1
|
Recka N, Simons A, Cornell RA, Van Otterloo E. Epidermal loss of PRMT5 leads to the emergence of an atypical basal keratinocyte-like cell population and defective skin stratification. J Invest Dermatol 2025:S0022-202X(25)00449-X. [PMID: 40339790 DOI: 10.1016/j.jid.2025.04.008] [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: 11/11/2024] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 05/10/2025]
Abstract
During skin development, ectoderm-derived cells undergo precisely coordinated proliferation, differentiation, and adhesion to yield stratified epidermis. Disruptions in these processes can result in congenital anomalies including ectodermal dysplasia and harlequin ichthyosis. Protein Arginine Methyl Transferase 5 (PRMT5)-an enzyme responsible for methylating arginine residues in histones and other proteins-maintains progenitor status in germ and limb bud cells. Similarly, in vitro evidence suggests that PRMT5 prevents differentiation of basal keratinocytes, leading us to hypothesize that PRMT5 preserves the stem-cell phenotype of keratinocytes in vivo. To test this possibility, we generated conditional knockout (cKO) mice lacking Prmt5 in early ectoderm (E7.5), impacting the entire epidermis. Prmt5 cKOs exhibited gross skin defects, compromised skin barrier function, and reduced postnatal viability. Histological analyses revealed significant defects in epidermal stratification, without alterations in apoptosis or proliferation. Single-cell RNA and ATAC-seq analysis identified an atypical population of basal keratinocyte-like cells in Prmt5 cKOs, that exhibited a senescence-like program, characterized by increased Cdkn1a (p21), elevated senescence-associated secretory phenotype (SASP) molecules (Igfbp2), and decreased developmental transcription factor (Trp63) expression. Our findings suggest that PRMT5 prevents basal keratinocyte senescence by repressing Cdkn1a, shedding light on the epigenetic regulation of basal keratinocyte maintenance and senescence in congenital skin disorders.
Collapse
Affiliation(s)
- Nicole Recka
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA; Iowa Institute of Oral Health Research, College of Dentistry & Dental Clinics, University of Iowa, Iowa City, IA, 52242, USA; Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, 52242, USA
| | - Andrean Simons
- Department of Radiation Oncology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Robert A Cornell
- Department of Oral Health Sciences, University of Washington, School of Dentistry, Seattle, WA, 98195, USA
| | - Eric Van Otterloo
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA; Iowa Institute of Oral Health Research, College of Dentistry & Dental Clinics, University of Iowa, Iowa City, IA, 52242, USA; Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA, 52242, USA; Department of Periodontics, College of Dentistry & Dental Clinics, University of Iowa, Iowa City, IA, 52242, USA; Craniofacial Anomalies Research Center, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
2
|
Liang H, Fisher ML, Wu C, Ballon C, Sun X, Mills AA. PRMT5/WDR77 Enhances the Proliferation of Squamous Cell Carcinoma via the ΔNp63α-p21 Axis. Cancers (Basel) 2024; 16:3789. [PMID: 39594744 PMCID: PMC11592282 DOI: 10.3390/cancers16223789] [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: 09/19/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Protein arginine methyltransferase 5 (PRMT5) is a critical oncogenic factor in various cancers, and its inhibition has shown promise in suppressing tumor growth. However, the role of PRMT5 in squamous cell carcinoma (SCC) remains largely unexplored. In this study, we analyzed SCC patient data from The Cancer Genome Atlas (TCGA) and the Cancer Dependency Map (DepMap) to investigate the relationship between PRMT5 and SCC proliferation. We employed competition-based cell proliferation assays, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assays, flow cytometry, and in vivo mouse modeling to examine the regulatory roles of PRMT5 and its binding partner WDR77 (WD repeat domain 77). We identified downstream targets, including the p63 isoform ΔNp63α and the cyclin-dependent kinase inhibitor p21, through single-cell RNA-seq, RT-qPCR, and Western blot analyses. Our findings demonstrate that upregulation of PRMT5 and WDR77 correlates with the poor survival of head and neck squamous cell carcinoma (HNSCC) patients. PRMT5/WDR77 regulates the HNSCC-specific transcriptome and facilitates SCC proliferation by promoting cell cycle progression. The PRMT5 and WDR77 stabilize the ΔNp63α Protein, which in turn, inhibits p21. Moreover, depletion of PRMT5 and WDR77 repress SCC in vivo. This study reveals for the first time that PRMT5 and WDR77 synergize to promote SCC proliferation via the ΔNp63α-p21 axis, highlighting a novel therapeutic target for SCC.
Collapse
Affiliation(s)
- Heng Liang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; (H.L.); (M.L.F.); (C.W.); (C.B.); (X.S.)
- Molecular and Cell Biology Graduate Program, Stony Brook University, Stony Brook, NY 11794, USA
| | - Matthew L. Fisher
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; (H.L.); (M.L.F.); (C.W.); (C.B.); (X.S.)
| | - Caizhi Wu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; (H.L.); (M.L.F.); (C.W.); (C.B.); (X.S.)
| | - Carlos Ballon
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; (H.L.); (M.L.F.); (C.W.); (C.B.); (X.S.)
| | - Xueqin Sun
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; (H.L.); (M.L.F.); (C.W.); (C.B.); (X.S.)
| | - Alea A. Mills
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; (H.L.); (M.L.F.); (C.W.); (C.B.); (X.S.)
| |
Collapse
|
3
|
Zheng Z, Nan B, Liu C, Tang D, Li W, Zhao L, Nie G, He Y. Inhibition of histone methyltransferase PRMT5 attenuates cisplatin-induced hearing loss through the PI3K/Akt-mediated mitochondrial apoptotic pathway. J Pharm Anal 2023; 13:590-602. [PMID: 37440906 PMCID: PMC10334280 DOI: 10.1016/j.jpha.2023.04.014] [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: 12/22/2022] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 07/15/2023] Open
Abstract
This study aimed to evaluate the therapeutic potential of inhibiting protein arginine methyltransferase 5 (PRMT5) in cisplatin-induced hearing loss. The effects of PRMT5 inhibition on cisplatin-induced auditory injury were determined using immunohistochemistry, apoptosis assays, and auditory brainstem response. The mechanism of PRMT5 inhibition on hair cell survival was assessed using RNA-seq and Cleavage Under Targets and Tagment-quantitative polymerase chain reaction (CUT&Tag-qPCR) analyses in the HEI-OC1 cell line. Pharmacological inhibition of PRMT5 significantly alleviated cisplatin-induced damage to hair cells and spiral ganglion neurons in the cochlea and decreased apoptosis by protecting mitochondrial function and preventing the accumulation of reactive oxygen species. CUT&Tag-qPCR analysis demonstrated that inhibition of PRMT5 in HEI-OC1 cells reduced the accumulation of H4R3me2s/H3R8me2s marks at the promoter region of the Pik3ca gene, thus activating the expression of Pik3ca. These findings suggest that PRMT5 inhibitors have strong potential as agents against cisplatin-induced ototoxicity and can lay the foundation for further research on treatment strategies of hearing loss.
