1
|
Wen R, Huang R, Yang M, Yang J, Yi X. Regulation of protein arginine methyltransferase in osteoporosis: a narrative review. Front Cell Dev Biol 2025; 13:1453624. [PMID: 40342926 PMCID: PMC12058719 DOI: 10.3389/fcell.2025.1453624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 04/14/2025] [Indexed: 05/11/2025] Open
Abstract
Osteoporosis (OP), a systemic bone disease characterised by increased bone fragility and susceptibility to fracture, is mainly caused by a decline in bone mineral density (BMD) and quality caused by an imbalance between bone formation and resorption. Protein arginine methyltransferases (PRMTs) are epigenetic factors and post-translational modification (PTM) enzymes participating in various biological processes, including mRNA splicing, DNA damage repair, transcriptional regulation, and cell signalling. They act by catalysing the transfer and modification of arginine residues and, thus, have become therapeutic targets for OP. In-depth studies have found that these enzymes also play key roles in bone matrix protein metabolism, skeletal cell proliferation and differentiation, and signal pathway regulation to regulate bone formation, bone resorption balance, or both and jointly maintain bone health and stability. However, the expression changes and mechanisms of action of multiple members of the PRMT family differ in OP. Therefore, this paper discusses the biological functions, mechanisms of action, and influencing factors of PRMTs in OP, which is expected to provide a new understanding of the pathogenesis of OP. Furthermore, we present theoretical support for the development of more precise and effective treatment strategies as well as for further study of the molecular mechanisms of PRMTs.
Collapse
Affiliation(s)
| | | | | | | | - Xuejie Yi
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| |
Collapse
|
2
|
Zhang Y, Qing J, Li Y, Gao X, Lu D, Wang Y, Gu L, Zhang H, Li Z, Wang X, Zhou Y, Zhang P. PRMT7-Mediated PTEN Activation Enhances Bone Regeneration in Female Mice. Int J Mol Sci 2025; 26:2981. [PMID: 40243588 PMCID: PMC11988880 DOI: 10.3390/ijms26072981] [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: 02/18/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Epigenetic regulation provides new insights into the mechanisms of osteogenic differentiation and identifies potential targets for treating bone-related diseases. However, the specific regulatory networks and mechanisms involved still need further investigation. In this study, we identify PRMT7 as a novel epigenetic regulator of mesenchymal stem cells (MSCs) osteogenic commitment. Conditional knockout of Prmt7 in mice reveals a significant impairment in osteogenesis and bone regeneration, specifically in females, affecting both femurs and mandibles, with no noticeable effect in males. Mechanistically, PRMT7 modulates MSCs osteogenic differentiation by activating PTEN. Specifically, PRMT7 enhances PTEN transcription by increasing H3R2me1 levels at the PTEN promoter. Additionally, PRMT7 interacts with the PTEN protein and stabilizes nuclear PTEN, revealing an unprecedented pathway. Notably, overexpression of PTEN alleviates the osteogenic deficits observed in Prmt7-deficient mice. This research establishes PRMT7 as a potential therapeutic target for promoting bone formation/regeneration and offers novel molecular insights into the PRMT7-PTEN regulatory axis, underscoring its significance in bone biology and regenerative medicine.
Collapse
Affiliation(s)
- Yingfei Zhang
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Jia Qing
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Yang Li
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Xin Gao
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Dazhuang Lu
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Yiyang Wang
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Lanxin Gu
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Hui Zhang
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Zechuan Li
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Xu Wang
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| | - Ping Zhang
- Department of Prosthodontics, Peking University Hospital of Stomatology, Beijing 100081, China; (Y.Z.); (J.Q.); (Y.L.); (X.G.); (D.L.); (Y.W.); (L.G.); (H.Z.); (Z.L.); (X.W.)
- National Clinical Research Center for Oral Diseases, Peking University Hospital of Stomatology, Beijing 100081, China
| |
Collapse
|
3
|
Yousefi F, Foster LA, Selim OA, Zhao C. Integrating Physical and Biochemical Cues for Muscle Engineering: Scaffolds and Graft Durability. Bioengineering (Basel) 2024; 11:1245. [PMID: 39768063 PMCID: PMC11673930 DOI: 10.3390/bioengineering11121245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Muscle stem cells (MuSCs) are essential for skeletal muscle regeneration, influenced by a complex interplay of mechanical, biochemical, and molecular cues. Properties of the extracellular matrix (ECM) such as stiffness and alignment guide stem cell fate through mechanosensitive pathways, where forces like shear stress translate into biochemical signals, affecting cell behavior. Aging introduces senescence which disrupts the MuSC niche, leading to reduced regenerative capacity via epigenetic alterations and metabolic shifts. Transplantation further challenges MuSC viability, often resulting in fibrosis driven by dysregulated fibro-adipogenic progenitors (FAPs). Addressing these issues, scaffold designs integrated with pharmacotherapy emulate ECM environments, providing cues that enhance graft functionality and endurance. These scaffolds facilitate the synergy between mechanotransduction and intracellular signaling, optimizing MuSC proliferation and differentiation. Innovations utilizing human pluripotent stem cell-derived myogenic progenitors and exosome-mediated delivery exploit bioactive properties for targeted repair. Additionally, 3D-printed and electrospun scaffolds with adjustable biomechanical traits tackle scalability in treating volumetric muscle loss. Advanced techniques like single-cell RNA sequencing and high-resolution imaging unravel muscle repair mechanisms, offering precise mapping of cellular interactions. Collectively, this interdisciplinary approach fortifies tissue graft durability and MuSC maintenance, propelling therapeutic strategies for muscle injuries and degenerative diseases.
Collapse
Affiliation(s)
- Farbod Yousefi
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| | - Lauren Ann Foster
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
- Atlanta Veterans Affairs Medical Center, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Omar A. Selim
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| | - Chunfeng Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| |
Collapse
|
4
|
Zhang J, Liu H, Liu Y, Luo E, Liu S. Unlocking the potential of histone modification in regulating bone metabolism. Biochimie 2024; 227:286-298. [PMID: 39154977 DOI: 10.1016/j.biochi.2024.08.004] [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: 05/23/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 08/20/2024]
Abstract
Bone metabolism plays a crucial role in maintaining normal bone tissue homeostasis and function. Imbalances between bone formation and resorption can lead to osteoporosis, osteoarthritis, and other bone diseases. The dynamic and complex process of bone remodeling is driven by various factors, including epigenetics. Histone modification, one of the most important and well-studied components of epigenetic regulation, has emerged as a promising area of research in bone metabolism. Different histone proteins and modification sites exert diverse effects on osteogenesis and osteoclastogenesis. In this review, we summarize recent progress in understanding histone modifications in bone metabolism, including specific modification sites and potential regulatory enzymes. Comprehensive knowledge of histone modifications in bone metabolism could reveal new therapeutic targets and treatment strategies for bone diseases.
