognostication, considerations of epigenetic therapy in CNS tumors, and for emerging molecular neuropathology classification approaches. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01156-9.
Collapse
|
18
|
Yin C, Tian Y, Hu L, Yu Y, Wu Z, Zhang Y, Wang X, Miao Z, Qian A. MACF1 alleviates aging-related osteoporosis via HES1. J Cell Mol Med 2021; 25:6242-6257. [PMID: 34133068 PMCID: PMC8366449 DOI: 10.1111/jcmm.16579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 03/24/2021] [Accepted: 04/08/2021] [Indexed: 12/30/2022] Open
Abstract
Ageing-related osteoporosis is becoming an emerging threat to human health along with the ageing of human population. The decreased rate of osteogenic differentiation and bone formation is the major cause of ageing-related osteoporosis. Microtubule actin cross-linking factor 1 (MACF1) is an important cytoskeletal factor that promotes osteogenic differentiation and bone formation. However, the relationship between MACF1 expression and ageing-related osteoporosis remains unclear. This study has investigated the expression pattern of MACF1 in bone tissues of ageing-related osteoporosis patients and ageing mice. The study has further elucidated the mechanism of MACF1 promoting bone formation by inhibiting HES1 expression and activity. Moreover, the therapeutic effect of MACF1 on ageing-related osteoporosis and post-menopausal osteoporosis was evaluated through in situ injection of the MACF1 overexpression plasmid. The study supplemented the molecular mechanisms between ageing and bone formation, and provided novel targets and potential therapeutic strategy for ageing-related osteoporosis.
Collapse
Affiliation(s)
- Chong Yin
- Lab for Bone MetabolismXi'an Key Laboratory of Special Medicine and Health EngineeringKey Lab for Space Biosciences and BiotechnologyResearch Center for Special Medicine and Health Systems EngineeringNPU‐UAB Joint Laboratory for Bone MetabolismSchool of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
- Lab of Epigenetics and RNA TherapyDepartment of Clinical Laboratory, Academician (Expert) WorkstationAffiliated Hospital of North Sichuan Medical CollegeNanchongChina
- Department of Laboratory MedicineNorth Sichuan Medical CollegeNanchongChina
- Translational Medicine Research CenterNorth Sichuan Medical CollegeNanchongChina
| | - Ye Tian
- Lab for Bone MetabolismXi'an Key Laboratory of Special Medicine and Health EngineeringKey Lab for Space Biosciences and BiotechnologyResearch Center for Special Medicine and Health Systems EngineeringNPU‐UAB Joint Laboratory for Bone MetabolismSchool of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Lifang Hu
- Lab for Bone MetabolismXi'an Key Laboratory of Special Medicine and Health EngineeringKey Lab for Space Biosciences and BiotechnologyResearch Center for Special Medicine and Health Systems EngineeringNPU‐UAB Joint Laboratory for Bone MetabolismSchool of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Yang Yu
- Tianjin Key Laboratory on Technologies Enabling Development Clinical Therapeutics and Diagnostics (Theranostics)School of PharmacyTianjin Medical UniversityTianjinChina
| | - Zixiang Wu
- Lab for Bone MetabolismXi'an Key Laboratory of Special Medicine and Health EngineeringKey Lab for Space Biosciences and BiotechnologyResearch Center for Special Medicine and Health Systems EngineeringNPU‐UAB Joint Laboratory for Bone MetabolismSchool of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Yan Zhang
