1
|
Zou D, Feng S, Hu B, Guo M, Lv Y, Ma R, Du Y, Feng J. Bromodomain proteins as potential therapeutic targets for B-cell non-Hodgkin lymphoma. Cell Biosci 2024; 14:143. [PMID: 39580422 PMCID: PMC11585172 DOI: 10.1186/s13578-024-01326-1] [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: 06/26/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND B-cell non-Hodgkin lymphoma (B-NHL) is the most common type of lymphoma and is significantly heterogeneous among various subtypes. Despite of considerable advancements in treatment strategies for B-NHL, the prognosis of relapsed/refractory patients remains poor. MAIN TEXT It has been indicated that epigenetic dysregulation is critically associated with the pathogenesis of most hematological malignancies, resulting in the clinical targeting of epigenetic modifications. Bromodomain (BRD) proteins are essential epigenetic regulators which contain eight subfamilies, including BRD and extra-terminal domain (BET) family, histone acetyltransferases (HATs) and HAT-related proteins, transcriptional coactivators, transcriptional mediators, methyltransferases, helicases, ATP-dependent chromatin-remodeling complexes, and nuclear-scaffolding proteins. Most pre-clinical and clinical studies on B-NHL have focused predominantly on the BET family and the use of BET inhibitors as mono-treatment or co-treatment with other anti-tumor drugs. Furthermore, preclinical models of B-NHL have revealed that BET degraders are more active than BET inhibitors. Moreover, with the development of BET inhibitors and degraders, non-BET BRD protein inhibitors have also been designed and have shown antitumor activities in B-NHL preclinical models. This review summarized the mechanism of BRD proteins and the recent progress of BRD protein-related drugs in B-NHL. This study aimed to collect the most recent evidences and summarize possibility on whether BRD proteins can serve as therapeutic targets for B-NHL. CONCLUSION In summary, BRD proteins are critical epigenetic regulatory factors and may be potential therapeutic targets for B-NHL.
Collapse
Affiliation(s)
- Dan Zou
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Sitong Feng
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Bowen Hu
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Mengya Guo
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yan Lv
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Rong Ma
- Research Center for Clinical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yuxin Du
- Research Center for Clinical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.
| | - Jifeng Feng
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China.
| |
Collapse
|
2
|
Dai R, Xiang Y, Fang R, Zheng HH, Zhao QS, Wang Y. Lonicerin alleviates ovalbumin-induced asthma of mice via inhibiting enhancer of zeste homolog 2/nuclear factor-kappa B signaling pathway. Exp Anim 2024; 73:154-161. [PMID: 37952975 PMCID: PMC11091354 DOI: 10.1538/expanim.23-0068] [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: 05/26/2023] [Accepted: 10/31/2023] [Indexed: 11/14/2023] Open
Abstract
Asthma is the most common chronic disease in the respiratory system of children caused by abnormal immunity that responses to common antigens. Lonicerin exerts anti-inflammatory activity in other inflammatory models through targeting enhancer of zeste homolog 2 (EZH2) that is related to asthma. We sought to explore the role and mechanism of lonicerin in regulating allergic airway inflammation. Mice were intraperitoneally injected 10 µg ovalbumin (OVA) on postnatal day 5 (P5) and P10, and then inhaled 3% aerosolized OVA for 10 min every day on P18-20, to establish asthmatic mice model. Lonicerin (10 or 30 mg/kg) was given to mice by intragastric administration on P16-P20. Notably, the administration of lonicerin amended infiltration of inflammatory cells and mucus hypersecretion. OVA-specific IgE level, inflammatory cell count and inflammatory cytokines in asthmatic mice were reduced after lonicerin treatment. Moreover, it suppressed the activity of EZH2 and activation of nuclear factor-kappa B (NF-κB) as evidenced by decreasing tri-methylation of histone H3 at lysine 27 and reducing nuclear translocation of NF-κB p65. In a word, Lonicerin may attenuate asthma by inhibiting EZH2/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Rui Dai
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678, Furong Road, Hefei 230601, Anhui, P.R. China
| | - Yun Xiang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678, Furong Road, Hefei 230601, Anhui, P.R. China
| | - Rui Fang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678, Furong Road, Hefei 230601, Anhui, P.R. China
| | - Hai-Han Zheng
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678, Furong Road, Hefei 230601, Anhui, P.R. China
| | - Qing-Song Zhao
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678, Furong Road, Hefei 230601, Anhui, P.R. China
| | - Yan Wang
- Department of Pediatrics, The Second Affiliated Hospital of Anhui Medical University, No. 678, Furong Road, Hefei 230601, Anhui, P.R. China
| |
Collapse
|
3
|
Guefack MGF, Bhatnagar S. Advances in Epigenetic Therapeutics for Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1465:89-97. [PMID: 39586995 DOI: 10.1007/978-3-031-66686-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
The epigenetic deregulations correlate with tumorigenesis, resistance to therapy, and metastasis of breast cancer cells. Given the predominance of aberrant epigenomic mechanisms, there is a growing emphasis on targeting epigenetic mechanisms for breast cancer therapeutic development. Selective inhibitors of epigenetic enzymes and the combined approach of epigenetic therapies with chemotherapies or hormone therapies in the treatment of breast cancer represent promising therapeutic strategies. In this chapter, we review the targeting of epigenetic mechanisms and highlight current epigenetic research in the development of breast cancer therapy.