Collapse
Affiliation(s)
- Jiayuan Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yao Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shibo Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
5
|
Yang J, Yang S, Ge X, Yuan L, Qi Y, Huang Z, Yang G, Zhang R. Protein arginine methyltransferase 5 in osteoblasts promotes the healing of extraction sockets. Oral Dis 2024; 30:3951-3961. [PMID: 38297969 DOI: 10.1111/odi.14826] [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: 06/10/2023] [Revised: 11/07/2023] [Accepted: 11/17/2023] [Indexed: 02/02/2024]
Abstract
OBJECTIVES To explore the effect of protein arginine methyltransferase 5 (PRMT5) on tooth extraction sockets healing, we established an extraction sockets model in osteoblast-conditional Prmt5 knockout mice. The results provided clues for promoting extraction sockets healing in clinical settings. MATERIALS AND METHODS Maxillary first molars were extracted from 6 to 8-week-old mice to establish an extraction fossa model. Microcomputed tomography (Micro-CT), histology, and immunostaining assays were performed on samples harvested at 3-, 7-, and 14-day post-extraction. Prmt5-silenced cell lines were employed to explore the regulatory mechanisms underlying the osteigenic differentiation. RESULTS PRMT5 expression was higher in the early stage of socket healing. Micro-CT analysis showed that the percentage of new bone in the extraction sockets was lower in OC-Cre; Prmt5fl/fl mice than in the control group, consistent with Masson staining. We found that, Prmt5 deficiency delayed the osteogenesis during extraction socket healing, which might be achieved through the decrease of H4R3me2s in the Sp7 promoter region. CONCLUSION PRMT5 in osteoblasts may promote the differentiation of osteoblasts by regulating the Sp7 promoter H4R3me2s and participate in the healing of tooth extraction sockets.
Collapse
Affiliation(s)
- Jie Yang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
- Department of Oral Pathology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Shurong Yang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
- Department of Endodontics, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, China
| | - Xuejun Ge
- Department of Oral Pathology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Lu Yuan
- Department of Oral Pathology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Yini Qi
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Zhen Huang
- Shanxi Health Vocational College, Taiyuan, China
| | - Guan Yang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Ran Zhang
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Research Unit of Precision Pathologic Diagnosis in Tumors of the Oral and Maxillofacial Regions, Chinese Academy of Medical Sciences (2019RU034), Beijing, China
| |
Collapse
|
6
|
Vuong TA, Zhang Y, Kim J, Leem YE, Kang JS. Prmt7 is required for the osteogenic differentiation of mesenchymal stem cells via modulation of BMP signaling. BMB Rep 2024; 57:330-335. [PMID: 38627951 PMCID: PMC11289507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/13/2023] [Accepted: 01/15/2024] [Indexed: 08/03/2024] Open
Abstract
Arginine methylation, which is catalyzed by protein arginine methyltransferases (Prmts), is known to play a key role in various biological processes. However, the function of Prmts in osteogenic differentiation of mesenchymal stem cells (MSCs) has not been clearly understood. In the current study, we attempted to elucidate a positive role of Prmt7 in osteogenic differentiation. Prmt7-depleted C3H/10T1/2 cells or bone marrow mesenchymal stem cells (BMSCs) showed the attenuated expression of osteogenic specific genes and Alizarin red staining compared to the wild-type cells. Furthermore, we found that Prmt7 deficiency reduced the activation of bone morphogenetic protein (BMP) signaling cascade, which is essential for the regulation of cell fate commitment and osteogenesis. Taken together, our data indicate that Prmt7 plays important regulatory roles in osteogenic differentiation. [BMB Reports 2024; 57(7): 330-335].
Collapse
Affiliation(s)
- Tuan Anh Vuong
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Yan Zhang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - June Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Young-Eun Leem
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| |
Collapse
|
7
|
Chen C, Ding Y, Huang Q, Zhang C, Zhao Z, Zhou H, Li D, Zhou G. Relationship between arginine methylation and vascular calcification. Cell Signal 2024; 119:111189. [PMID: 38670475 DOI: 10.1016/j.cellsig.2024.111189] [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: 01/30/2024] [Revised: 04/11/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
In patients on maintenance hemodialysis (MHD), vascular calcification (VC) is an independent predictor of cardiovascular disease (CVD), which is the primary cause of death in chronic kidney disease (CKD). The main component of VC in CKD is the vascular smooth muscle cells (VSMCs). VC is an ordered, dynamic activity. Under the stresses of oxidative stress and calcium-‑phosphorus imbalance, VSMCs undergo osteogenic phenotypic transdifferentiation, which promotes the formation of VC. In addition to traditional epigenetics like RNA and DNA control, post-translational modifications have been discovered to be involved in the regulation of VC in recent years. It has been reported that the process of osteoblast differentiation is impacted by catalytic histone or non-histone arginine methylation. Its function in the osteogenic process is comparable to that of VC. Thus, we propose that arginine methylation regulates VC via many signaling pathways, including as NF-B, WNT, AKT/PI3K, TGF-/BMP/SMAD, and IL-6/STAT3. It might also regulate the VC-related calcification regulatory factors, oxidative stress, and endoplasmic reticulum stress. Consequently, we propose that arginine methylation regulates the calcification of the arteries and outline the regulatory mechanisms involved.
Collapse
Affiliation(s)
- Chen Chen
- Department of Nephrology, Shengjing Hospital, China Medical University, China
| | - Yuanyuan Ding
- Department of Pain Management, Shengjing Hospital, China Medical University, China
| | - Qun Huang
- Department of Nephrology, Shengjing Hospital, China Medical University, China
| | - Chen Zhang
- Department of Nephrology, Shengjing Hospital, China Medical University, China
| | - Zixia Zhao
- Department of Nephrology, Shengjing Hospital, China Medical University, China
| | - Hua Zhou
- Department of Nephrology, Shengjing Hospital, China Medical University, China
| | - Detian Li
- Department of Nephrology, Shengjing Hospital, China Medical University, China
| | - Guangyu Zhou
- Department of Nephrology, Shengjing Hospital, China Medical University, China.
| |
Collapse
|
8
|
Hoang PM, Torre D, Jaynes P, Ho J, Mohammed K, Alvstad E, Lam WY, Khanchandani V, Lee JM, Toh CMC, Lee RX, Anbuselvan A, Lee S, Sebra RP, Martin J. Walsh, Marazzi I, Kappei D, Guccione E, Jeyasekharan AD. A PRMT5-ZNF326 axis mediates innate immune activation upon replication stress. SCIENCE ADVANCES 2024; 10:eadm9589. [PMID: 38838142 PMCID: PMC11804791 DOI: 10.1126/sciadv.adm9589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/25/2024] [Indexed: 06/07/2024]
Abstract
DNA replication stress (RS) is a widespread phenomenon in carcinogenesis, causing genomic instability and extensive chromatin alterations. DNA damage leads to activation of innate immune signaling, but little is known about transcriptional regulators mediating such signaling upon RS. Using a chemical screen, we identified protein arginine methyltransferase 5 (PRMT5) as a key mediator of RS-dependent induction of interferon-stimulated genes (ISGs). This response is also associated with reactivation of endogenous retroviruses (ERVs). Using quantitative mass spectrometry, we identify proteins with PRMT5-dependent symmetric dimethylarginine (SDMA) modification induced upon RS. Among these, we show that PRMT5 targets and modulates the activity of ZNF326, a zinc finger protein essential for ISG response. Our data demonstrate a role for PRMT5-mediated SDMA in the context of RS-induced transcriptional induction, affecting physiological homeostasis and cancer therapy.