- Lab for Bone MetabolismXi'an Key Laboratory of Special Medicine and Health EngineeringKey Lab for Space Biosciences and BiotechnologyResearch Center for Special Medicine and Health Systems EngineeringNPU‐UAB Joint Laboratory for Bone MetabolismSchool of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Xue Wang
- Lab for Bone MetabolismXi'an Key Laboratory of Special Medicine and Health EngineeringKey Lab for Space Biosciences and BiotechnologyResearch Center for Special Medicine and Health Systems EngineeringNPU‐UAB Joint Laboratory for Bone MetabolismSchool of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Zhiping Miao
- Lab for Bone MetabolismXi'an Key Laboratory of Special Medicine and Health EngineeringKey Lab for Space Biosciences and BiotechnologyResearch Center for Special Medicine and Health Systems EngineeringNPU‐UAB Joint Laboratory for Bone MetabolismSchool of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| | - Airong Qian
- Lab for Bone MetabolismXi'an Key Laboratory of Special Medicine and Health EngineeringKey Lab for Space Biosciences and BiotechnologyResearch Center for Special Medicine and Health Systems EngineeringNPU‐UAB Joint Laboratory for Bone MetabolismSchool of Life SciencesNorthwestern Polytechnical UniversityXi'anChina
| |
Collapse
|
19
|
Li G, Li B, Li B, Zhao J, Wang X, Luo R, Li Y, Liu J, Hu R. The role of biomechanical forces and MALAT1/miR-329-5p/PRIP signalling on glucocorticoid-induced osteonecrosis of the femoral head. J Cell Mol Med 2021; 25:5164-5176. [PMID: 33939272 PMCID: PMC8178276 DOI: 10.1111/jcmm.16510] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/25/2021] [Accepted: 03/12/2021] [Indexed: 12/19/2022] Open
Abstract
Glucocorticoid‐induced osteonecrosis of the femoral head (GIONFH) is a common orthopaedic disease. GIONFH primarily manifests clinically as hip pain in the early stages, followed by the collapse of the femoral head, narrowing of the hip joint space and damage to the acetabulum, resulting in severely impaired mobility. However, the pathogenesis of GIONFH is not clearly understood. Recently, biomechanical forces and non‐coding RNAs have been suggested to play important roles in the pathogenesis of GIONFH. This study aimed to evaluate the role of biomechanical forced and non‐coding RNAs in GIONFH. We utilized an in vivo, rat model of GIONFH and used MRI, μCT, GIONFH‐TST (tail suspension test), GIONFH‐treadmill, haematoxylin and eosin staining, qRT‐PCR and Western blot analysis to analyse the roles of biomechanical forces and non‐coding RNAs in GIONFH. We used RAW264.7 cells and MC3T3E1 cells to verify the role of MALAT1/miR‐329‐5p/PRIP signalling using a dual luciferase reporter assay, qRT‐PCR and Western blot analysis. The results demonstrated that MALAT1 and PRIP were up‐regulated in the femoral head tissues of GIONFH rats, RAW264.7 cells, and MC3T3E1 cells exposed to dexamethasone (Dex). Knockdown of MALAT1 decreased PRIP expression in rats and cultured cells and rescued glucocorticoid‐induced osteonecrosis of femoral head in rats. The dual luciferase reporter gene assay revealed a targeting relationship for MALAT1/miR‐329‐5p and miR‐329‐5p/PRIP in MC3T3E1 and RAW264.7 cells. In conclusion, MALAT1 played a vital role in the pathogenesis of GIONFH by binding to (‘sponging’) miR‐329‐5p to up‐regulate PRIP. Also, biomechanical forces aggravated the pathogenesis of GIONFH through MALAT1/miR‐329‐5p/PRIP signalling.