Collapse
Affiliation(s)
- Zhiwei Zheng
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, NHC Key Laboratory of Hearing Medicine (Fudan University), Fudan University, Shanghai, 200031, China
| | - Benyu Nan
- Department of Otorhinolaryngology-Head and Neck Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Chang Liu
- Department of Otolaryngology-Head and Neck Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Dongmei Tang
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, NHC Key Laboratory of Hearing Medicine (Fudan University), Fudan University, Shanghai, 200031, China
| | - Wen Li
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, NHC Key Laboratory of Hearing Medicine (Fudan University), Fudan University, Shanghai, 200031, China
| | - Liping Zhao
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, NHC Key Laboratory of Hearing Medicine (Fudan University), Fudan University, Shanghai, 200031, China
| | - Guohui Nie
- Department of Otolaryngology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong, China
| | - Yingzi He
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, NHC Key Laboratory of Hearing Medicine (Fudan University), Fudan University, Shanghai, 200031, China
| |
Collapse
|
4
|
Goruppi S, Clocchiatti A, Bottoni G, Di Cicco E, Ma M, Tassone B, Neel V, Demehri S, Simon C, Paolo Dotto G. The ULK3 kinase is a determinant of keratinocyte self-renewal and tumorigenesis targeting the arginine methylome. Nat Commun 2023; 14:887. [PMID: 36797248 PMCID: PMC9935893 DOI: 10.1038/s41467-023-36410-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/26/2023] [Indexed: 02/18/2023] Open
Abstract
Epigenetic mechanisms oversee epidermal homeostasis and oncogenesis. The identification of kinases controlling these processes has direct therapeutic implications. We show that ULK3 is a nuclear kinase with elevated expression levels in squamous cell carcinomas (SCCs) arising in multiple body sites, including skin and Head/Neck. ULK3 loss by gene silencing or deletion reduces proliferation and clonogenicity of human keratinocytes and SCC-derived cells and affects transcription impinging on stem cell-related and metabolism programs. Mechanistically, ULK3 directly binds and regulates the activity of two histone arginine methyltransferases, PRMT1 and PRMT5 (PRMT1/5), with ULK3 loss compromising PRMT1/5 chromatin association to specific genes and overall methylation of histone H4, a shared target of these enzymes. These findings are of translational significance, as downmodulating ULK3 by RNA interference or locked antisense nucleic acids (LNAs) blunts the proliferation and tumorigenic potential of SCC cells and promotes differentiation in two orthotopic models of skin cancer.
Collapse
Affiliation(s)
- Sandro Goruppi
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, 02129, MA, USA.
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA.
| | - Andrea Clocchiatti
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, 02129, MA, USA
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA
| | - Giulia Bottoni
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, 02129, MA, USA
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA
| | - Emery Di Cicco
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, 02129, MA, USA
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA
| | - Min Ma
- Personalized Cancer Prevention Research Unit and Head and Neck Surgery Division, Centre Hospitalier Universitaire Vaudois, Lausanne, 1011, Switzerland
- Department of Immunobiology, University of Lausanne, Epalinges, 1066, Switzerland
| | - Beatrice Tassone
- Personalized Cancer Prevention Research Unit and Head and Neck Surgery Division, Centre Hospitalier Universitaire Vaudois, Lausanne, 1011, Switzerland
- Department of Immunobiology, University of Lausanne, Epalinges, 1066, Switzerland
| | - Victor Neel
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA
| | - Shadhmer Demehri
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, 02129, MA, USA
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA
| | - Christian Simon
- Personalized Cancer Prevention Research Unit and Head and Neck Surgery Division, Centre Hospitalier Universitaire Vaudois, Lausanne, 1011, Switzerland
- Department of Immunobiology, University of Lausanne, Epalinges, 1066, Switzerland
- International Cancer Prevention Institute, Epalinges, 1066, Switzerland
| | - G Paolo Dotto
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, 02129, MA, USA.
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA.
- Personalized Cancer Prevention Research Unit and Head and Neck Surgery Division, Centre Hospitalier Universitaire Vaudois, Lausanne, 1011, Switzerland.
- Department of Immunobiology, University of Lausanne, Epalinges, 1066, Switzerland.
- International Cancer Prevention Institute, Epalinges, 1066, Switzerland.
| |
Collapse
|
5
|
A cytotoxic effect of human lactoferrin fusion with Fc domain of IgG. Biometals 2022; 36:617-627. [PMID: 36136256 DOI: 10.1007/s10534-022-00443-z] [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: 05/14/2022] [Accepted: 09/06/2022] [Indexed: 11/02/2022]
Abstract
Lactoferrin (LTF) is a natural iron-binding protein with a potential for clinical utility in many human immune disorders, including cancer. A fusion of LTF with the Fc domain of IgG2 (FcLTF) was designed with inherent properties of an extended the half-life in circulation. Furthermore, the effects of LTF and FcLTF were assessed for influence on the activity of natural killer (NK) cells isolated from human peripheral blood, on the NK-92 cell line, and on human monocytes. The NK cytotoxic activity induced by LTF and FcLTF was determined against the human leukemia K562 cell line, and also for monocytes, by measuring TNFα and granzyme B production, and in an assay for Jurkat cell viability. Selected gene expression in NK-92 cells and monocytes, induced by LTF and FcLTF, was performed by Real Time PCR. No significant difference was observed in NK-92 cytotoxicity stimulated by LTF and FcLTF. The effects on NK cells isolated from the human peripheral blood were varied, possibly due to the immunoregulatory nature of LTF sensing the immune status of donors. Furthermore, only the FcLTF group strongly stimulated production of TNFα and granzyme B in isolated monocytes. In addition, only supernatants from the monocyte cultures treated with FcLTF decreased the viability of Jurkat cells. The ability of FcLTF to induce TNFα in monocytes was strongly inhibited by anti-CD32 and moderately inhibited by anti-CD14 antibody. Lastly, it was demonstrated that FcLTF, strongly induced expression of PI3K, with subsequent activation of AKT/mTOR signaling pathway. Overall, it was demonstrated that this novel fusion molecule may be a perferred choice for clinical utility than the wild type LTF.