Collapse
Affiliation(s)
- Michel-Gael F Guefack
- Department of Medical Microbiology and Immunology, University of California Davis School of Medicine, Davis, CA, USA
| | - Sanchita Bhatnagar
- Department of Medical Microbiology and Immunology, University of California Davis School of Medicine, Davis, CA, USA.
| |
Collapse
|
4
|
Peng Y, Bui CH, Zhang XJ, Chen JS, Tham CC, Chu WK, Chen LJ, Pang CP, Yam JC. The role of EZH2 in ocular diseases: a narrative review. Epigenomics 2023; 15:557-570. [PMID: 37458071 DOI: 10.2217/epi-2023-0147] [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] [Indexed: 07/18/2023] Open
Abstract
EZH2, acting as a catalytic subunit of PRC2 to catalyze lysine 27 in histone H3, induces the suppression of gene expression. EZH2 can regulate cell proliferation and differentiation of retinal progenitors, which are required for physiological retinal development. Meanwhile, an abnormal level of EZH2 has been observed in ocular tumors and other pathological tissues. This review summarizes the current knowledge on EZH2 in retinal development and ocular diseases, including inherited retinal diseases, ocular tumors, corneal injury, cataract, glaucoma, diabetic retinopathy and age-related retinal degeneration. We highlight the potential of targeting EZH2 as a precision therapeutic target in ocular diseases.
Collapse
Affiliation(s)
- Yu Peng
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, 999077, Hong Kong
| | - Christine Ht Bui
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, 999077, Hong Kong
| | - Xiu J Zhang
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, 999077, Hong Kong
| | - Jian S Chen
- Aier School of Ophthalmology, Central South University, Changsha, Hunan Province, 410000, China
- Aier Eye Institute, Changsha, Hunan Province, 410000, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, Guangdong Province, 510000, China
| | - Clement C Tham
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, 999077, Hong Kong
- Hong Kong Eye Hospital, Kowloon, 999077, Hong Kong
- Department of Ophthalmology & Visual Sciences, Prince of Wales Hospital, 999077, Hong Kong
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, 999077, Hong Kong
| | - Wai K Chu
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, 999077, Hong Kong
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, 999077, Hong Kong
| | - Li J Chen
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, 999077, Hong Kong
- Department of Ophthalmology & Visual Sciences, Prince of Wales Hospital, 999077, Hong Kong
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, 999077, Hong Kong
| | - Chi P Pang
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, 999077, Hong Kong
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, 999077, Hong Kong
| | - Jason C Yam
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, 999077, Hong Kong
- Hong Kong Eye Hospital, Kowloon, 999077, Hong Kong
- Department of Ophthalmology, Hong Kong Children's Hospital, 999077, Hong Kong
- Department of Ophthalmology & Visual Sciences, Prince of Wales Hospital, 999077, Hong Kong
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, 999077, Hong Kong
| |
Collapse
|
5
|
Roh J, Im M, Kang J, Youn B, Kim W. Long non-coding RNA in glioma: novel genetic players in temozolomide resistance. Anim Cells Syst (Seoul) 2023; 27:19-28. [PMID: 36819921 PMCID: PMC9937017 DOI: 10.1080/19768354.2023.2175497] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