Collapse
Affiliation(s)
- Guomin Li
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, China
| | - Bing Li
- Department of Joint, Tianjin Hospital, Tianjin, China
| | - Bo Li
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jie Zhao
- Department of Joint, Tianjin Hospital, Tianjin, China
| | - Xiaoquan Wang
- Department of Joint, Tianjin Hospital, Tianjin, China
| | - Rui Luo
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yankun Li
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jun Liu
- Department of Joint, Tianjin Hospital, Tianjin, China
| | - Ruyin Hu
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
20
|
Li B, Zhao J, Ma J, Chen W, Zhou C, Wei W, Li S, Li G, Xin G, Zhang Y, Liu J, Wang Y, Ma X. Cross-talk Between Histone and DNA Methylation Mediates Bone Loss in Hind Limb Unloading. J Bone Miner Res 2021; 36:956-967. [PMID: 33465813 DOI: 10.1002/jbmr.4253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/11/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022]
Abstract
Bone loss induced by mechanical unloading is a common skeletal disease, but the precise mechanism remains unclear. The current study investigated the role of histone methylation, a key epigenetic marker, and its cross-talk with DNA methylation in bone loss induced by mechanical unloading. The expression of G9a, ubiquitin-like with PHD and ring finger domains 1 (UHRF1), and DNA methylation transferase 1 (DNMT1) were increased in hind limb unloading (HLU) rats. This was accompanied by an increased level of histone H3 lysine 9 (H3K9) di-/tri-methylation at lncH19 promoter. Then, alteration of G9a, DNMT1, or UHRF1 expression significantly affected lncH19 level and osteogenic activity in UMR106 cells. Osteogenic gene expression and matrix mineralization were robustly promoted after simultaneous knockdown of G9a, DNMT1, and UHRF1. Furthermore, physical interactions of lncH19 promoter with G9a and DNMT1, as well as direct interactions among DNMT1, G9a, and UHRF1 were detected. Importantly, overexpression of DNMT1, G9a, or UHRF1, respectively, resulted in enrichment of H3K9me2/me3 and 5-methylcytosine at lncH19 promoter. Finally, in vivo rescue experiments indicated that knockdown of DNMT1, G9a, or UHRF1 significantly relieved bone loss in HLU rats. In conclusion, our research demonstrated the critical role of H3K9 methylation and its cross-talk with DNA methylation in regulating lncH19 expression and bone loss in HLU rats. Combined targeting of DNMT1, G9a, and UHRF1 could be a promising strategy for the treatment of bone loss induced by mechanical unloading. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Bing Li
- Joint Department, Tianjin Hospital, Tianjin, China
| | - Jie Zhao
- Orthopedic Department, Tianjin Hospital, Tianjin, China
| | - Jianxiong Ma
- Tianjin Orthopedic Research Institute, Tianjin, China
| | - Weibo Chen
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Ce Zhou
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Wuzeng Wei
- Joint Department, Tianjin Hospital, Tianjin, China
| | - Shuai Li
- Joint Department, Tianjin Hospital, Tianjin, China
| | - Guomin Li
- Joint Department, Tianjin Hospital, Tianjin, China
| | - Guosheng Xin
- Tianjin Orthopedic Research Institute, Tianjin, China
| | - Yang Zhang
- Tianjin Orthopedic Research Institute, Tianjin, China
| | - Jun Liu
- Joint Department, Tianjin Hospital, Tianjin, China
| | - Yinsong Wang
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xinlong Ma
- Joint Department, Tianjin Hospital, Tianjin, China.,Orthopedic Department, Tianjin Hospital, Tianjin, China.,Tianjin Orthopedic Research Institute, Tianjin, China
| |
Collapse
|
21
|
Chi J, Zhao J, Wei S, Li Y, Zhi J, Wang H, Hou X, Hu L, Zheng X, Gao M. A CRISPR-Cas9-Based Near-Infrared Upconversion-Activated DNA Methylation Editing System. ACS APPLIED MATERIALS & INTERFACES 2021; 13:6043-6052. [PMID: 33525876 DOI: 10.1021/acsami.0c21223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
DNA methylation is a kind of a crucial epigenetic marker orchestrating gene expression, molecular function, and cellular phenotype. However, manipulating the methylation status of specific genes remains challenging. Here, a clustered regularly interspaced palindromic repeats-Cas9-based near-infrared upconversion-activated DNA methylation editing system (CNAMS) was designed for the optogenetic editing of DNA methylation. The fusion proteins of photosensitive CRY2PHR, the catalytic domain of DNMT3A or TET1, and the fusion proteins for CIBN and catalytically inactive Cas9 (dCas9) were engineered. The CNAMS could control DNA methylation editing in response to blue light, thus allowing methylation editing in a spatiotemporal manner. Furthermore, after combination with upconversion nanoparticles, the spectral sensitivity of DNA methylation editing was extended from the blue light to near-infrared (NIR) light, providing the possibility for remote DNA methylation editing. These results demonstrated a meaningful step forward toward realizing the specific editing of DNA methylation, suggesting the wide utility of our CNAMS for functional studies on epigenetic regulation and potential therapeutic strategies for related diseases.