Collapse
|
6
|
Ye Q, Zhang J, Zhang C, Yi B, Kazama K, Liu W, Sun X, Liu Y, Sun J. Endothelial PRMT5 plays a crucial role in angiogenesis after acute ischemic injury. JCI Insight 2022; 7:e152481. [PMID: 35531958 PMCID: PMC9090242 DOI: 10.1172/jci.insight.152481] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 04/06/2022] [Indexed: 11/17/2022] Open
Abstract
Arginine methylation mediated by protein arginine methyltransferases (PRMTs) has been shown to be an important posttranslational mechanism involved in various biological processes. Herein, we sought to investigate whether PRMT5, a major type II enzyme, is involved in pathological angiogenesis and, if so, to elucidate the molecular mechanism involved. Our results show that PRMT5 expression is significantly upregulated in ischemic tissues and hypoxic endothelial cells (ECs). Endothelial-specific Prmt5-KO mice were generated to define the role of PRMT5 in hindlimb ischemia-induced angiogenesis. We found that these mice exhibited impaired recovery of blood perfusion and motor function of the lower limbs, an impairment that was accompanied by decreased vascular density and increased necrosis as compared with their WT littermates. Furthermore, both pharmacological and genetic inhibition of PRMT5 significantly attenuated EC proliferation, migration, tube formation, and aortic ring sprouting. Mechanistically, we showed that inhibition of PRMT5 markedly attenuated hypoxia-induced factor 1-α (HIF-1α) protein stability and vascular endothelial growth factor-induced (VEGF-induced) signaling pathways in ECs. Our results provide compelling evidence demonstrating a crucial role of PRMT5 in hypoxia-induced angiogenesis and suggest that inhibition of PRMT5 may provide novel therapeutic strategies for the treatment of abnormal angiogenesis-related diseases, such as cancer and diabetic retinopathy.
Collapse
Affiliation(s)
- Qing Ye
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jian Zhang
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Zhang
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Bing Yi
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Kyosuke Kazama
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Wennan Liu
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Xiaobo Sun
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Yan Liu
- Department of Clinical Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianxin Sun
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Luo Y, Gao Y, Liu W, Yang Y, Jiang J, Wang Y, Tang W, Yang S, Sun L, Cai J, Guo X, Takahashi S, Krausz KW, Qu A, Chen L, Xie C, Gonzalez FJ. Myelocytomatosis-Protein Arginine N-Methyltransferase 5 Axis Defines the Tumorigenesis and Immune Response in Hepatocellular Carcinoma. Hepatology 2021; 74:1932-1951. [PMID: 33896016 PMCID: PMC9942701 DOI: 10.1002/hep.31864] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 03/14/2021] [Accepted: 04/09/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS HCC is a leading cause of cancer-related deaths globally with poor outcome and limited therapeutic options. Although the myelocytomatosis (MYC) oncogene is frequently dysregulated in HCC, it is thought to be undruggable. Thus, the current study aimed to identify the critical downstream metabolic network of MYC and develop therapies for MYC-driven HCC. APPROACH AND RESULTS Liver cancer was induced in mice with hepatocyte-specific disruption of Myc and control mice by administration of diethylnitrosamine. Liquid chromatography coupled with mass spectrometry-based metabolomic analyses revealed that urinary dimethylarginine, especially symmetric dimethylarginine (SDMA), was increased in the HCC mouse model in an MYC-dependent manner. Analyses of human samples demonstrated a similar induction of SDMA in the urines from patients with HCC. Mechanistically, Prmt5, encoding protein arginine N-methyltransferase 5, which catalyzes SDMA formation from arginine, was highly induced in HCC and identified as a direct MYC target gene. Moreover, GSK3326595, a PRMT5 inhibitor, suppressed the growth of liver tumors in human MYC-overexpressing transgenic mice that spontaneously develop HCC. Inhibition of PRMT5 exhibited antiproliferative activity through up-regulation of the tumor suppressor gene Cdkn1b/p27, encoding cyclin-dependent kinase inhibitor 1B. In addition, GSK3326595 induced lymphocyte infiltration and major histocompatibility complex class II expression, which might contribute to the enhanced antitumor immune response. Combination of GSK3326595 with anti-programed cell death protein 1 (PD-1) immune checkpoint therapy (ICT) improved therapeutic efficacy in HCC. CONCLUSIONS This study reveals that PRMT5 is an epigenetic executer of MYC, leading to repression of the transcriptional regulation of downstream genes that promote hepatocellular carcinogenesis, highlights a mechanism-based therapeutic strategy for MYC-driven HCC by PRMT5 inhibition through synergistically suppressed proliferation and enhanced antitumor immunity, and finally provides an opportunity to mitigate the resistance of "immune-cold" tumor to ICT.
Collapse
Affiliation(s)
- Yuhong Luo
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yuqing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China,University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Weiwei Liu
- Department of Laboratory Medicine and Central Laboratory, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, P.R. China
| | - Yuan Yang
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, P.R. China
| | - Jie Jiang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Ying Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Wei Tang
- Molecular Epidemiology Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shoumei Yang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lulu Sun
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jie Cai
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xiaozhen Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Shogo Takahashi
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA,Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, USA
| | - Kristopher W. Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China,Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing, P.R. China
| | - Lei Chen
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, P.R. China,International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, P.R. China
| | - Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA,State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China,School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Frank J. Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
8
|
Chakrapani B, Khan MIK, Kadumuri RV, Gupta S, Verma M, Awasthi S, Govindaraju G, Mahesh A, Rajavelu A, Chavali S, Dhayalan A. The uncharacterized protein FAM47E interacts with PRMT5 and regulates its functions. Life Sci Alliance 2021; 4:e202000699. [PMID: 33376131 PMCID: PMC7772775 DOI: 10.26508/lsa.202000699] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 12/16/2022] Open
Abstract
Protein arginine methyltransferase 5 (PRMT5) symmetrically dimethylates arginine residues in various proteins affecting diverse cellular processes such as transcriptional regulation, splicing, DNA repair, differentiation, and cell cycle. Elevated levels of PRMT5 are observed in several types of cancers and are associated with poor clinical outcomes, making PRMT5 an important diagnostic marker and/or therapeutic target for cancers. Here, using yeast two-hybrid screening, followed by immunoprecipitation and pull-down assays, we identify a previously uncharacterized protein, FAM47E, as an interaction partner of PRMT5. We report that FAM47E regulates steady-state levels of PRMT5 by affecting its stability through inhibition of its proteasomal degradation. Importantly, FAM47E enhances the chromatin association and histone methylation activity of PRMT5. The PRMT5-FAM47E interaction affects the regulation of PRMT5 target genes expression and colony-forming capacity of the cells. Taken together, we identify FAM47E as a protein regulator of PRMT5, which promotes the functions of this versatile enzyme. These findings imply that disruption of PRMT5-FAM47E interaction by small molecules might be an alternative strategy to attenuate the oncogenic function(s) of PRMT5.