Glioma is the most common primary malignant brain tumor in adults and accounts for approximately 80% of brain and central nervous system tumors. In 2021, the World Health Organization (WHO) published a new taxonomy for glioma based on its histological features and molecular alterations. Isocitrate dehydrogenase (IDH) catalyzes the decarboxylation of isocitrate, a critical metabolic reaction in energy generation in cells. Mutations in the IDH genes interrupt cell differentiation and serve as molecular biomarkers that can be used to classify gliomas. For example, the mutant IDH is widely detected in low-grade gliomas, whereas the wild type is in high-grade ones, including glioblastomas. Long non-coding RNAs (lncRNAs) are epigenetically involved in gene expression and contribute to glioma development. To investigate the potential use of lncRNAs as biomarkers, we examined lncRNA dysregulation dependent on the IDH mutation status. We found that several lncRNAs, namely, AL606760.2, H19, MALAT1, PVT1 and SBF2-AS1 may function as glioma risk factors, whereas AC068643.1, AC079228.1, DGCR5, FAM13A-AS1, HAR1A and WDFY3-AS2 may have protective effects. Notably, H19, MALAT1, PVT1, and SBF2-AS1 have been associated with temozolomide resistance in glioma patients. This review study suggests that targeting glioma-associated lncRNAs might aid the treatment of glioma.
Collapse
Affiliation(s)
- Jungwook Roh
- Department of Science Education, Korea National University of Education, Cheongju-si, Republic of Korea
| | - Mijung Im
- Department of Science Education, Korea National University of Education, Cheongju-si, Republic of Korea
| | - JiHoon Kang
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory, Emory University School of Medicine, Atlanta, GA, USA
| | - BuHyun Youn
- Department of Biological Sciences, Pusan National University, Busan, Republic of Korea, BuHyun Youn Department of Biological Sciences, Pusan National University, Busandaehak-ro 63beon-gil 2, Geumjeong-gu, Busan46241, Republic of Korea; Wanyeon Kim Department of Biology Education, Korea National University of Education, 250 Taeseongtabyeon-ro, Gangnae-myeon, Heungdeok-gu, Cheongju-si, Chungbuk28173, Republic of Korea
| | - Wanyeon Kim
- Department of Science Education, Korea National University of Education, Cheongju-si, Republic of Korea,Department of Biology Education, Korea National University of Education, Cheongju-si, Republic of Korea, BuHyun Youn Department of Biological Sciences, Pusan National University, Busandaehak-ro 63beon-gil 2, Geumjeong-gu, Busan46241, Republic of Korea; Wanyeon Kim Department of Biology Education, Korea National University of Education, 250 Taeseongtabyeon-ro, Gangnae-myeon, Heungdeok-gu, Cheongju-si, Chungbuk28173, Republic of Korea
| |
Collapse
|
6
|
Huang S, Zhen Y, Yin X, Yang Z, Li X, Wang R, Wen H, Zhong H, Yan J, Sun Q. KMT2C Induced by FABP5P3 Aggravates Keratinocyte Hyperproliferation and Psoriasiform Skin Inflammation by Upregulating the Transcription of PIK3R3. J Invest Dermatol 2023; 143:37-47.e8. [PMID: 35870559 DOI: 10.1016/j.jid.2022.06.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/01/2022] [Accepted: 06/13/2022] [Indexed: 12/24/2022]
Abstract
The extensive involvement of lysine methyltransferase 2C (KMT2C) in the inflammatory response is well-documented. However, little is known about the role of KMT2C in psoriasis. We identified that KMT2C was significantly upregulated in the epidermis of psoriatic skin lesions and the psoriasiform cell model. KMT2C knockdown diminished keratinocyte proliferation and the secretion of IL-6, IL-8, CCL20, and S100A9 in vitro and in vivo. In psoriasiform keratinocytes, KMT2C promoted the transcription of PIK3R3 by regulating the enrichment of histone H3 lysine 4 trimethylation at the PIK3R3 promoter and histone 3 lysine 4 monomethylation at the enhancer. The PIK3R3/protein kinase B/NF-κB pathway is a vital step in KMT2C-mediated alleviation of cytokine-primed inflammation. The long noncoding RNA FABP5P3 sustained KMT2C mRNA stability by recruiting human antigen R. Furthermore, inhibition of KMT2C attenuated epidermal hyperplasia and skin inflammation in mice with psoriasis. Taken together, our findings indicated a link between KMT2C and psoriasis and opened the possibility of using KMT2C as a potential therapeutic target for psoriasis treatment.