Collapse
Affiliation(s)
- Jiadong Chi
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical, Research Center of Cancer, 300060Tianjin, China
| | - Jingzhu Zhao
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical, Research Center of Cancer, 300060Tianjin, China
| | - Songfeng Wei
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical, Research Center of Cancer, 300060Tianjin, China
| | - Yigong Li
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical, Research Center of Cancer, 300060Tianjin, China
| | - Jingtai Zhi
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical, Research Center of Cancer, 300060Tianjin, China
| | - Huijuan Wang
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical, Research Center of Cancer, 300060Tianjin, China
| | - Xiukun Hou
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical, Research Center of Cancer, 300060Tianjin, China
| | - Linfei Hu
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical, Research Center of Cancer, 300060Tianjin, China
| | - Xiangqian Zheng
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical, Research Center of Cancer, 300060Tianjin, China
| | - Ming Gao
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, Oncology Key Laboratory of Cancer Prevention and Therapy, National Clinical, Research Center of Cancer, 300060Tianjin, China
| |
Collapse
|
22
|
Tan L, Song X, Ren Y, Wang M, Guo C, Guo D, Gu Y, Li Y, Cao Z, Deng Y. Anti-inflammatory effects of cordycepin: A review. Phytother Res 2020; 35:1284-1297. [PMID: 33090621 DOI: 10.1002/ptr.6890] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/25/2020] [Accepted: 09/13/2020] [Indexed: 01/08/2023]
Abstract
Cordycepin is the major bioactive component extracted from Cordyceps militaris. In recent years, cordycepin has received increasing attention owing to its multiple pharmacological activities. This study reviews recent researches on the anti-inflammatory effects and the related activities of cordycepin. The results from our review indicate that cordycepin exerts protective effects against inflammatory injury for many diseases including acute lung injury (ALI), asthma, rheumatoid arthritis, Parkinson's disease (PD), hepatitis, atherosclerosis, and atopic dermatitis. Cordycepin regulates the NF-κB, RIP2/Caspase-1, Akt/GSK-3β/p70S6K, TGF-β/Smads, and Nrf2/HO-1 signaling pathways among others. Several studies focusing on cordycepin derivatives were reviewed and found to down metabolic velocity of cordycepin and increase its bioavailability. Moreover, cordycepin enhanced immunity, inhibited the proliferation of viral RNA, and suppressed cytokine storms, thereby suggesting its potential to treat COVID-19 and other viral infections. From the collected and reviewed information, this article provides the theoretical basis for the clinical applications of cordycepin and discusses the path for future studies focusing on expanding the medicinal use of cordycepin. Taken together, cordycepin and its analogs show great potential as the next new class of anti-inflammatory agents.
Collapse
Affiliation(s)
- Lu Tan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Xiaominting Song
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Yali Ren
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Miao Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Chuanjie Guo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Dale Guo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Yucheng Gu
- Syngenta Jealott's Hill International Research Centre, Berkshire, UK
| | - Yuzhi Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Zhixing Cao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| | - Yun Deng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine; Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education; National Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Chengdu, China
| |
Collapse
|
23
|
Zhao J, Li B, Ma J, Jin W, Ma X. Photoactivatable RNA N 6 -Methyladenosine Editing with CRISPR-Cas13. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1907301. [PMID: 32583968 DOI: 10.1002/smll.201907301] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 04/27/2020] [Indexed: 06/11/2023]
Abstract
RNA has important and diverse biological roles, but the molecular methods to manipulate it spatiotemporally are limited. Here, an engineered photoactivatable RNA N6 -methyladenosine (m6 A) editing system with CRISPR-Cas13 is designed to direct specific m6 A editing. Light-inducible heterodimerizing proteins CIBN and CRY2PHR are fused to catalytically inactive PguCas13 (dCas13) and m6 A effectors, respectively. This system, referred to as PAMEC, enables the spatiotemporal control of m6 A editing in response to blue light. Further optimization of this system to create a highly efficient version, known as PAMECR , allows the manipulation of multiple genes robustly and simultaneously. When coupled with an upconversion nanoparticle film, the optogenetic operation window is extended from the visible range to tissue-penetrable near-infrared wavelengths, which offers an appealing avenue to remotely control RNA editing. These results show that PAMEC is a promising optogenetic platform for flexible and efficient targeting of RNA, with broad applicability for epitranscriptome engineering, imaging, and future therapeutic development.