Collapse
Affiliation(s)
- Baskar Chakrapani
- Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Mohd Imran K Khan
- Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Rajashekar Varma Kadumuri
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | - Somlee Gupta
- Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Mamta Verma
- Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Sharad Awasthi
- Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Gayathri Govindaraju
- Interdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | - Arun Mahesh
- Department of Biotechnology, Pondicherry University, Puducherry, India
| | - Arumugam Rajavelu
- Interdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum, India
| | - Sreenivas Chavali
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, India
| | | |
Collapse
|
9
|
Beketova E, Fang S, Owens JL, Liu S, Chen X, Zhang Q, Asberry AM, Deng X, Malola J, Huang J, Li C, Pili R, Elzey BD, Ratliff TL, Wan J, Hu CD. Protein Arginine Methyltransferase 5 Promotes pICln-Dependent Androgen Receptor Transcription in Castration-Resistant Prostate Cancer. Cancer Res 2020; 80:4904-4917. [PMID: 32999000 PMCID: PMC7669631 DOI: 10.1158/0008-5472.can-20-1228] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/30/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022]
Abstract
The majority of advanced prostate cancer therapies aim to inhibit androgen receptor (AR) signaling. However, AR reactivation inevitably drives disease progression to castration-resistant prostate cancer (CRPC). Here we demonstrate that protein arginine methyltransferase 5 (PRMT5) functions as an epigenetic activator of AR transcription in CRPC, requiring cooperation with a methylosome subunit pICln. In vitro and in xenograft tumors in mice, targeting PRMT5 or pICln suppressed growth of CRPC cells. Full-length AR and AR-V7 transcription activation required both PRMT5 and pICln but not MEP50. This activation of transcription was accompanied by PRMT5-mediated symmetric dimethylation of H4R3 at the proximal AR promoter. Further, knockdown of PRMT5 abolished the binding of pICln (but not vice versa) to the AR proximal promoter region, suggesting that PRMT5 recruits pICln to the AR promoter to activate AR transcription. Differential gene expression analysis in 22Rv1 cells confirmed that PRMT5 and pICln both regulate the androgen signaling pathway. In addition, PRMT5 and pICln protein expression positively correlated with AR and AR-V7 protein expression in CRPC tissues and their expression was highly correlated at the mRNA level across multiple publicly available CRPC datasets. Our results suggest that targeting PRMT5 or pICln may be explored as a novel therapy for CRPC treatment by suppressing expression of AR and AR splice variants to circumvent AR reactivation. SIGNIFICANCE: This study provides evidence that targeting PRMT5 can eliminate expression of AR and can be explored as a novel therapeutic approach to treat metastatic hormone-naïve and castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Elena Beketova
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana.,Purdue University Interdisciplinary Life Sciences Graduate Program, Purdue University, West Lafayette, Indiana
| | - Shuyi Fang
- Department of BioHealth Informatics, Indiana University School of Informatics and Computing, Indiana University - Purdue University Indianapolis, Indianapolis, Indiana
| | - Jake L Owens
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana.,The Indiana University Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana
| | - Xufeng Chen
- Department of Pathology, Duke University School of Medicine, Durham, North Caroline
| | - Qingfu Zhang
- Department of Pathology, Duke University School of Medicine, Durham, North Caroline
| | - Andrew M Asberry
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana.,Purdue University Interdisciplinary Life Sciences Graduate Program, Purdue University, West Lafayette, Indiana
| | - Xuehong Deng
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| | - Jonathan Malola
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| | - Jiaoti Huang
- Department of Pathology, Duke University School of Medicine, Durham, North Caroline
| | - Chenglong Li
- Department of Medicinal Chemistry, University of Florida College of Pharmacy, Gainesville, Florida
| | - Roberto Pili
- Department of Medical Oncology, Indiana University Simon Comprehensive Cancer Center, Indianapolis, Indiana
| | - Bennett D Elzey
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Timothy L Ratliff
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Jun Wan
- Department of BioHealth Informatics, Indiana University School of Informatics and Computing, Indiana University - Purdue University Indianapolis, Indianapolis, Indiana. .,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana.,The Indiana University Simon Comprehensive Cancer Center, Indiana University, Indianapolis, Indiana.,The Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Chang-Deng Hu
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana. .,Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana
| |
Collapse
|
10
|
Liu Z, Ramachandran J, Vokes SA, Gray RS. Regulation of terminal hypertrophic chondrocyte differentiation in Prmt5 mutant mice modeling infantile idiopathic scoliosis. Dis Model Mech 2019; 12:dmm.041251. [PMID: 31848143 PMCID: PMC6955203 DOI: 10.1242/dmm.041251] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/18/2019] [Indexed: 12/20/2022] Open
Abstract
Idiopathic scoliosis (IS) is the most common type of musculoskeletal defect affecting children worldwide, and is classified by age of onset, location and degree of spine curvature. Although rare, IS with onset during infancy is the more severe and rapidly progressive form of the disease, associated with increased mortality due to significant respiratory compromise. The pathophysiology of IS, in particular for infantile IS, remains elusive. Here, we demonstrate the role of PRMT5 in the infantile IS phenotype in mouse. Conditional genetic ablation of PRMT5 in osteochondral progenitors results in impaired terminal hypertrophic chondrocyte differentiation and asymmetric defects of endochondral bone formation in the perinatal spine. Analysis of these several markers of endochondral ossification revealed increased type X collagen (COLX) and Ihh expression, coupled with a dramatic reduction in Mmp13 and RUNX2 expression, in the vertebral growth plate and in regions of the intervertebral disc in the Prmt5 conditional mutant mice. We also demonstrate that PRMT5 has a continuous role in the intervertebral disc and vertebral growth plate in adult mice. Altogether, our results establish PRMT5 as a critical promoter of terminal hypertrophic chondrocyte differentiation and endochondral bone formation during spine development and homeostasis. This article has an associated First Person interview with the first author of the paper. Summary: Loss of Prmt5 in osteochondral progenitors impairs terminal hypertrophic chondrocyte differentiation, leading to defects in endochondral bone formation and models infantile idiopathic scoliosis in mouse.