Collapse
Affiliation(s)
- Phuong Mai Hoang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Denis Torre
- Center for OncoGenomics and Innovative Therapeutics (COGIT), Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetic and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Patrick Jaynes
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Jessica Ho
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Kevin Mohammed
- Center for OncoGenomics and Innovative Therapeutics (COGIT), Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetic and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Erik Alvstad
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, CA 92697, USA
| | - Wan Yee Lam
- Department of Genetic and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Vartika Khanchandani
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Jie Min Lee
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Chin Min Clarissa Toh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Rui Xue Lee
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Akshaya Anbuselvan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Sukchan Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, South Korea
| | - Robert P. Sebra
- Department of Genetic and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Martin J. Walsh
- Department of Genetic and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ivan Marazzi
- Department of Biological Chemistry, University of California Irvine, Irvine, CA 92697, USA
- Center for Epigenetics and Metabolism, University of California Irvine, Irvine, CA 92697, USA
| | - Dennis Kappei
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ernesto Guccione
- Center for OncoGenomics and Innovative Therapeutics (COGIT), Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetic and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Anand D. Jeyasekharan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Haematology-Oncology, National University Hospital, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
9
|
Yi SJ, Lim J, Kim K. Exploring epigenetic strategies for the treatment of osteoporosis. Mol Biol Rep 2024; 51:398. [PMID: 38453825 DOI: 10.1007/s11033-024-09353-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/14/2024] [Indexed: 03/09/2024]
Abstract
The worldwide trend toward an aging population has resulted in a higher incidence of chronic conditions, such as osteoporosis. Osteoporosis, a prevalent skeletal disorder characterized by decreased bone mass and increased fracture risk, encompasses primary and secondary forms, each with distinct etiologies. Mechanistically, osteoporosis involves an imbalance between bone resorption by osteoclasts and bone formation by osteoblasts. Current pharmacological interventions for osteoporosis, such as bisphosphonates, denosumab, and teriparatide, aim to modulate bone turnover and preserve bone density. Hormone replacement therapy and lifestyle modifications are also recommended to manage the condition. While current medications offer therapeutic options, they are not devoid of limitations. Recent studies have highlighted the importance of epigenetic mechanisms, including DNA methylation and histone modifications, in regulating gene expression during bone remodeling. The use of epigenetic drugs, or epidrugs, to target these mechanisms offers a promising avenue for therapeutic intervention in osteoporosis. In this review, we comprehensively examine the recent advancements in the application of epidrugs for treating osteoporosis.
Collapse
Affiliation(s)
- Sun-Ju Yi
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jaeho Lim
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Kyunghwan Kim
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.
| |
Collapse
|
10
|
Zhang Y, Li X, Zhang S, Li J, Liu M, Lu Y, Han J. Role of IFITM2 in osteogenic differentiation of C3H10T1/2 mesenchymal stem cells. Intractable Rare Dis Res 2024; 13:42-50. [PMID: 38404731 PMCID: PMC10883848 DOI: 10.5582/irdr.2023.01108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 02/27/2024] Open
Abstract
Interferon-inducible transmembrane (IFITM) are a family of small proteins localized to plasma and endolysosomal membranes. Their functions beyond restricting viral entry and replication have been revealed in recent years. IFITM5 is involved in bone mineralization and is an osteogenic cell surface marker. IFITM1 and 3 interact with desmin and myosin, and are involved in myogenic differentiation. This study found upregulation of Ifitm2 during osteogenic differentiation of C3H10T1/2 cells. This positively correlated to the expression of osteogenic differentiation markers Col1a1, Alp, Runx2, and Ocn. Knockdown of Ifitm2 by siRNAs inhibited osteogenic differentiation, calcium deposition, and osteogenic marker expression of C3H10T1/2 cells. The osteoblast transcriptome revealed that knocking down Ifitm2 affected the expression Wnt signaling pathway-related genes, including Wnt family members, their receptors Lrp, Frizzled, and Lgr, and transmembrane molecule Rnf43 that suppresses the Wnt signaling pathway. Luciferase assays indicated enhancement of canonical Wnt signaling pathways by Ifitm2 overexpression. Furthermore, IFITM2 was colocalized in the metaphyseal bone and growth plate of the mouse tibial bone with SP7, a transcription factor essential for osteoblast differentiation and bone formation. These findings reveal a possible novel function and potential mechanisms of Ifitm2 in osteogenic differentiation.
Collapse
Affiliation(s)
- Yongtao Zhang
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
| | - Xiangdong Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Shanshan Zhang
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
| | - Junfeng Li
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
| | - Meilin Liu
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
| | - Yanqin Lu
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| | - Jinxiang Han
- Key Laboratory for Biotech Drugs of the National Health Commission, Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, Shandong, China
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Ji'nan, Shandong, China
| |
Collapse
|
11
|
Kumar D, Jain S, Coulter DW, Joshi SS, Chaturvedi NK. PRMT5 as a Potential Therapeutic Target in MYC-Amplified Medulloblastoma. Cancers (Basel) 2023; 15:5855. [PMID: 38136401 PMCID: PMC10741595 DOI: 10.3390/cancers15245855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
MYC amplification or overexpression is most common in Group 3 medulloblastomas and is positively associated with poor clinical outcomes. Recently, protein arginine methyltransferase 5 (PRMT5) overexpression has been shown to be associated with tumorigenic MYC functions in cancers, particularly in brain cancers such as glioblastoma and medulloblastoma. PRMT5 regulates oncogenes, including MYC, that are often deregulated in medulloblastomas. However, the role of PRMT5-mediated post-translational modification in the stabilization of these oncoproteins remains poorly understood. The potential impact of PRMT5 inhibition on MYC makes it an attractive target in various cancers. PRMT5 inhibitors are a promising class of anti-cancer drugs demonstrating preclinical and preliminary clinical efficacies. Here, we review the publicly available preclinical and clinical studies on PRMT5 targeting using small molecule inhibitors and discuss the prospects of using them in medulloblastoma therapy.
Collapse
Affiliation(s)
- Devendra Kumar
- Department of Pediatrics, Division of Hematology and Oncology, University of Nebraska Medical Center, Omaha, NE 69198, USA; (D.K.); (S.J.); (D.W.C.)
| | - Stuti Jain
- Department of Pediatrics, Division of Hematology and Oncology, University of Nebraska Medical Center, Omaha, NE 69198, USA; (D.K.); (S.J.); (D.W.C.)
| | - Don W. Coulter
- Department of Pediatrics, Division of Hematology and Oncology, University of Nebraska Medical Center, Omaha, NE 69198, USA; (D.K.); (S.J.); (D.W.C.)
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 69198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 69198, USA
| | - Shantaram S. Joshi
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 69198, USA;
| | - Nagendra K. Chaturvedi
- Department of Pediatrics, Division of Hematology and Oncology, University of Nebraska Medical Center, Omaha, NE 69198, USA; (D.K.); (S.J.); (D.W.C.)