Collapse
Affiliation(s)
- Jie Zhao
- Department of Orthopedics, Tianjin University Tianjin Hospital, Tianjin, 300211, China
| | - Bing Li
- Department of Orthopedics, Tianjin University Tianjin Hospital, Tianjin, 300211, China
| | - Jianxiong Ma
- Orthopedic Research Institute, Tianjin University Tianjin Hospital, Tianjin, 300050, China
| | - Weilin Jin
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, China
| | - Xinlong Ma
- Department of Orthopedics, Tianjin University Tianjin Hospital, Tianjin, 300211, China
| |
Collapse
|
24
|
Chen G, Wang Q, Li Z, Yang Q, Liu Y, Du Z, Zhang G, Song Y. Circular RNA CDR1as promotes adipogenic and suppresses osteogenic differentiation of BMSCs in steroid-induced osteonecrosis of the femoral head. Bone 2020; 133:115258. [PMID: 32018039 DOI: 10.1016/j.bone.2020.115258] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 01/31/2020] [Accepted: 01/31/2020] [Indexed: 12/13/2022]
Abstract
Steroid-induced osteonecrosis of the femoral head (SONFH) is a common debilitating orthopedic disease. The bone marrow mesenchymal stem cells (BMSCs) are a type of mesenchymal stem cells which play crucial roles in bone repair. The adipogenic/osteogenic differentiation disorder of BMSCs has been widely perceived contributing to SONFH. However, the regulatory mechanism of BMSCs differentiation disorder still remains unclear. Circular RNA (circRNA), a kind of stable ncRNA, plays important roles in regulating gene expression via various ways. To date, there are no studies to uncover the circRNA expression profile and screen out the key circRNAs playing crucial roles in adipogenic/osteogenic differentiation disorder of SONFH-BMSCs. In present study, we detected the circRNA expression profiles in SONFH-BMSCs for the first time. A total of 820 circRNAs were differentially expressed in SONFH-BMSCs, including 460 up- and 360 down-regulated circRNAs. Bioinformatics analysis indicates circRNA CDR1as, one up-regulated circRNA, may play crucial role in adipogenic/osteogenic differentiation disorder of SONFH-BMSCs via CDR1as-miR-7-5p-WNT5B axis. Knocking-down CDR1as resulted in increasing of osteogenic differentiation and decreasing of adipogenic differentiation of BMSCs, while over-expressing CDR1as resulted in decreasing of osteogenic differentiation and increasing of adipogenic differentiation of BMSCs. The miR-7-5p binding sites of CDR1as and WNT5B were verified by luciferase reporter gene assay. Our study may provide new insights into the molecular mechanisms of osteogenic/adipogenic differentiation disorder of SONFH-BMSCs and new biomarkers for the diagnosis and treatment of SONFH.
Collapse
Affiliation(s)
- Gaoyang Chen
- Department of Orthopedics, The Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; Research Centre of the Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Ziqiang Street 218, Changchun, Jilin 130041, China.
| | - Qingyu Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; Research Centre of the Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Ziqiang Street 218, Changchun, Jilin 130041, China
| | - Zhaoyan Li
- Department of Orthopedics, The Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; Research Centre of the Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Ziqiang Street 218, Changchun, Jilin 130041, China
| | - Qiwei Yang
- Research Centre of the Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Ziqiang Street 218, Changchun, Jilin 130041, China.
| | - Yuzhe Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Ziqiang Street 218, Changchun, Jilin 130041, China.