Collapse
Affiliation(s)
- Zhaoyang Liu
- Department of Pediatrics, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd, The University of Texas at Austin, Dell Medical School, Austin, TX 78723, USA
| | - Janani Ramachandran
- Department of Molecular Biosciences, 2500 Speedway, The University of Texas at Austin, Austin, TX 78712, USA
| | - Steven A Vokes
- Department of Molecular Biosciences, 2500 Speedway, The University of Texas at Austin, Austin, TX 78712, USA
| | - Ryan S Gray
- Department of Pediatrics, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd, The University of Texas at Austin, Dell Medical School, Austin, TX 78723, USA .,Department of Nutritional Sciences, 200 W 24th Street, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
11
|
Ghufran MS, Soni P, Kanade SR. Aflatoxin-induced upregulation of protein arginine methyltransferase 5 is mediated by protein kinase C and extracellular signal-regulated kinase. Cell Biol Toxicol 2018; 35:67-80. [DOI: 10.1007/s10565-018-9439-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/27/2018] [Indexed: 12/24/2022]
|
12
|
Saha K, Fisher ML, Adhikary G, Grun D, Eckert RL. Sulforaphane suppresses PRMT5/MEP50 function in epidermal squamous cell carcinoma leading to reduced tumor formation. Carcinogenesis 2017; 38:827-836. [PMID: 28854561 PMCID: PMC5862259 DOI: 10.1093/carcin/bgx044] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/14/2017] [Accepted: 05/04/2017] [Indexed: 12/19/2022] Open
Abstract
Protein arginine methyltransferase 5 (PRMT5) cooperates with methylosome protein 50 (MEP50) to arginine methylate histone H3 and H4 to silence gene expression, and increased PRMT5 activity is associated with enhanced cancer cell survival. We have studied the role of PRMT5 and MEP50 in epidermal squamous cell carcinoma. We show that knockdown of PRMT5 or MEP50 results in reduced H4R3me2s formation, and reduced cell proliferation, invasion, migration and tumor formation. We further show that treatment with sulforaphane (SFN), a cancer preventive agent derived from cruciferous vegetables, reduces PRMT5 and MEP50 level and H4R3me2s formation, and this is associated with reduced cell proliferation, invasion and migration. The SFN-dependent reduction in PRMT5 and MEP50 level requires proteasome activity. Moreover, SFN-mediated responses are partially reversed by forced PRMT5 or MEP50 expression. SFN treatment of tumors results in reduced MEP50 level and H4R3me2s formation, confirming that that SFN impacts this complex in vivo. These studies suggest that the PRMT5/MEP50 is required for tumor growth and that reduced expression of this complex is a part of the mechanism of SFN suppression of tumor formation.
Collapse
Affiliation(s)
| | | | | | - Daniel Grun
- Department of Biochemistry and Molecular Biology
| | - Richard L Eckert
- Department of Biochemistry and Molecular Biology
- Department of Dermatology
- Department of Obstetrics and Gynecology and
- The Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
13
|
Jeong HJ, Lee HJ, Vuong TA, Choi KS, Choi D, Koo SH, Cho SC, Cho H, Kang JS. Prmt7 Deficiency Causes Reduced Skeletal Muscle Oxidative Metabolism and Age-Related Obesity. Diabetes 2016; 65:1868-82. [PMID: 27207521 DOI: 10.2337/db15-1500] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 04/19/2016] [Indexed: 11/13/2022]
Abstract
Maintenance of skeletal muscle function is critical for metabolic health and the disruption of which exacerbates many chronic diseases such as obesity and diabetes. Skeletal muscle responds to exercise or metabolic demands by a fiber-type switch regulated by signaling-transcription networks that remains to be fully defined. Here, we report that protein arginine methyltransferase 7 (Prmt7) is a key regulator for skeletal muscle oxidative metabolism. Prmt7 is expressed at the highest levels in skeletal muscle and decreased in skeletal muscles with age or obesity. Prmt7(-/-) muscles exhibit decreased oxidative metabolism with decreased expression of genes involved in muscle oxidative metabolism, including PGC-1α. Consistently, Prmt7(-/-) mice exhibited significantly reduced endurance exercise capacities. Furthermore, Prmt7(-/-) mice exhibit decreased energy expenditure, which might contribute to the exacerbated age-related obesity of Prmt7(-/-) mice. Similarly to Prmt7(-/-) muscles, Prmt7 depletion in myoblasts also reduces PGC-1α expression and PGC-1α-promoter driven reporter activities. Prmt7 regulates PGC-1α expression through interaction with and activation of p38 mitogen-activated protein kinase (p38MAPK), which in turn activates ATF2, an upstream transcriptional activator for PGC-1α. Taken together, Prmt7 is a novel regulator for muscle oxidative metabolism via activation of p38MAPK/ATF2/PGC-1α.
Collapse
Affiliation(s)
- Hyeon-Ju Jeong
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon, South Korea
| | - Hye-Jin Lee
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon, South Korea
| | - Tuan Anh Vuong
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon, South Korea
| | - Kyu-Sil Choi
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, South Korea
| | - Dahee Choi
- Division of Life Science, Korea University, Seoul, South Korea
| | - Sung-Hoi Koo
- Division of Life Science, Korea University, Seoul, South Korea
| | - Sung Chun Cho
- Well Aging Research Center, Samsung Advanced Institute of Technology, Samsung Electronics Co. Ltd., Suwon, South Korea
| | - Hana Cho
- Department of Physiology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon, South Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Suwon, South Korea
| |
Collapse
|
14
|
Ghufran MS, Ghosh K, Kanade SR. Aflatoxin B1 induced upregulation of protein arginine methyltransferase 5 in human cell lines. Toxicon 2016; 119:117-21. [PMID: 27242039 DOI: 10.1016/j.toxicon.2016.05.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/17/2016] [Accepted: 05/26/2016] [Indexed: 11/29/2022]
Abstract
The exposure of naturally occurring mycotoxins affects human health and play a vital role in cancer initiation and progression. Aflatoxin B1 is a difuranocoumarin mycotoxin, classified as a group I carcinogen. The present study was conducted to assess the effect of aflatoxin B1 on epigenetic regulatory proteins. The protein arginine methyltransferase 5 expression was induced upon aflatoxin B1 treatment in a dose and time dependent manner. Further global arginine methylation was also increased in the same manner. This is the first report showing the induction of epigenetic regulatory protein, protein arginine methyltransferase 5 upon aflatoxin B1 treatment. Further study is required to establish the detailed pathway of PRMT5 induction.