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 69198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 69198, USA
| |
Collapse
|
12
|
Zhang K, Li C, Sun J, Tian X. PRMT5 inhibition ameliorates inflammation and promotes the osteogenic differentiation of LPS‑induced periodontal stem cells via STAT3/NF‑κB signaling. Exp Ther Med 2023; 25:264. [PMID: 37206565 PMCID: PMC10189754 DOI: 10.3892/etm.2023.11963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/06/2022] [Indexed: 05/21/2023] Open
Abstract
It has been reported that protein arginine methyltransferase 5 (PRMT5) serves a significant role in osteogenic differentiation and inflammatory response. Nevertheless, its role in periodontitis as well as its underlying mechanism remain to be elucidated. The aim of the present study was to explore the role of PRMT5 in periodontitis and whether PRMT5 could reduce liposaccharide (LPS)-induced inflammation of human periodontal ligament stem cells (hPDLSCs) and promote osteogenic differentiation through STAT3/NF-κB signaling. In the current study, the expression levels of PRMT5 were determined in LPS-induced hPDLSCs by reverse transcription-quantitative PCR and western blot analysis. ELISA and western blot analysis were employed to assess the secretion and expression levels of inflammatory factors, respectively. The osteogenic differentiation and mineralization potential of hPDLSCs were evaluated using alkaline phosphatase (ALP) activity assay, Alizarin red staining and western blot analysis. Additionally, western blot analysis was applied to determine the expression levels of the STAT3/NF-κB signaling pathway-related proteins. The results showed that the expression levels of PRMT5 were significantly enhanced in LPS-induced hPDLSCs. Additionally, PRMT5 knockdown reduced the contents of IL-1β, IL-6, TNF-α, inducible nitric oxide synthase and cyclooxygenase-2. PRMT5 depletion also enhanced ALP activity, improved the mineralization ability and upregulated bone morphogenetic protein 2, osteocalcin and runt-related transcription factor 2 in LPS-induced hPDLSCs. Furthermore, PRMT5 knockdown inhibited inflammation and promoted the osteogenic differentiation of hPDLSCs via blocking the activation of the STAT3/NF-κB signaling pathway. In conclusion, PRMT5 inhibition suppressed LPS-induced inflammation and accelerated osteogenic differentiation in hPDLSCs via regulating STAT3/NF-κB signaling, thus providing a potential targeted therapy for the improvement of periodontitis.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Cariology and Endodontics, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
- School of Stomatology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Changshun Li
- Department of Cariology and Endodontics, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
- School of Stomatology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Jian Sun
- Department of Cariology and Endodontics, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
- School of Stomatology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Xiaobei Tian
- Department of Cariology and Endodontics, The Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
- School of Stomatology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
- Correspondence to: Professor Xiaobei Tian, Department of Cariology and Endodontics, The Affiliated Stomatological Hospital of Xuzhou Medical University, 130 Huaihai West Road, Xuzhou, Jiangsu 221004, P.R. China
| |
Collapse
|
13
|
Dong XL, Yuan BH, Yu SZ, Liu H, Pan XH, Sun J, Pan LL. Adriamycin induces cardiac fibrosis in mice via PRMT5-mediated cardiac fibroblast activation. Acta Pharmacol Sin 2023; 44:573-583. [PMID: 36056082 PMCID: PMC9958096 DOI: 10.1038/s41401-022-00963-x] [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: 03/20/2022] [Accepted: 07/21/2022] [Indexed: 11/08/2022]
Abstract
Long-term treatment with adriamycin (ADR) is associated with higher incidences of cumulative cardiotoxicity manifest as heart failure. ADR-induced cardiomyopathy is characterized by extensive fibrosis that is caused by cardiac fibroblast activation. To date, however, no specific treatment is available to alleviate ADR-induced cardiotoxicity. Protein arginine methyltransferase 5 (PRMT5), a major enzyme responsible for methylation of arginine, regulates numerous cellular processes such as cell differentiation. In the present study we investigated the role of PRMT5 in cardiac fibrosis. Mice were administered ADR (3 mg/kg, i.p., every 2 days) for 2 weeks. We showed that aberrant PRMT5 expression was largely co-localized with α-SMA-positive activated cardiac fibroblasts in ADR-injected mice and in ADR-treated cardiac fibroblasts in vitro. PRMT5-overexpression exacerbated, whereas PRMT5 knockdown alleviated ADR-induced cardiac fibrosis in vivo and TGF-β1-induced cardiac fibroblast activation in vitro. We demonstrated that PRMT5-overexpression enhanced methylated-Smad3 levels in vivo and in vitro. Pretreatment with a specific PRMT5 inhibitor EPZ015666 (5 nM) or overexpression of a catalytically inactive mutant of PRMT5, PRMT5(E444Q), reduced PRMT5-induced methylation of Smad3, thus suppressing PRMT5-mediated cardiac fibroblast activation in vitro. Furthermore, ADR activated cardiac fibroblasts was depending on autocrine TGF-β1. Taken together, our results demonstrate that PRMT5 promotes ADR-induced cardiac fibrosis via activating cardiac fibroblasts, suggesting that it may be a potential therapeutic target of ADR-caused cardiotoxicity.
Collapse
Affiliation(s)
- Xiao-Liang Dong
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Bao-Hui Yuan
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Sheng-Zhou Yu
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - He Liu
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Xiao-Hua Pan
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Jia Sun
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.
| | - Li-Long Pan
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
14
|
Yu J, Yu C, Bayliss G, Zhuang S. Protein arginine methyltransferases in renal development, injury, repair, and fibrosis. Front Pharmacol 2023; 14:1123415. [PMID: 36817133 PMCID: PMC9935595 DOI: 10.3389/fphar.2023.1123415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Protein arginine methyltransferases (PRMTs) methylate a range of histone and non-histone substrates and participate in multiple biological processes by regulating gene transcription and post-translational modifications. To date, most studies on PRMTs have focused on their roles in tumors and in the physiological and pathological conditions of other organs. Emerging evidence indicates that PRMTs are expressed in the kidney and contribute to renal development, injury, repair, and fibrosis. In this review, we summarize the role and the mechanisms of PRMTs in regulating these renal processes and provide a perspective for future clinical applications.
Collapse
Affiliation(s)
- Jianjun Yu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chao Yu
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Georgia Bayliss
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, United States
| | - Shougang Zhuang
- Department of Nephrology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, RI, United States
| |
Collapse
|
15
|
BRG1: Promoter or Suppressor of Cancer? The Outcome of BRG1's Interaction with Specific Cellular Pathways. Int J Mol Sci 2023; 24:ijms24032869. [PMID: 36769189 PMCID: PMC9917617 DOI: 10.3390/ijms24032869] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
BRG1 is one of two catalytic subunits of the SWI/SNF ATP-dependent chromatin-remodeling complex. In cancer, it has been hypothesized that BRG1 acts as a tumor suppressor. Further study has shown that, under certain circumstances, BRG1 acts as an oncogene. Targeted knockout of BRG1 has proven successful in most cancers in suppressing tumor growth and proliferation. Furthermore, BRG1 effects cancer proliferation in oncogenic KRAS mutated cancers, with varying directionality. Thus, dissecting BRG1's interaction with various cellular pathways can highlight possible intermediates that can facilitate the design of different treatment methods, including BRG1 inhibition. Autophagy and apoptosis are two important cellular responses to stress. BRG1 plays a direct role in autophagy and apoptosis and likely promotes autophagy and suppresses apoptosis, supporting unfettered cancer growth. PRMT5 inhibits transcription by interacting with ATP-dependent chromatin remodeling complexes, such as SWI/SNF. When PRMT5 associates with the SWI/SNF complex, including BRG1, it represses tumor suppressor genes. The Ras/Raf/MAPK/ERK1/2 pathway in cancers is a signal transduction pathway involved in the transcription of genes related to cancer survival. BRG1 has been shown to effect KRAS-driven cancer growth. BRG1 associates with several proteins within the signal transduction pathway. In this review, we analyze BRG1 as a promising target for cancer inhibition and possible synergy with other cancer treatments.