Collapse
Affiliation(s)
- Shan Huang
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China; Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Yunyue Zhen
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China; Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Xiran Yin
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China; Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Zhenxian Yang
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China; Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Xueqing Li
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China; Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Ruijie Wang
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China; Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - He Wen
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China
| | - Hua Zhong
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China
| | - Jianjun Yan
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China; Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Qing Sun
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
7
|
Barros II, Leão V, Santis JO, Rosa RCA, Brotto DB, Storti CB, Siena ÁDD, Molfetta GA, Silva WA. Non-Syndromic Intellectual Disability and Its Pathways: A Long Noncoding RNA Perspective. Noncoding RNA 2021; 7:ncrna7010022. [PMID: 33799572 PMCID: PMC8005948 DOI: 10.3390/ncrna7010022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Non-syndromic intellectual disability (NS-ID or idiopathic) is a complex neurodevelopmental disorder that represents a global health issue. Although many efforts have been made to characterize it and distinguish it from syndromic intellectual disability (S-ID), the highly heterogeneous aspect of this disorder makes it difficult to understand its etiology. Long noncoding RNAs (lncRNAs) comprise a large group of transcripts that can act through various mechanisms and be involved in important neurodevelopmental processes. In this sense, comprehending the roles they play in this intricate context is a valuable way of getting new insights about how NS-ID can arise and develop. In this review, we attempt to bring together knowledge available in the literature about lncRNAs involved with molecular and cellular pathways already described in intellectual disability and neural function, to better understand their relevance in NS-ID and the regulatory complexity of this disorder.
Collapse
Affiliation(s)
- Isabela I. Barros
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
| | - Vitor Leão
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
| | - Jessica O. Santis
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
| | - Reginaldo C. A. Rosa
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
| | - Danielle B. Brotto
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
| | - Camila B. Storti
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
| | - Ádamo D. D. Siena
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
| | - Greice A. Molfetta
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
| | - Wilson A. Silva
- Department of Genetics at the Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Monte Alegre, Ribeirão Preto, São Paulo 14049-900, Brazil; (I.I.B.); (V.L.); (J.O.S.); (R.C.A.R.); (D.B.B.); (C.B.S.); (Á.D.D.S.); (G.A.M.)
- National Institute of Science and Technology in Stem Cell and Cell Therapy and Center for Cell Based Therapy, Ribeirão Preto Medical School, University of São Paulo, Rua Tenente Catão Roxo, 2501, Monte Alegre, Ribeirão Preto 14051-140, Brazil
- Center for Integrative Systems Biology-CISBi, NAP/USP, Ribeirão Preto Medical School, University of São Paulo, Rua Catão Roxo, 2501, Monte Alegre, Ribeirão Preto 14051-140, Brazil
- Department of Medicine at the Midwest State University of Paraná-UNICENTRO, and Guarapuava Institute for Cancer Research, Rua Fortim Atalaia, 1900, Cidade dos Lagos, Guarapuava 85100-000, Brazil
- Correspondence: ; Tel.: +55-16-3315-3293
| |
Collapse
|
8
|
Wang W, Chen Y, Zhao J, Chen L, Song W, Li L, Lin GN. Alternatively Splicing Interactomes Identify Novel Isoform-Specific Partners for NSD2. Front Cell Dev Biol 2021; 9:612019. [PMID: 33718354 PMCID: PMC7947288 DOI: 10.3389/fcell.2021.612019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/05/2021] [Indexed: 11/13/2022] Open
Abstract
Nuclear receptor SET domain protein (NSD2) plays a fundamental role in the pathogenesis of Wolf-Hirschhorn Syndrome (WHS) and is overexpressed in multiple human myelomas, but its protein-protein interaction (PPI) patterns, particularly at the isoform/exon levels, are poorly understood. We explored the subcellular localizations of four representative NSD2 transcripts with immunofluorescence microscopy. Next, we used label-free quantification to perform immunoprecipitation mass spectrometry (IP-MS) analyses of the transcripts. Using the interaction partners for each transcript detected in the IP-MS results, we identified 890 isoform-specific PPI partners (83% are novel). These PPI networks were further divided into four categories of the exon-specific interactome. In these exon-specific PPI partners, two genes, RPL10 and HSPA8, were successfully confirmed by co-immunoprecipitation and Western blotting. RPL10 primarily interacted with Isoforms 1, 3, and 5, and HSPA8 interacted with all four isoforms, respectively. Using our extended NSD2 protein interactions, we constructed an isoform-level PPI landscape for NSD2 to serve as reference interactome data for NSD2 spliceosome-level studies. Furthermore, the RNA splicing processes supported by these isoform partners shed light on the diverse roles NSD2 plays in WHS and myeloma development. We also validated the interactions using Western blotting, RPL10, and the three NSD2 (Isoform 1, 3, and 5). Our results expand gene-level NSD2 PPI networks and provide a basis for the treatment of NSD2-related developmental diseases.