Collapse
Affiliation(s)
- Md Sajid Ghufran
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Kasargod, Kerala, 671314, India
| | - Krishna Ghosh
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Kasargod, Kerala, 671314, India
| | - Santosh R Kanade
- Department of Biochemistry and Molecular Biology, Central University of Kerala, Kasargod, Kerala, 671314, India.
| |
Collapse
|
15
|
Saha K, Adhikary G, Eckert RL. MEP50/PRMT5 Reduces Gene Expression by Histone Arginine Methylation and this Is Reversed by PKCδ/p38δ Signaling. J Invest Dermatol 2016; 136:214-224. [PMID: 26763441 PMCID: PMC4899982 DOI: 10.1038/jid.2015.400] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 09/03/2015] [Accepted: 09/26/2015] [Indexed: 02/08/2023]
Abstract
PKCδ and p38δ are key proteins in a cascade that stimulates keratinocyte differentiation. This cascade activates transcription of involucrin (hINV) and other genes associated with differentiation. Protein arginine methyltransferase 5 (PRMT5) is an arginine methyltransferase that symmetrically dimethylates arginine residues. This protein interacts with a cofactor, MEP50, and symmetrically dimethylates arginine eight of histone 3 (H3R8me2s) and arginine three of histone 4 (H4R3me2s) to silence gene expression. We use the involucrin gene as a tool to understand the relationship between PKCδ/p38δ and PRMT5/MEP50 signaling. MEP50 suppresses hINV mRNA level and promoter activity. This is associated with increased arginine dimethylation of hINV gene-associated H3/H4. We further show that the PKCδ/p38δ keratinocyte differentiation cascade reduces PRMT5 and MEP50 expression, association with the hINV gene promoter, and H3R8me2s and H4R2me2s formation. We propose that PRMT5/MEP50-dependent methylation is an epigenetic mechanism that assists in silencing of hINV expression, and that PKCδ signaling activates gene expression by directly activating transcription and by suppressing PRMT5/MEP50 dependent arginine dimethylation of promoter associated histones. This is an example of crosstalk between PKCδ/p38δ signaling and PRMT5/MEP50 epigenetic silencing.
Collapse
Affiliation(s)
- Kamalika Saha
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Gautam Adhikary
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Richard L Eckert
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Dermatology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Obstetrics and Gynecology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
16
|
Prmt5 is a regulator of muscle stem cell expansion in adult mice. Nat Commun 2015; 6:7140. [PMID: 26028225 PMCID: PMC4458870 DOI: 10.1038/ncomms8140] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 04/09/2015] [Indexed: 01/16/2023] Open
Abstract
Skeletal muscle stem cells (MuSC), also called satellite cells, are indispensable for maintenance and regeneration of adult skeletal muscles. Yet, a comprehensive picture of the regulatory events controlling the fate of MuSC is missing. Here, we determine the proteome of MuSC to design a loss-of-function screen, and identify 120 genes important for MuSC function including the arginine methyltransferase Prmt5. MuSC-specific inactivation of Prmt5 in adult mice prevents expansion of MuSC, abolishes long-term MuSC maintenance and abrogates skeletal muscle regeneration. Interestingly, Prmt5 is dispensable for proliferation and differentiation of Pax7+ myogenic progenitor cells during mouse embryonic development, indicating significant differences between embryonic and adult myogenesis. Mechanistic studies reveal that Prmt5 controls proliferation of adult MuSC by direct epigenetic silencing of the cell cycle inhibitor p21. We reason that Prmt5 generates a poised state that keeps MuSC in a standby mode, thus allowing rapid MuSC amplification under disease conditions. Skeletal muscle satellite cells are important for muscle regeneration, but their regulatory mechanisms are largely unknown. Here the authors identify arginine methyltransferase Prmt5 as a key regulator of satellite cell maintenance and function in adult mice, and show that Prmt5 acts mainly but not exclusively on the cell cycle inhibitor p21.
Collapse
|
17
|
Stopa N, Krebs JE, Shechter D. The PRMT5 arginine methyltransferase: many roles in development, cancer and beyond. Cell Mol Life Sci 2015; 72:2041-59. [PMID: 25662273 PMCID: PMC4430368 DOI: 10.1007/s00018-015-1847-9] [Citation(s) in RCA: 370] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 01/10/2015] [Accepted: 01/29/2015] [Indexed: 10/24/2022]
Abstract
Post-translational arginine methylation is responsible for regulation of many biological processes. The protein arginine methyltransferase 5 (PRMT5, also known as Hsl7, Jbp1, Skb1, Capsuleen, or Dart5) is the major enzyme responsible for mono- and symmetric dimethylation of arginine. An expanding literature demonstrates its critical biological function in a wide range of cellular processes. Histone and other protein methylation by PRMT5 regulate genome organization, transcription, stem cells, primordial germ cells, differentiation, the cell cycle, and spliceosome assembly. Metazoan PRMT5 is found in complex with the WD-repeat protein MEP50 (also known as Wdr77, androgen receptor coactivator p44, or Valois). PRMT5 also directly associates with a range of other protein factors, including pICln, Menin, CoPR5 and RioK1 that may alter its subcellular localization and protein substrate selection. Protein substrate and PRMT5-MEP50 post-translation modifications induce crosstalk to regulate PRMT5 activity. Crystal structures of C. elegans PRMT5 and human and frog PRMT5-MEP50 complexes provide substantial insight into the mechanisms of substrate recognition and procession to dimethylation. Enzymological studies of PRMT5 have uncovered compelling insights essential for future development of specific PRMT5 inhibitors. In addition, newly accumulating evidence implicates PRMT5 and MEP50 expression levels and their methyltransferase activity in cancer tumorigenesis, and, significantly, as markers of poor clinical outcome, marking them as potential oncogenes. Here, we review the substantial new literature on PRMT5 and its partners to highlight the significance of understanding this essential enzyme in health and disease.