Collapse
|
16
|
Ruiz UEA, Santos IA, Grosche VR, Fernandes RS, de Godoy AS, Torres JDA, Freire MCLC, Mesquita NCDMR, Guevara-Vega M, Nicolau-Junior N, Sabino-Silva R, Mineo TWP, Oliva G, Jardim ACG. Imidazonaphthyridine effects on Chikungunya virus replication: Antiviral activity by dependent and independent of interferon type 1 pathways. Virus Res 2023; 324:199029. [PMID: 36565816 PMCID: PMC10194360 DOI: 10.1016/j.virusres.2022.199029] [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: 10/05/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
The Chikungunya virus (CHIKV) causes Chikungunya fever, a disease characterized by symptoms such as arthralgia/polyarthralgia. Currently, there are no antivirals approved against CHIKV, emphasizing the need to develop novel therapies. The imidazonaphthyridine compound (RO8191), an interferon-α (IFN-α) agonist, was reported as a potent inhibitor of HCV. Here RO8191 was investigated for its potential to inhibit CHIKV replication in vitro. RO8191 inhibited CHIKV infection in BHK-21 and Vero-E6 cells with a selectivity index (SI) of 12.3 and 37.3, respectively. Additionally, RO8191 was capable to protect cells against CHIKV infection, inhibit entry by virucidal activity, and strongly impair post-entry steps of viral replication. An effect of RO8191 on CHIKV replication was demonstrated in BHK-21 through type-1 IFN production mechanism and in Vero-E6 cells which has a defective type-1 IFN production, also suggesting a type-1 IFN independent mode of action. Molecular docking calculations demonstrated interactions of RO8191 with the CHIKV E proteins, corroborated by the ATR-FTIR assay, and with non-structural proteins, supported by the CHIKV-subgenomic replicon cells assay.
Collapse
Affiliation(s)
| | - Igor Andrade Santos
- Institute of Biomedical Science, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil
| | - Victória Riquena Grosche
- Institute of Biomedical Science, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil; Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), Campus São José do Rio Preto, SP, Brazil
| | | | | | | | | | | | - Marco Guevara-Vega
- Institute of Biomedical Science, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil
| | - Nilson Nicolau-Junior
- Institute of Biotechnology, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil
| | - Robinson Sabino-Silva
- Institute of Biomedical Science, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil
| | | | - Glaucius Oliva
- Sao Carlos Institute of Physics, University of Sao Paulo (USP), São Carlos, SP, Brazil
| | - Ana Carolina Gomes Jardim
- Institute of Biomedical Science, Federal University of Uberlândia (UFU), Uberlândia, MG, Brazil; Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), Campus São José do Rio Preto, SP, Brazil.
| |
Collapse
|
17
|
Semenova D, Zabirnyk A, Lobov A, Boyarskaya N, Kachanova O, Uspensky V, Zainullina B, Denisov E, Gerashchenko T, Kvitting JPE, Kaljusto ML, Thiede B, Kostareva A, Stensløkken KO, Vaage J, Malashicheva A. Multi-omics of in vitro aortic valve calcification. Front Cardiovasc Med 2022; 9:1043165. [PMID: 36407442 PMCID: PMC9669078 DOI: 10.3389/fcvm.2022.1043165] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 09/23/2022] [Indexed: 09/10/2023] Open
Abstract
Heart valve calcification is an active cellular and molecular process that partly remains unknown. Osteogenic differentiation of valve interstitial cells (VIC) is a central mechanism in calcific aortic valve disease (CAVD). Studying mechanisms in CAVD progression is clearly needed. In this study, we compared molecular mechanisms of osteogenic differentiation of human VIC isolated from healthy donors or patients with CAVD by RNA-seq transcriptomics in early timepoint (48 h) and by shotgun proteomics at later timepoint (10th day). Bioinformatic analysis revealed genes and pathways involved in the regulation of VIC osteogenic differentiation. We found a high amount of stage-specific differentially expressed genes and good accordance between transcriptomic and proteomic data. Functional annotation of differentially expressed proteins revealed that osteogenic differentiation of VIC involved many signaling cascades such as: PI3K-Akt, MAPK, Ras, TNF signaling pathways. Wnt, FoxO, and HIF-1 signaling pathways were modulated only at the early timepoint and thus probably involved in the commitment of VIC to osteogenic differentiation. We also observed a significant shift of some metabolic pathways in the early stage of VIC osteogenic differentiation. Lentiviral overexpression of one of the most upregulated genes (ZBTB16, PLZF) increased calcification of VIC after osteogenic stimulation. Analysis with qPCR and shotgun proteomics suggested a proosteogenic role of ZBTB16 in the early stages of osteogenic differentiation.
Collapse
Affiliation(s)
- Daria Semenova
- Institute of Cytology Russian Academy of Science, St. Petersburg, Russia
- Almazov National Medical Research Center Russia, St. Petersburg, Russia
| | - Arsenii Zabirnyk
- Heart Physiology Research Group, Division of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Oslo University Hospital, Oslo, Norway
| | - Arseniy Lobov
- Institute of Cytology Russian Academy of Science, St. Petersburg, Russia
| | | | - Olga Kachanova
- Almazov National Medical Research Center Russia, St. Petersburg, Russia
| | - Vladimir Uspensky
- Almazov National Medical Research Center Russia, St. Petersburg, Russia
| | - Bozhana Zainullina
- Centre for Molecular and Cell Technologies, St. Petersburg State University, St. Petersburg, Russia
| | - Evgeny Denisov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Tatiana Gerashchenko
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - John-Peder Escobar Kvitting
- Heart Physiology Research Group, Division of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Oslo University Hospital, Oslo, Norway
| | | | - Bernd Thiede
- Heart Physiology Research Group, Division of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Anna Kostareva
- Almazov National Medical Research Center Russia, St. Petersburg, Russia
| | - Kåre-Olav Stensløkken
- Heart Physiology Research Group, Division of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jarle Vaage
- Heart Physiology Research Group, Division of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Oslo University Hospital, Oslo, Norway
| | - Anna Malashicheva
- Institute of Cytology Russian Academy of Science, St. Petersburg, Russia
| |
Collapse
|
18
|
Sun P, Huang T, Huang C, Wang Y, Tang D. Role of histone modification in the occurrence and development of osteoporosis. Front Endocrinol (Lausanne) 2022; 13:964103. [PMID: 36093077 PMCID: PMC9458911 DOI: 10.3389/fendo.2022.964103] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoporosis is a systemic degenerative bone disease characterized by low bone mass and damage to bone microarchitecture, which increases bone fragility and susceptibility to fracture. The risk of osteoporosis increases with age; with the aging of the global population, osteoporosis is becoming more prevalent, adding to the societal healthcare burden. Histone modifications such as methylation, acetylation, ubiquitination, and ADP-ribosylation are closely related to the occurrence and development of osteoporosis. This article reviews recent studies on the role of histone modifications in osteoporosis. The existing evidence indicates that therapeutic targeting of these modifications to promote osteogenic differentiation and bone formation may be an effective treatment for this disease.