| | - Zhenwu Du
- Department of Orthopedics, The Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; Research Centre of the Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Ziqiang Street 218, Changchun, Jilin 130041, China
| | - Guizhen Zhang
- Department of Orthopedics, The Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; Research Centre of the Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Ziqiang Street 218, Changchun, Jilin 130041, China
| | - Yang Song
- Department of Orthopedics, The Second Hospital of Jilin University, Ziqiang Street 218, Changchun, Jilin 130041, China; The Engineering Research Centre of Molecular Diagnosis and Cell Treatment for Metabolic Bone Diseases of Jilin Province, Ziqiang Street 218, Changchun, Jilin 130041, China.
| |
Collapse
|
25
|
Zhang Y, Zheng D, Fang Q, Zhong M. Aberrant hydroxymethylation of ANGPTL4 is associated with selective intrauterine growth restriction in monochorionic twin pregnancies. Epigenetics 2020; 15:887-899. [PMID: 32114885 DOI: 10.1080/15592294.2020.1737355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Selective intrauterine growth restriction (sIUGR) is a severe complication in monochorionic (MC) twin pregnancies, and it carries increased risks of poor prognosis. Current data suggest that vascular anastomoses and unequal placental sharing may be the key contributor to discordant foetal growth. While MC twins derive from a single zygote and have almost identical genetic information, the precise mechanisms remain unknown. DNA hydroxymethylation is a newly discovered epigenetic feature associated with gene regulation and modification. Here, we investigate discordant hydroxymethylation patterns between two placental shares of sIUGR and analyse the potential role of aberrant hydroxymethylation of angiopoietin-like 4 (ANGPTL4) in placental dysplasia. Hydroxymethylation DNA immunoprecipitation (hMeDIP)-chip and mRNA sequencing were performed to identify hydroxymethylation-associated genes. Real-time qPCR, western blotting, and immunohistochemistry were used to confirm ANGPTL4 expression. The mechanisms regulating ANGPTL4 were investigated by cell migration assay, invasion assay, viability assay, and apoptotic ratio assays, western blotting and hMeDIP-qPCR. Decreased ANGPTL4 was detected in the smaller placental shares of sIUGR. ANGPTL4 knockdown suppressed trophoblast invasiveness and migration, which possibly occurred through hypoxia inducible factor 1α (HIF-1α) and HIF-1 signalling pathway. Hypoxia leads to aberrant expression of ANGPTL4 and HIF-1α, positively correlated with their aberrant hydroxymethylation levels in promoter regions. Aberrant hydroxymethylation of ANGPTL4 may contribute to placental impairment by the HIF-1 signalling pathway in smaller placental shares of sIUGR.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Dezhong Zheng
- Department of Cardiology, The Third Affiliated Hospital of Southern Medical University, Southern Medical University , Guangzhou, China.,Department of Cardiology, Nanfang Hospital, Southern Medical University , Guangzhou, China
| | - Qun Fang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University , Guangzhou, China
| | - Mei Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University , Guangzhou, China
| |
Collapse
|
26
|
Zhang J, Li J, Zhu Y, Miao Z, Tian Y. Forced running exercise mitigates radiation-induced cognitive deficits via regulated DNA hydroxymethylation. Epigenomics 2020; 12:385-396. [PMID: 32041423 DOI: 10.2217/epi-2019-0370] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: Roles of forced running exercise (FE) in remediation of neurogenesis inhibition and radiation-induced cognitive dysfunction were investigated in a whole-brain irradiation mice model via the regulation of DNA 5-hydroxymethylation modification (5 hmC) and its catalytic enzymes ten-eleven translocation (Tet) proteins. Materials & methods: Hippocampal neurogenesis and cognitive function, DNA 5 hmC level and Tet expression were determined in mice. Results: The expression of DNA 5 hmC and Tet2, brain-derived neurotrophic factor significantly decreased in hippocampus postradiation. FE mitigated radiation-induced neurogenesis deficits and cognitive dysfunction. Furthermore, FE increased 5 hmC and brain-derived neurotrophic factor expression. SC1, a Tet inhibitor, reversed partly such changes. Conclusion: Tet-mediated 5 hmC modification represents a kind of diagnostic biomarkers of radiation-induced cognitive dysfunction. Targeting Tet-related epigenetic modification may be a novel therapeutic strategy for radiation-induced brain injury.