Collapse
Affiliation(s)
- Nicole Stopa
- Department of Biological Sciences, University of Alaska Anchorage, 3211 Providence Drive, Anchorage, AK 99508, USA
| | - Jocelyn E. Krebs
- Department of Biological Sciences, University of Alaska Anchorage, 3211 Providence Drive, Anchorage, AK 99508, USA
| | - David Shechter
- Department of Biochemistry, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| |
Collapse
|
18
|
Saha K, Eckert RL. Methylosome Protein 50 and PKCδ/p38δ Protein Signaling Control Keratinocyte Proliferation via Opposing Effects on p21Cip1 Gene Expression. J Biol Chem 2015; 290:13521-30. [PMID: 25851901 PMCID: PMC4505598 DOI: 10.1074/jbc.m115.642868] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/19/2015] [Indexed: 11/06/2022] Open
Abstract
Protein arginine methyltransferase 5 (PRMT5) is a key epigenetic regulator that symmetrically dimethylates arginine residues on histones H3 and H4 to silence gene expression. PRMT5 is frequently observed in a complex with the cofactor methylosome protein 50 (MEP50), which is required for PRMT5 activity. PKCδ/p38δ signaling, a key controller of keratinocyte proliferation and differentiation, increases p21(Cip1) expression to suppress keratinocyte proliferation. We now show that MEP50 enhances keratinocyte proliferation and survival via mechanisms that include silencing of p21(Cip1) expression. This is associated with enhanced PRMT5-MEP50 interaction at the p21(Cip1) promoter and enhanced arginine dimethylation of the promoter-associated histones H3 and H4. It is also associated with a MEP50-dependent reduction in the level of p53, a key controller of p21(Cip1) gene expression. We confirm an important biological role for MEP50 and PRMT5 in regulating keratinocyte proliferation using a stratified epidermal equivalent model that mimics in vivo epidermal keratinocyte differentiation. In this model, PRMT5 or MEP50 knockdown results in reduced keratinocyte proliferation. We further show that PKCδ/p38δ signaling suppresses MEP50 expression, leading to reduced H3/H4 arginine dimethylation at the p21(Cip1) promoter, and that this is associated with enhanced p21(Cip1) expression and reduced cell proliferation. These findings describe an opposing action between PKCδ/p38δ MAPK signaling and PRMT5/MEP50 epigenetic silencing mechanisms in regulating cell proliferation.
Collapse
Affiliation(s)
- Kamalika Saha
- From the Departments of Biochemistry and Molecular Biology
| | - Richard L Eckert
- From the Departments of Biochemistry and Molecular Biology, Dermatology, and Obstetrics and Gynecology and the Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
19
|
Zhang HT, Zhang D, Zha ZG, Hu CD. Transcriptional activation of PRMT5 by NF-Y is required for cell growth and negatively regulated by the PKC/c-Fos signaling in prostate cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1839:1330-40. [PMID: 25281873 DOI: 10.1016/j.bbagrm.2014.09.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 09/17/2014] [Accepted: 09/24/2014] [Indexed: 01/11/2023]
Abstract
Protein arginine methyltransferase 5 (PRMT5) symmetrically methylates arginine residues of histones and non-histone protein substrates and regulates a variety of cellular processes through epigenetic control of target gene expression or post-translational modification of signaling molecules. Recent evidence suggests that PRMT5 may function as an oncogene and its overexpression contributes to the development and progression of several human cancers. However, the mechanism underlying the regulation of PRMT5 expression in cancer cells remains largely unknown. In the present study, we have mapped the proximal promoter of PRMT5 to the -240bp region and identified nuclear transcription factor Y (NF-Y) as a critical transcription factor that binds to the two inverted CCAAT boxes and regulates PRMT5 expression in multiple cancer cell lines. Further, we present evidence that loss of PRMT5 is responsible for cell growth inhibition induced by knockdown of NF-YA, a subunit of NF-Y that forms a heterotrimeric complex with NF-YB and NF-YC for function. Significantly, we have found that activation of protein kinase C (PKC) by phorbol 12-myristate 13-acetate (PMA) in LNCaP prostate cancer cells down-regulates the expression of NF-YA and PRMT5 at the transcription level in a c-Fos-dependent manner. Given that down-regulation of several PKC isozymes is implicated in the development and progression of several human cancers, our findings suggest that the PKC-c-Fos-NF-Y signaling pathway may be responsible for PRMT5 overexpression in a subset of human cancer patients.
Collapse
Affiliation(s)
- Huan-Tian Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology and the Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA; Institute of Orthopedic Diseases and Department of Orthopedics of the First Affiliated Hospital, Jinan University, Guangzhou 510630, People's Republic of China
| | - Dabao Zhang
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Zhen-Gang Zha
- Department of Medicinal Chemistry and Molecular Pharmacology and the Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Chang-Deng Hu
- Department of Medicinal Chemistry and Molecular Pharmacology and the Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
20
|
Baek JH, Rubinstein M, Scheuer T, Trimmer JS. Reciprocal changes in phosphorylation and methylation of mammalian brain sodium channels in response to seizures. J Biol Chem 2014; 289:15363-73. [PMID: 24737319 PMCID: PMC4140893 DOI: 10.1074/jbc.m114.562785] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 04/10/2014] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium (Nav) channels initiate action potentials in brain neurons and are primary therapeutic targets for anti-epileptic drugs controlling neuronal hyperexcitability in epilepsy. The molecular mechanisms underlying abnormal Nav channel expression, localization, and function during development of epilepsy are poorly understood but can potentially result from altered posttranslational modifications (PTMs). For example, phosphorylation regulates Nav channel gating, and has been proposed to contribute to acquired insensitivity to anti-epileptic drugs exhibited by Nav channels in epileptic neurons. However, whether changes in specific brain Nav channel PTMs occur acutely in response to seizures has not been established. Here, we show changes in PTMs of the major brain Nav channel, Nav1.2, after acute kainate-induced seizures. Mass spectrometry-based proteomic analyses of Nav1.2 purified from the brains of control and seizure animals revealed a significant down-regulation of phosphorylation at nine sites, primarily located in the interdomain I-II linker, the region of Nav1.2 crucial for phosphorylation-dependent regulation of activity. Interestingly, Nav1.2 in the seizure samples contained methylated arginine (MeArg) at three sites. These MeArgs were adjacent to down-regulated sites of phosphorylation, and Nav1.2 methylation increased after seizure. Phosphorylation and MeArg were not found together on the same tryptic peptide, suggesting reciprocal regulation of these two PTMs. Coexpression of Nav1.2 with the primary brain arginine methyltransferase PRMT8 led to a surprising 3-fold increase in Nav1.2 current. Reciprocal regulation of phosphorylation and MeArg of Nav1.2 may underlie changes in neuronal Nav channel function in response to seizures and also contribute to physiological modulation of neuronal excitability.