Collapse
Affiliation(s)
- Pan Sun
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tingrui Huang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Huang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yongjun Wang
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Yongjun Wang, ; Dezhi Tang,
| | - Dezhi Tang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Yongjun Wang, ; Dezhi Tang,
| |
Collapse
|
19
|
Xu J, Richard S. Cellular pathways influenced by protein arginine methylation: Implications for cancer. Mol Cell 2021; 81:4357-4368. [PMID: 34619091 PMCID: PMC8571027 DOI: 10.1016/j.molcel.2021.09.011] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/24/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023]
Abstract
Arginine methylation is an influential post-translational modification occurring on histones, RNA binding proteins, and many other cellular proteins, affecting their function by altering their protein-protein and protein-nucleic acid interactions. Recently, a wealth of information has been gathered, implicating protein arginine methyltransferases (PRMTs), enzymes that deposit arginine methylation, in transcription, pre-mRNA splicing, DNA damage signaling, and immune signaling with major implications for cancer therapy, especially immunotherapy. This review summarizes this recent progress and the current state of PRMT inhibitors, some in clinical trials, as promising drug targets for cancer.
Collapse
Affiliation(s)
- Jian Xu
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA; Department of Biochemistry and Molecular Medicine, Norris Comprehensive Cancer Center, and Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Stéphane Richard
- Segal Cancer Center, Lady Davis Institute for Medical Research, Gerald Bronfman Department of Oncology, and Departments of Medicine, Human Genetics, and Biochemistry, McGill University, Montréal, QC H3T 1E2, Canada.
| |
Collapse
|
20
|
Protein arginine methylation: from enigmatic functions to therapeutic targeting. Nat Rev Drug Discov 2021; 20:509-530. [PMID: 33742187 DOI: 10.1038/s41573-021-00159-8] [Citation(s) in RCA: 248] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2021] [Indexed: 02/06/2023]
Abstract
Protein arginine methyltransferases (PRMTs) are emerging as attractive therapeutic targets. PRMTs regulate transcription, splicing, RNA biology, the DNA damage response and cell metabolism; these fundamental processes are altered in many diseases. Mechanistically understanding how these enzymes fuel and sustain cancer cells, especially in specific metabolic contexts or in the presence of certain mutations, has provided the rationale for targeting them in oncology. Ongoing inhibitor development, facilitated by structural biology, has generated tool compounds for the majority of PRMTs and enabled clinical programmes for the most advanced oncology targets, PRMT1 and PRMT5. In-depth mechanistic investigations using genetic and chemical tools continue to delineate the roles of PRMTs in regulating immune cells and cancer cells, and cardiovascular and neuronal function, and determine which pathways involving PRMTs could be synergistically targeted in combination therapies for cancer. This research is enhancing our knowledge of the complex functions of arginine methylation, will guide future clinical development and could identify new clinical indications.
Collapse
|
21
|
Ma D, Yang M, Wang Q, Sun C, Shi H, Jing W, Bi Y, Shen X, Ma X, Qin Z, Lin Y, Zhu L, Zhao Y, Cheng Y, Han L. Arginine methyltransferase PRMT5 negatively regulates cGAS-mediated antiviral immune response. SCIENCE ADVANCES 2021; 7:7/13/eabc1834. [PMID: 33762328 PMCID: PMC7990331 DOI: 10.1126/sciadv.abc1834] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 02/04/2021] [Indexed: 05/07/2023]
Abstract
Cyclic GMP-AMP synthase (cGAS) functions as an essential DNA sensor, which senses the cytoplasmic double-stranded DNA and activates the antiviral response. However, the posttranslational modification of cGAS remains to be fully understood and whether it has arginine methylation modification remains unknown. Here, we identified protein arginine methyltransferase 5 (PRMT5) as a direct binding partner of cGAS, and it catalyzed the arginine symmetrical dimethylation of cGAS at the Arg124 residue. Further investigation demonstrated that methylation of cGAS by PRMT5 attenuated cGAS-mediated antiviral immune response by blocking the DNA binding ability of cGAS. Oral administration of PRMT5 inhibitors significantly protected mice from HSV-1 infection and prolonged the survival time of these infected mice. Therefore, our findings revealed an essential regulatory effect of PRMT5 on cGAS-mediated antiviral immune response and provided a promising potential antiviral strategy by modulating PRMT5.
Collapse
Affiliation(s)
- Dapeng Ma
- Shandong Provincial Key Laboratory of Infection and Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Min Yang
- Shandong Provincial Key Laboratory of Infection and Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Qiushi Wang
- Department of Geriatric Gastroenterology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Caiyu Sun
- Shandong Provincial Key Laboratory of Infection and Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Hongbiao Shi
- Key Laboratory for Experimental Teratology, Ministry of Education and Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Weiqiang Jing
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yuxuan Bi
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xuecheng Shen
- Shandong Provincial Key Laboratory of Infection and Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xiaomin Ma
- Shandong Provincial Key Laboratory of Infection and Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Zhenzhi Qin
- Shandong Provincial Key Laboratory of Infection and Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yueke Lin
- Shandong Provincial Key Laboratory of Infection and Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lihui Zhu
- Shandong Provincial Key Laboratory of Infection and Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Yunxue Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yeping Cheng
- Shandong Provincial Key Laboratory of Infection and Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lihui Han
- Shandong Provincial Key Laboratory of Infection and Immunology, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
22
|
Kota SK, Lim ZW, Kota SB. Elavl1 Impacts Osteogenic Differentiation and mRNA Levels of Genes Involved in ECM Organization. Front Cell Dev Biol 2021; 9:606971. [PMID: 33614643 PMCID: PMC7889968 DOI: 10.3389/fcell.2021.606971] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/05/2021] [Indexed: 12/21/2022] Open
Abstract
Posttranscriptional gene regulation by Adenylate Uridylate (AU) rich element RNA binding protein, Elavl1 has been implicated in embryonic development as well as progenitor cell differentiation. Elavl1 binds to hundreds of cellular messenger RNAs predominantly through interactions with AU-rich elements (AREs) found in the untranslated regions (UTRs) and functions by regulating their stability. Biological functions of Elavl1 during osteogenic differentiation of bone marrow derived mesenchymal stem cells is not well-understood. Here we report that specific knockdown of nuclear localized Elavl1 by RNA interference in multipotent BMSCs led to increased osteogenic differentiation. Differential gene expression analysis following unbiased total RNA sequencing upon Elavl1 depletion during osteogenic differentiation of BMSCs showed increased levels of multiple mRNAs that are involved in extracellular matrix organization. We further show that many of these mRNAs contain Elavl1 binding consensus motifs that are preserved in their 3′ UTRs. RNA stability analyses indicated that depletion of Elavl1 prolongs the steady state RNA levels of several of these mRNAs. Together, our data points to Elavl1 mediated negative regulation of multiple genes involved in ECM organization that play a functional role in MSC osteogenic differentiation.