Collapse
Affiliation(s)
- Weidi Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China
| | - Yucan Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jingjing Zhao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Liang Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Weichen Song
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Li Li
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guan Ning Lin
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China
| |
Collapse
|
9
|
Fernández-Ponce C, Navarro Quiroz R, Díaz Perez A, Aroca Martinez G, Cadena Bonfanti A, Acosta Hoyos A, Gómez Escorcia L, Hernández Agudelo S, Orozco Sánchez C, Villarreal Camacho J, Atencio Ibarra L, Consuegra Machado J, Espinoza Garavito A, García-Cózar F, Navarro Quiroz E. MicroRNAs overexpressed in Crohn's disease and their interactions with mechanisms of epigenetic regulation explain novel aspects of Crohn's disease pathogenesis. Clin Epigenetics 2021; 13:39. [PMID: 33602320 PMCID: PMC7890887 DOI: 10.1186/s13148-021-01022-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Background In this review, we were interested to identify the wide universe of enzymes associated with epigenetic modifications, whose gene expression is regulated by miRNAs with a high relative abundance in Crohn's disease (CD) affected tissues, with the aim to determine their impact in the pathogenesis and evolution of the disease. Methods We used HMDD and Bibliometrix R-package in order to identify the miRNAs overexpressed in CD. The identified enzymes associated with epigenetic mechanisms and post-translational modifications, regulated by miRNAs upregulated in CD, were analyzed using String v11 database. Results We found 190 miRNAs with great abundance in patients with CD, of which 26 miRNAs regulate the gene expression of enzymes known to catalyze epigenetic modifications involved in essentials pathophysiological processes, such as chromatin architecture reorganization, immune response regulation including CD4+ T cells polarization, integrity of gut mucosa, gut microbiota composition and tumorigenesis. Conclusion The integrated analysis of miRNAs with a high relative abundance in patients with CD showed a combined and superimposed gene expression regulation of enzymes associated with relevant epigenetic mechanisms and that could explain, in part, the pathogenesis of CD. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01022-8.