Collapse
Affiliation(s)
- Je-Hyun Baek
- From the Department of Neurobiology, Physiology, and Behavior and
| | - Moran Rubinstein
- the Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington 98195-7280
| | - Todd Scheuer
- the Department of Pharmacology, University of Washington School of Medicine, Seattle, Washington 98195-7280
| | - James S Trimmer
- From the Department of Neurobiology, Physiology, and Behavior and the Department of Physiology and Membrane Biology, University of California, Davis, California 95616 and
| |
Collapse
|
21
|
Nicholas C, Yang J, Peters SB, Bill MA, Baiocchi RA, Yan F, Sïf S, Tae S, Gaudio E, Wu X, Grever MR, Young GS, Lesinski GB. PRMT5 is upregulated in malignant and metastatic melanoma and regulates expression of MITF and p27(Kip1.). PLoS One 2013; 8:e74710. [PMID: 24098663 PMCID: PMC3786975 DOI: 10.1371/journal.pone.0074710] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 08/05/2013] [Indexed: 12/16/2022] Open
Abstract
Protein arginine methyltransferase-5 (PRMT5) is a Type II arginine methyltransferase that regulates various cellular functions. We hypothesized that PRMT5 plays a role in regulating the growth of human melanoma cells. Immunohistochemical analysis indicated significant upregulation of PRMT5 in human melanocytic nevi, malignant melanomas and metastatic melanomas as compared to normal epidermis. Furthermore, nuclear PRMT5 was significantly decreased in metastatic melanomas as compared to primary cutaneous melanomas. In human metastatic melanoma cell lines, PRMT5 was predominantly cytoplasmic, and associated with its enzymatic cofactor Mep50, but not STAT3 or cyclin D1. However, histologic examination of tumor xenografts from athymic mice revealed heterogeneous nuclear and cytoplasmic PRMT5 expression. Depletion of PRMT5 via siRNA inhibited proliferation in a subset of melanoma cell lines, while it accelerated growth of others. Loss of PRMT5 also led to reduced expression of MITF (microphthalmia-associated transcription factor), a melanocyte-lineage specific oncogene, and increased expression of the cell cycle regulator p27Kip1. These results are the first to report elevated PRMT5 expression in human melanoma specimens and indicate this protein may regulate MITF and p27Kip1 expression in human melanoma cells.
Collapse
Affiliation(s)
- Courtney Nicholas
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Jennifer Yang
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Sara B. Peters
- Department of Pathology, Division of Dermatopathology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Matthew A. Bill
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Robert A. Baiocchi
- Department of Internal Medicine, Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Fengting Yan
- Department of Internal Medicine, Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Saïd Sïf
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Sookil Tae
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, Ohio, United States of America
| | - Eugenio Gaudio
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, Ohio, United States of America
| | - Xin Wu
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Michael R. Grever
- Department of Internal Medicine, Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States of America
| | - Gregory S. Young
- Center for Biostatistics, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Gregory B. Lesinski
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
22
|
AP1 transcription factors in epidermal differentiation and skin cancer. J Skin Cancer 2013; 2013:537028. [PMID: 23762562 PMCID: PMC3676924 DOI: 10.1155/2013/537028] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 05/02/2013] [Indexed: 01/17/2023] Open
Abstract
AP1 (jun/fos) transcription factors (c-jun, junB, junD, c-fos, FosB, Fra-1, and Fra-2) are key regulators of epidermal keratinocyte survival and differentiation and important drivers of cancer development. Understanding the role of these factors in epidermis is complicated by the fact that each protein is expressed, at different levels, in multiple cells layers in differentiating epidermis, and because AP1 transcription factors regulate competing processes (i.e., proliferation, apoptosis, and differentiation). Various in vivo genetic approaches have been used to study these proteins including targeted and conditional knockdown, overexpression, and expression of dominant-negative inactivating AP1 transcription factors in epidermis. Taken together, these studies suggest that individual AP1 transcription factors have different functions in the epidermis and in cancer development and that altering AP1 transcription factor function in the basal versus suprabasal layers differentially influences the epidermal differentiation response and disease and cancer development.
Collapse
|
23
|
Ho MC, Wilczek C, Bonanno JB, Xing L, Seznec J, Matsui T, Carter LG, Onikubo T, Kumar PR, Chan MK, Brenowitz M, Cheng RH, Reimer U, Almo SC, Shechter D. Structure of the arginine methyltransferase PRMT5-MEP50 reveals a mechanism for substrate specificity. PLoS One 2013; 8:e57008. [PMID: 23451136 PMCID: PMC3581573 DOI: 10.1371/journal.pone.0057008] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 01/16/2013] [Indexed: 01/07/2023] Open
Abstract
The arginine methyltransferase PRMT5-MEP50 is required for embryogenesis and is misregulated in many cancers. PRMT5 targets a wide variety of substrates, including histone proteins involved in specifying an epigenetic code. However, the mechanism by which PRMT5 utilizes MEP50 to discriminate substrates and to specifically methylate target arginines is unclear. To test a model in which MEP50 is critical for substrate recognition and orientation, we determined the crystal structure of Xenopus laevis PRMT5-MEP50 complexed with S-adenosylhomocysteine (SAH). PRMT5-MEP50 forms an unusual tetramer of heterodimers with substantial surface negative charge. MEP50 is required for PRMT5-catalyzed histone H2A and H4 methyltransferase activity and binds substrates independently. The PRMT5 catalytic site is oriented towards the cross-dimer paired MEP50. Histone peptide arrays and solution assays demonstrate that PRMT5-MEP50 activity is inhibited by substrate phosphorylation and enhanced by substrate acetylation. Electron microscopy and reconstruction showed substrate centered on MEP50. These data support a mechanism in which MEP50 binds substrate and stimulates PRMT5 activity modulated by substrate post-translational modifications.
Collapse
Affiliation(s)
- Meng-Chiao Ho
- Department of Biochemistry, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei, Taiwan
| | - Carola Wilczek
- Department of Biochemistry, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
| | - Jeffrey B. Bonanno
- Department of Biochemistry, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
| | - Li Xing
- Department of Molecular and Cellular Biology, University of California Davis, Davis, California, United States of America
| | | | - Tsutomu Matsui
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, California, United States of America
| | - Lester G. Carter
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Menlo Park, California, United States of America
| | - Takashi Onikubo
- Department of Biochemistry, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
| | - P. Rajesh Kumar
- Department of Biochemistry, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
| | - Man K. Chan
- Department of Biochemistry, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
| | - Michael Brenowitz
- Department of Biochemistry, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
| | - R. Holland Cheng
- Department of Molecular and Cellular Biology, University of California Davis, Davis, California, United States of America
| | - Ulf Reimer
- JPT Peptide Technologies, Berlin, Germany
| | - Steven C. Almo
- Department of Biochemistry, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
- Department of Physiology and Biophysics, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
| | - David Shechter
- Department of Biochemistry, Albert Einstein College of Medicine of Yeshiva University, Bronx, New York, United States of America
| |
Collapse
|
24
|
|