Collapse
Affiliation(s)
- Satya K Kota
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, MA, United States
| | - Zhu Wei Lim
- Division of Bone and Mineral Research, Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Harvard University, Boston, MA, United States
| | - Savithri B Kota
- Division of Nephrology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
23
|
Yuan Y, Nie H. Protein arginine methyltransferase 5: a potential cancer therapeutic target. Cell Oncol (Dordr) 2021; 44:33-44. [PMID: 33469838 DOI: 10.1007/s13402-020-00577-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND PRMT5 is a type II protein arginine methyltransferase that methylates histone or non-histone proteins. Arginine methylation by PRMT5 has been implicated in gene transcription, ribosome biogenesis, RNA transport, pre-mRNA splicing and signal transduction. High expression of PRMT5 has been observed in various cancers and PRMT5 overexpression has been reported to improve cancer cell survival, proliferation, migration and metabolism and to inhibit cancer cell apoptosis. In addition, PRMT5 has been found to be required for cancer stem cell survival, self-renewal and differentiation. Several microRNAs have been shown to regulate PRMT5 expression. As PRMT5 has oncogene-like properties, several PRMT5 inhibitors have been used to explore their efficacy as potential drugs for different types of cancer, and three of them are now being tested in clinical trials. CONCLUSIONS In this review, we summarize current knowledge on the role of PRMT5 in cancer development and progression, including its functions and underlying mechanisms. In addition, we highlight the rapid development of PRMT5 inhibitors and summarize ongoing clinical trials for cancer therapy. By affecting both tumor cells and the tumor microenvironment, PRMT5 inhibitors may serve as effective anti-cancer agents, especially when combined with immune therapies.
Collapse
Affiliation(s)
- Yuanyang Yuan
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, China
| | - Hong Nie
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, China.
| |
Collapse
|
24
|
Deng Z, Ng C, Inoue K, Chen Z, Xia Y, Hu X, Greenblatt M, Pernis A, Zhao B. Def6 regulates endogenous type-I interferon responses in osteoblasts and suppresses osteogenesis. eLife 2020; 9:e59659. [PMID: 33373293 PMCID: PMC7771961 DOI: 10.7554/elife.59659] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 12/15/2020] [Indexed: 12/28/2022] Open
Abstract
Bone remodeling involves a balance between bone resorption and formation. The mechanisms underlying bone remodeling are not well understood. DEF6 is recently identified as a novel loci associated with bone mineral density. However, it is unclear how Def6 impacts bone remodeling. We identify Def6 as a novel osteoblastic regulator that suppresses osteoblastogenesis and bone formation. Def6 deficiency enhances both bone resorption and osteogenesis. The enhanced bone resorption in Def6-/- mice dominates, leading to osteoporosis. Mechanistically, Def6 inhibits the differentiation of both osteoclasts and osteoblasts via a common mechanism through endogenous type-I IFN-mediated feedback inhibition. RNAseq analysis shows expression of a group of IFN stimulated genes (ISGs) during osteoblastogenesis. Furthermore, we found that Def6 is a key upstream regulator of IFNβ and ISG expression in osteoblasts. Collectively, our results identify a novel immunoregulatory function of Def6 in bone remodeling, and shed insights into the interaction between immune system and bone.
Collapse
Affiliation(s)
- Zhonghao Deng
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special SurgeryNew YorkUnited States
| | - Courtney Ng
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special SurgeryNew YorkUnited States
| | - Kazuki Inoue
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special SurgeryNew YorkUnited States
- Department of Medicine, Weill Cornell Medical CollegeNew YorkUnited States
| | - Ziyu Chen
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special SurgeryNew YorkUnited States
| | - Yuhan Xia
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special SurgeryNew YorkUnited States
| | - Xiaoyu Hu
- Institute for Immunology and School of Medicine, Tsinghua UniversityBeijingChina
| | - Matthew Greenblatt
- Pathology and Laboratory Medicine, Weill Cornell Medical CollegeNew YorkUnited States
- Research Division, Hospital for Special SurgeryNew YorkUnited States
| | - Alessandra Pernis
- Autoimmunity and Inflammation Program, Hospital for Special SurgeryNew YorkUnited States
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special SurgeryNew YorkUnited States
- Department of Medicine, Weill Cornell Medical CollegeNew YorkUnited States
- Graduate Program in Cell and Development Biology, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| |
Collapse
|
25
|
The Histone Demethylase KDM3B Promotes Osteo-/Odontogenic Differentiation, Cell Proliferation, and Migration Potential of Stem Cells from the Apical Papilla. Stem Cells Int 2020; 2020:8881021. [PMID: 33082788 PMCID: PMC7563049 DOI: 10.1155/2020/8881021] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/11/2020] [Accepted: 09/20/2020] [Indexed: 02/07/2023] Open
Abstract
Understanding the regulation mechanisms of mesenchymal stem cells (MSCs) can assist in tissue regeneration. The histone demethylase (KDM) family has a crucial role in differentiation and cell proliferation of MSCs, while the function of KDM3B in MSCs is not well understood. In this study, we used the stem cells from the apical papilla (SCAPs) to test whether KDM3B could regulate the function of MSCs. By an alkaline phosphatase (ALP) activity assay, Alizarin red staining, real-time RT-PCR, and western blot analysis, we found that KDM3B enhanced the ALP activity and mineralization of SCAPs and promoted the expression of runt-related transcription factor 2 (RUNX2), osterix (OSX), dentin sialophosphoprotein (DSPP), and osteocalcin (OCN). Additionally, the CFSE, CCK-8, and flow cytometry assays revealed that KDM3B improved cell proliferation by accelerating cell cycle transition from the G1 to S phase. Scratch and transwell migration assays displayed that KDM3B promoted the migration potential of SCAPs. Mechanically, microarray results displayed that 98 genes were upregulated, including STAT1, CCND1, and FGF5, and 48 genes were downregulated after KDM3B overexpression. Besides, we found that the Toll-like receptor and JAK-STAT signaling pathway may be involved in the regulating function of KDM3B in SCAPs. In brief, we discovered that KDM3B promoted the osteo-/odontogenic differentiation, cell proliferation, and migration potential of SCAPs and provided a novel target and theoretical basis for regenerative medicine.