Collapse
Affiliation(s)
- Cecilia Fernández-Ponce
- Department of Biomedicine, Biotechnology and Public Health, University of Cadiz, Cadiz, Spain
| | - Roberto Navarro Quiroz
- CMCC-Centro de Matemática, Computação E Cognição, Laboratório do Biología Computacional e Bioinformática-LBCB, Universidade Federal Do ABC, Sao Paulo, 01023, Brazil
| | - Anderson Díaz Perez
- Facultad de Ciencias Básicas y Biomédicas, Universidad Simon Bolivar, 080001, Barranquilla, Colombia.,Universidad Rafael Nuñez, 130001, Cartagena, Colombia
| | - Gustavo Aroca Martinez
- Facultad de Ciencias Básicas y Biomédicas, Universidad Simon Bolivar, 080001, Barranquilla, Colombia.,Department of Nephrology, Clinica de La Costa, 080001, Barranquilla, Colombia
| | - Andrés Cadena Bonfanti
- Facultad de Ciencias Básicas y Biomédicas, Universidad Simon Bolivar, 080001, Barranquilla, Colombia.,Department of Nephrology, Clinica de La Costa, 080001, Barranquilla, Colombia
| | - Antonio Acosta Hoyos
- Facultad de Ciencias Básicas y Biomédicas, Universidad Simon Bolivar, 080001, Barranquilla, Colombia
| | - Lorena Gómez Escorcia
- Facultad de Ciencias Básicas y Biomédicas, Universidad Simon Bolivar, 080001, Barranquilla, Colombia.,Universidad Rafael Nuñez, 130001, Cartagena, Colombia
| | - Sandra Hernández Agudelo
- Facultad de Ciencias Básicas y Biomédicas, Universidad Simon Bolivar, 080001, Barranquilla, Colombia.,Department of Nephrology, Clinica de La Costa, 080001, Barranquilla, Colombia
| | - Christian Orozco Sánchez
- Facultad de Ciencias Básicas y Biomédicas, Universidad Simon Bolivar, 080001, Barranquilla, Colombia
| | | | | | | | - Alberto Espinoza Garavito
- Facultad de Ciencias Básicas y Biomédicas, Universidad Simon Bolivar, 080001, Barranquilla, Colombia
| | - Francisco García-Cózar
- Department of Biomedicine, Biotechnology and Public Health, University of Cadiz, Cadiz, Spain
| | - Elkin Navarro Quiroz
- Facultad de Ciencias Básicas y Biomédicas, Universidad Simon Bolivar, 080001, Barranquilla, Colombia. .,Centro de Investigación E Innovación en Biomoléculas, C4U S.A.S, 080001, Barranquilla, Colombia.
| |
Collapse
|
10
|
Novel DNMT3A Germline Variant in a Patient with Multiple Paragangliomas and Papillary Thyroid Carcinoma. Cancers (Basel) 2020; 12:cancers12113304. [PMID: 33182397 PMCID: PMC7697455 DOI: 10.3390/cancers12113304] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The use of next generation technologies has helped to unravel the genetics of rare inherited diseases, facilitating the discovery of new susceptibility genes. Nonetheless, the sequencing of all protein-coding genes of an individual may lead to doubtful assignments of causality for non-pathological variants, so it is mandatory to perform comprehensive studies and screening of additional patients for new mutations. Here we describe a novel DNMT3A germline variant identified by whole-exome sequencing in a patient with multiple paragangliomas and papillary thyroid carcinoma. The increased methylation of DNMT3A target genes observed in the proband’s sample points towards a gain-of-function effect of the variant, contrasting with the inactivation caused by loss-of-function alterations commonly seen in other neoplasia and in patients with Tatton-Brown-Rahman syndrome. This finding stresses the diverse molecular outcomes and suggests a heterogeneous phenotypic spectrum related to DNMT3A germline variants. Abstract Over the past few years, next generation technologies have been applied to unravel the genetics of rare inherited diseases, facilitating the discovery of new susceptibility genes. We recently found germline DNMT3A gain-of-function variants in two patients with head and neck paragangliomas causing a characteristic hypermethylated DNA profile. Here, whole-exome sequencing identifies a novel germline DNMT3A variant (p.Gly332Arg) in a patient with bilateral carotid paragangliomas, papillary thyroid carcinoma and idiopathic intellectual disability. The variant, located in the Pro-Trp-Trp-Pro (PWWP) domain of the protein involved in chromatin targeting, affects a residue mutated in papillary thyroid tumors and located between the two residues found mutated in microcephalic dwarfism patients. Structural modelling of the variant in the DNMT3A PWWP domain predicts that the interaction with H3K36me3 will be altered. An increased methylation of DNMT3A target genes, compatible with a gain-of-function effect of the alteration, was observed in saliva DNA from the proband and in one independent acute myeloid leukemia sample carrying the same p.Gly332Arg variant. Although further studies are needed to support a causal role of DNMT3A variants in paraganglioma, the description of a new DNMT3A alteration in a patient with multiple clinical features suggests a heterogeneous phenotypic spectrum related to DNMT3A germline variants.