Collapse
Affiliation(s)
- Junjun Zhang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou City, 215000, PR China.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou City, 215000, PR China.,Suzhou Key Laboratory for Radiation Oncology, Suzhou City, 215000, PR China
| | - Junyan Li
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou City, 215000, PR China.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou City, 215000, PR China.,Suzhou Key Laboratory for Radiation Oncology, Suzhou City, 215000, PR China
| | - Yiwen Zhu
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou City, 215000, PR China.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou City, 215000, PR China.,Suzhou Key Laboratory for Radiation Oncology, Suzhou City, 215000, PR China
| | - Zhigang Miao
- Institute of Neuroscience, Soochow University, Suzhou City, 215000, PR China
| | - Ye Tian
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou City, 215000, PR China.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou City, 215000, PR China.,Suzhou Key Laboratory for Radiation Oncology, Suzhou City, 215000, PR China
| |
Collapse
|
27
|
Pan H, Wang H, Yu J, Huang X, Hao Y, Zhang C, Ji W, Yang M, Gong X, Wu X, Chang J. Near-infrared light remotely up-regulate autophagy with spatiotemporal precision via upconversion optogenetic nanosystem. Biomaterials 2019; 199:22-31. [PMID: 30735893 DOI: 10.1016/j.biomaterials.2019.01.042] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/15/2019] [Accepted: 01/30/2019] [Indexed: 12/31/2022]
Abstract
In vivo noninvasively manipulating biological functions by the mediation of biosafe near infrared (NIR) light is becoming increasingly popular. For these applications, upconversion rare-earth nanomaterial holds great promise as a novel photonic element, and has been widely adopted in optogenetics. In this article, an upconversion optogenetic nanosystem that was promised to achieve autophagy up-regulation with spatiotemporal precision was designed. The implantable, wireless, recyclable, less-invasive and biocompatible system worked via two separated parts: blue light-receptor optogenetics-autophagy upregulation plasmids, for protein import; upconversion rods-encapsulated flexible capsule (UCRs-capsule), for converting tissue-penetrative NIR light into local visible blue light. Results validated that this system could achieve up-regulation of autophagy in vitro (in both HeLa and 293T cell lines) and remotely penetrate tissue (∼3.5 mm) in vivo. Since autophagy serves at a central position in intracellular signalling pathways, which is correlative with diverse pathologies, we expect that this method could establish an upconversion material-based autophagy up-regulation strategy for fundamental and clinical applications.
Collapse
Affiliation(s)
- Huizhuo Pan
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Hanjie Wang
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China.
| | - Jingxian Yu
- School of Precision Instrument and Opto-Electronics Engineering, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Xian Huang
- School of Precision Instrument and Opto-Electronics Engineering, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Yafeng Hao
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Chaonan Zhang
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Wanying Ji
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Minye Yang
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Xiaoqun Gong
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Xiaoli Wu
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Jin Chang
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China.
| |
Collapse
|
28
|
Li B, Zhao J, Ma JX, Li GM, Zhang Y, Xing GS, Liu J, Ma XL. Overexpression of DNMT1 leads to hypermethylation of H19 promoter and inhibition of Erk signaling pathway in disuse osteoporosis. Bone 2018; 111:82-91. [PMID: 29555308 DOI: 10.1016/j.bone.2018.03.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 03/14/2018] [Accepted: 03/15/2018] [Indexed: 12/28/2022]
Abstract
Disuse osteoporosis (DOP) is a common complication of the lack of mechanical loading. The precise mechanism underlying DOP remains unknown, although epigenetic modifications may be a major cause. Recently, cumulative research has revealed that DNA methyltransferase (DNMT) proteins can catalyze the conversion of cytosine to 5-methylcytosine (5mC), altering the epigenetic state of DNA. Here, we report that DNMT1 expression and lncRNA-H19 methylation are upregulated in the femoral tissues of DOP rats, accompanied with inhibited Erk signaling pathway. Overexpression of DNMT1 in UMR-106 cells mimics 5mC enrichment in the H19 promoter, inhibition of Erk signaling and impairment of osteogenesis, which can be rescued by 5'-aza-deoxycytidine (5'-Aza) treatment. Moreover, local intramedullary injection of Dnmt1 siRNA (siDNMT1) in Sprague-Dawley (SD) rats abrogated disuse lncRNA-H19 (H19) downregulation, Erk signaling inhibition, histopathological changes, and bone microstructure declines in the distal femur in vivo. Therefore, our data identify for the first time a new signaling cascade in DOP: mechanical unloading causes upregulation of DNMT1 and hypermethylation of H19 promoter, which subsequently leads to downregulation of lncRNA-H19 and inhibition of the ERK signaling, suggesting a new potential therapeutic target.