Collapse
|
26
|
Inhibition of PRMT5 Attenuates Oxidative Stress-Induced Pyroptosis via Activation of the Nrf2/HO-1 Signal Pathway in a Mouse Model of Renal Ischemia-Reperfusion Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2345658. [PMID: 31885778 PMCID: PMC6899313 DOI: 10.1155/2019/2345658] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 10/11/2019] [Accepted: 11/01/2019] [Indexed: 02/07/2023]
Abstract
Background Extensive evidence has demonstrated that oxidative stress, pyroptosis, and proinflammatory programmed cell death are related to renal ischemia/reperfusion (I/R) injury. However, the underlying mechanism remains to be illustrated. Protein arginine methylation transferase 5 (PRMT5), which mediates arginine methylation involved in the regulation of epigenetics, exhibits a variety of biological functions and essential roles in diseases. The present study investigated the role of PRMT5 in oxidative stress and pyroptosis induced by I/R injury in a mouse model and in a hypoxia/reoxygenation (H/R) model of HK-2 cells. Methods C57 mice were used as an animal model. All mice underwent right nephrectomy, and the left renal pedicles were either clamped or not. Renal I/R injury was induced by ligating the left renal pedicle for 30 min followed by reperfusion for 24 h. HK-2 cells were exposed to normal conditions or stimulation through H/R. EPZ015666(EPZ)—a selective potent chemical inhibitor—and small interfering RNA (siRNA) were administered to suppress the function and expression of PRMT5. The levels of urea nitrogen and creatinine in the serum and renal tissue injury were assessed. Immunohistochemistry, western blotting, and reverse transcription-polymerase chain reaction were used to evaluate pyroptosis-related proteins including nod-like receptor protein-3, ASC, caspase-1, caspase-11, GSDMD-N, and interleukin-1β. Cell apoptosis and cell viability were detected through flow cytometry, and the levels of reactive oxygen species (ROS) and hydrogen peroxide (H2O2) were measured. Ki-67 was used to assess the proliferation of renal tubular epithelium. In addition, the activity of malondialdehyde and superoxide dismutase was determined. Results I/R or H/R induced an increase in the expression of PRMT5. Inhibition of PRMT5 by EPZ alleviated oxidative stress and I/R- or H/R-induced pyroptosis. In renal tissue, the application of EPZ promoted the proliferation of tubular epithelium. In addition, H/R-induced pyroptosis in HK-2 cells was dependent on oxidative stress in vitro. Administration of either EPZ or siRNA led to decreased expression of pyroptosis-related proteins. Inhibition of PRMT5 also attenuated the I/R- or H/R-induced oxidative stress in vivo and in HK-2 cells, respectively. It also resulted in a distinct decrease in the levels of malondialdehyde and H2O2, and an apparent increase in superoxide dismutase activity in mouse renal tissue. Moreover, it led to a significant decrease in the levels of ROS and H2O2 in HK-2 cells. When activated, NF-E2-related factor/heme oxygenase-1 (Nrf2/HO-1)—a key regulator of various cytoprotective proteins that withstand oxidative damage—can decrease the generation of ROS. Nrf2/HO-1 was downregulated during I/R in tissues and H/R in HK-2 cells, and this effect was reversed by the PRMT5 inhibitor. Furthermore, the expressions of Nrf2 and HO-1 proteins were markedly upregulated by EPZ or siRNA against PRMT5. Conclusion PRMT5 is involved in ischemia- and hypoxia-induced oxidative stress and pyroptosis in vitro and in vivo. Inhibition of PRMT5 may ameliorate renal I/R injury by suppressing oxidative stress and pyroptosis via the activation of the Nrf2/HO-1 pathway, as well as promoting the proliferation of tubular epithelium. Therefore, PRMT5 may be a promising therapeutic target.
Collapse
|
27
|
Ramachandran J, Liu Z, Gray RS, Vokes SA. PRMT5 is necessary to form distinct cartilage identities in the knee and long bone. Dev Biol 2019; 456:154-163. [PMID: 31442442 DOI: 10.1016/j.ydbio.2019.08.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/19/2019] [Accepted: 08/19/2019] [Indexed: 01/08/2023]
Abstract
During skeletal development, limb progenitors become specified as chondrocytes and subsequently differentiate into specialized cartilage compartments. We previously showed that the arginine dimethyl transferase, PRMT5, is essential for regulating the specification of progenitor cells into chondrocytes within early limb buds. Here, we report that PRMT5 regulates the survival of a separate progenitor domain that gives rise to the patella. Independent of its role in knee development, PRMT5 regulates several distinct types of chondrocyte differentiation within the long bones. Chondrocytes lacking PRMT5 have a striking blockage in hypertrophic chondrocyte differentiation and are marked by abnormal gene expression. PRMT5 remains important for articular cartilage and hypertrophic cell identity during adult stages, indicating an ongoing role in homeostasis of these tissues. We conclude that PRMT5 is required for distinct steps of early and late chondrogenic specialization and is thus a critical component of multiple aspects of long bone development and maintenance.
Collapse
Affiliation(s)
- Janani Ramachandran
- Department of Molecular Biosciences, University of Texas at Austin, 100 E 24th Street, Stop A5000, Austin, TX, 78712, USA
| | - Zhaoyang Liu
- Department of Pediatrics, Dell Pediatrics Research Institute, University of Texas at Austin Dell Medical School, 1400 Barbara Jordan Blvd, Austin, TX, 78723, USA
| | - Ryan S Gray
- Department of Nutritional Sciences, University of Texas at Austin, 103 W. 24th Street, A2703, Austin, TX, 78712, USA; Department of Pediatrics, Dell Pediatrics Research Institute, University of Texas at Austin Dell Medical School, 1400 Barbara Jordan Blvd, Austin, TX, 78723, USA
| | - Steven A Vokes
- Department of Molecular Biosciences, University of Texas at Austin, 100 E 24th Street, Stop A5000, Austin, TX, 78712, USA.
| |
Collapse
|
28
|
Asymmetrical methyltransferase PRMT3 regulates human mesenchymal stem cell osteogenesis via miR-3648. Cell Death Dis 2019; 10:581. [PMID: 31378783 PMCID: PMC6680051 DOI: 10.1038/s41419-019-1815-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 07/03/2019] [Accepted: 07/12/2019] [Indexed: 12/15/2022]
Abstract
Histone arginine methylation, which is catalyzed by protein arginine methyltransferases (PRMTs), plays a key regulatory role in various biological processes. Several PRMTs are involved in skeletal development; however, their role in the osteogenic differentiation of mesenchymal stem cells (MSCs) is not completely clear. In this study, we aimed to elucidate the function of PRMT3, a type-I PRMT that catalyzes the formation of ω-mono- or asymmetric dimethyl arginine, in MSCs osteogenesis. We found that PRMT3 promoted MSCs osteogenic commitment and bone remodeling. PRMT3 activated the expression of miR-3648 by enhancing histone H4 arginine 3 asymmetric dimethylation (H4R3me2a) levels at promoter region of the gene. Overexpression of miR-3648 rescued impaired osteogenesis in PRMT3-deficient cells. Moreover, administration of Prmt3 shRNA or a chemical inhibitor of PRMT3 (SGC707) caused an osteopenia phenotype in mice. These results indicate that PRMT3 is a potential therapeutic target for the treatment of bone regeneration and osteopenia disorders.
Collapse
|
29
|
Bone Remodeling: Histone Modifications as Fate Determinants of Bone Cell Differentiation. Int J Mol Sci 2019; 20:ijms20133147. [PMID: 31252653 PMCID: PMC6651527 DOI: 10.3390/ijms20133147] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 02/07/2023] Open
Abstract
The bone tissue is a dynamic complex that constitutes of several interdependent systems and is continuously remodeled through the concerted actions of bone cells. Osteoblasts are mononucleated cells, derived from mesenchymal stem cells, responsible for bone formation. Osteoclasts are large multinucleated cells that differentiate from hematopoietic progenitors of the myeloid lineage and are responsible for bone resorption. The lineage-specific differentiation of bone cells requires an epigenetic regulation of gene expressions involving chromatin dynamics. The key step for understanding gene regulatory networks during bone cell development lies in characterizing the chromatin modifying enzymes responsible for reorganizing and potentiating particular chromatin structure. This review covers the histone-modifying enzymes involved in bone development, discusses the impact of enzymes on gene expression, and provides future directions and clinical significance in this area.
Collapse
|