Collapse
|
11
|
Yang R, Wang M, Zhang G, Bao Y, Wu Y, Li X, Yang W, Cui H. E2F7-EZH2 axis regulates PTEN/AKT/mTOR signalling and glioblastoma progression. Br J Cancer 2020; 123:1445-1455. [PMID: 32814835 PMCID: PMC7591888 DOI: 10.1038/s41416-020-01032-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/24/2020] [Accepted: 07/27/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND E2F transcription factors are considered to be important drivers of tumour growth. E2F7 is an atypical E2F factor, and its role in glioblastoma remains undefined. METHODS E2F7 expression was examined in patients by IHC and qRT-PCR. The overall survival probability was determined by statistical analyses. MTT assay, colony formation, cell-cycle assay, cell metastasis and the in vivo model were employed to determine the functional role of E2F7 in glioblastoma. Chromatin immunoprecipitation, luciferase assay and western blot were used to explore the underlying mechanisms. RESULTS E2F7 was found to be up-regulated in glioblastoma patients, and high E2F7 expression was associated with poor overall survival in glioblastoma patients. Functional studies showed that E2F7 promoted cell proliferation, cell-cycle progression, cell metastasis and tumorigenicity abilities in vitro and in vivo. E2F7 promoted the transcription of EZH2 by binding to its promoter and increased H3K27me3 level. EZH2 recruited H3K27me3 to the promoter of PTEN and inhibited PTEN expression, and then activated the AKT/mTOR signalling pathway. In addition, restored expression of EZH2 recovered the abilities of cell proliferation and metastasis in E2F7-silencing cells. CONCLUSION Collectively, our findings indicate that E2F7 promotes cell proliferation, cell metastasis and tumorigenesis via EZH2-mediated PTEN/AKT/mTOR pathway in glioblastoma.
Collapse
Affiliation(s)
- Rui Yang
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, China. .,State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China.
| | - Mei Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China.,Guizhou Provincial College-based Key Laboratory for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi, China
| | - Guanghui Zhang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China.,Cancer Center, Medical Research Institute, Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, China
| | - Yonghua Bao
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Yanan Wu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China.,Cancer Center, Medical Research Institute, Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, China
| | - Xiuxiu Li
- Department of Pharmacy, The Second People's Hospital of Liaocheng, Liaocheng, China
| | - Wancai Yang
- Key Laboratory of Precision Oncology of Shandong Higher Education, Institute of Precision Medicine, Jining Medical University, Jining, China.,Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China. .,Cancer Center, Medical Research Institute, Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, China.
| |
Collapse
|
12
|
Belpinati F, Malerba G, Dal Toè M, Ceccuzzi L, Rodolfo M, Poli A, Turco A, Vergani E, Sangalli A, Gomez-Lira M. Enhancer of zeste 2 polycomb repressive complex 2 subunit polymorphisms in melanoma skin cancer risk. Exp Dermatol 2020; 29:980-986. [PMID: 32748461 DOI: 10.1111/exd.14163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 11/27/2022]
Abstract
Melanoma is the most deadly skin cancer, and its incidence is growing. EZH2, a member of the Polycomb Group (PcGs) proteins family, plays an important biological role in the occurrence and development of melanoma. EZH2 germline genetic polymorphisms have not been yet evaluated in melanoma predisposition. Three hundred thirty sporadic Italian melanoma patients and 333 healthy volunteers were genotyped to analyse the association between EZH2 variants rs6950683, rs2302427, rs3757441, rs2072408 and melanoma risk. The functionality of rs6950683 alleles was investigated in keratinocytes (HaCat), melanoma cells (A375) and human embryonic kidney cells (HEK293), using promoter-reporter assays. Genotype distribution of SNPs showed that rs6950683T and rs3757441C alleles were positively associated with melanoma risk (P = .003 and .004, respectively). Haplotype analysis revealed that TCCA and CCCG haplotypes were associated with a higher risk of melanoma (P = .02 and .04, respectively). Functional assays demonstrated that allele rs6950683T reduce promoter activity in the three cell lines analysed compared to C allele. rs6950683T and rs3757441C alleles in the EZH2 gene appear positively associated with melanoma risk in the analysed population. In addition, we demonstrated for the first time the functional role of rs6950683 upstream polymorphism on EZH2 gene expression regulation.