Collapse
Affiliation(s)
- Bing Li
- Joint Department, Tianjin Hospital, Tianjin 300211, People's Republic of China
| | - Jie Zhao
- Orthopedic Department, Tianjin Hospital, Tianjin 300211, People's Republic of China
| | - Jian-Xiong Ma
- Orthopedic Research Institute, Tianjin Hospital, Tianjin 300050, People's Republic of China
| | - Guo-Min Li
- Graduate School, Tianjin Medical University, Tianjin 300070, People's Republic of China
| | - Yang Zhang
- Orthopedic Research Institute, Tianjin Hospital, Tianjin 300050, People's Republic of China
| | - Guo-Sheng Xing
- Orthopedic Research Institute, Tianjin Hospital, Tianjin 300050, People's Republic of China
| | - Jun Liu
- Joint Department, Tianjin Hospital, Tianjin 300211, People's Republic of China.
| | - Xin-Long Ma
- Joint Department, Tianjin Hospital, Tianjin 300211, People's Republic of China; Orthopedic Research Institute, Tianjin Hospital, Tianjin 300050, People's Republic of China.
| |
Collapse
|
29
|
Zhang H, Zhou F, Pan Z, Bu X, Wang Y, Chen F. 11β-hydroxysteroid dehydrogenases-2 decreases the apoptosis of MC3T3/MLO-Y4 cells induced by glucocorticoids. Biochem Biophys Res Commun 2017; 490:1399-1406. [PMID: 28698139 DOI: 10.1016/j.bbrc.2017.07.046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 07/08/2017] [Indexed: 12/28/2022]
Abstract
The aim of the present study was to confirm the role of 11β-hydroxysteroid dehydrogenases type 2(11β-HSD-2) in steroid induced osteonecrosis of the femoral head(SANFH). We cultured mouse bone-like cells (MLO-Y4) and mouse osteoblast-like cells (MC3T3-E1). After overexpressed 11β-HSD-2 successfully, we induced cell apoptosis by dexamethasone (DXM). The level of cell apoptosis, the expression of Bcl-2 in MLO-Y4 cells and the expression of Fas and caspase8 in MC3T3-E1 cells were detected. Then, we constructed 11β-HSD-2 siRNA plasmid and represented it on MLO-Y4/MC3T3-E1 Cells, to down-regulate the 11β-HSD-2 expression. After that, we used dexamethasone to induce cell apoptosis. The level of cell apoptosis, the expression of Bcl-2 in MLO-Y4 cells and the expression of Fas and caspase8 in MC3T3-E1 cells were detected again. In the overexpression model of cells, we found that the amount of cell apoptosis, the expression of Fas and caspase8 in MC3T3-E1 cells are lower than that of control groups. The amount of cell apoptosis, the expression of Fas and caspase8 in MC3T3-E1 cells were more than before when we reduced the expression of 11β-HSD-2. In our study, we concluded that 11β-HSD-2 plays an important role in the development of bone or osteoblast cell apoptosis, and the decreased expression of 11β-HSD-2 may aggravate steroid induced bone/osteoblast cell apoptosis.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Zhenyu Pan
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China.
| | - Xiangpeng Bu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Yaoqing Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Fan Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, PR China
| |
Collapse
|