Collapse
Affiliation(s)
- Francesca Belpinati
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biology and Genetics, University of Verona, Verona, Italy
| | - Giovanni Malerba
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biology and Genetics, University of Verona, Verona, Italy
| | - Melissa Dal Toè
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biology and Genetics, University of Verona, Verona, Italy
| | - Laura Ceccuzzi
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biology and Genetics, University of Verona, Verona, Italy
| | - Monica Rodolfo
- Melanoma and Sarcoma Surgery, Unit of Immunotherapy, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Albino Poli
- Department of Diagnostic and Public Health, Environmental and Occupational Medicine, Section of Hygiene and Preventive, University of Verona, Verona, Italy
| | - Alberto Turco
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biology and Genetics, University of Verona, Verona, Italy
| | - Elisabetta Vergani
- Melanoma and Sarcoma Surgery, Unit of Immunotherapy, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Antonella Sangalli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biology and Genetics, University of Verona, Verona, Italy
| | - Macarena Gomez-Lira
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biology and Genetics, University of Verona, Verona, Italy
| |
Collapse
|
13
|
Yang YX, Shen HH, Cao F, Xie LY, Zhu GL, Sam NB, Wang DG, Pan HF. Therapeutic potential of enhancer of zeste homolog 2 in autoimmune diseases. Expert Opin Ther Targets 2019; 23:1015-1030. [PMID: 31747802 DOI: 10.1080/14728222.2019.1696309] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: Autoimmune diseases (ADs) are idiopathic and heterogeneous disorders with contentious pathophysiology. Great strides have been made in epigenetics and its involvement in ADs. Zeste homolog 2 (EZH2) has sparked extensive interest because of its pleiotropic roles in distinct pathologic contexts.Areas covered: This review summarizes the epigenetic functions and the biological significance of EZH2 in the etiology of rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), type 1 diabetes (T1D), inflammatory bowel disease (IBD), multiple sclerosis (MS), and systemic sclerosis (SSc). A brief recapitulation of the therapeutic potential of EZH2 targeting is provided.Expert opinion: There are questions marks and controversies surrounding the feasibility and safety of EZH2 targeting; it is recommended in RA and SLE, but queried in T1D, IBD, MS, and SSc. Future work should focus on contrast studies, systematic analyses and preclinical studies with optimizing methodologies. Selective research studies conducted in a stage-dependent manner are necessary because of the relapsing-remitting clinical paradigms.
Collapse
Affiliation(s)
- Yue-Xin Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hui-Hui Shen
- Department of Clinical Medicine, The second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Fan Cao
- Department of Clinical Medicine, The second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Liang-Yu Xie
- Department of Clinical Medicine, The second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Guang-Lin Zhu
- Department of Clinical Medicine, The second School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Napoleon Bellua Sam
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, China
| | - De-Guang Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, China
| |
Collapse
|
14
|
Peng B, Han X, Peng C, Luo X, Deng L, Huang L. G9α-dependent histone H3K9me3 hypomethylation promotes overexpression of cardiomyogenesis-related genes in foetal mice. J Cell Mol Med 2019; 24:1036-1045. [PMID: 31746096 PMCID: PMC6933410 DOI: 10.1111/jcmm.14824] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/08/2019] [Accepted: 09/16/2019] [Indexed: 12/19/2022] Open
Abstract
Alcohol consumption during pregnancy can cause foetal alcohol syndrome and congenital heart disease. Nonetheless, the underlying mechanism of alcohol‐induced cardiac dysplasia remains unknown. We previously reported that alcohol exposure during pregnancy can cause abnormal expression of cardiomyogenesis‐related genes, and histone H3K9me3 hypomethylation was observed in alcohol‐treated foetal mouse heart. Hence, an imbalance in histone methylation may be involved in alcohol‐induced cardiac dysplasia. In this study, we investigated the involvement of G9α histone methyltransferase in alcohol‐induced cardiac dysplasia in vivo and in vitro using heart tissues of foetal mice and primary cardiomyocytes of neonatal mice. Western blotting revealed that alcohol caused histone H3K9me3 hypomethylation by altering G9α histone methyltransferase expression in cardiomyocytes. Moreover, overexpression of cardiomyogenesis‐related genes (MEF2C, Cx43, ANP and β‐MHC) was observed in alcohol‐exposed foetal mouse heart. Additionally, we demonstrated that G9α histone methyltransferase directly interacted with histone H3K9me3 and altered its methylation. Notably, alcohol did not down‐regulate H3K9me3 methylation after G9α suppression by short hairpin RNA in primary mouse cardiomyocytes, preventing MEF2C, Cx43, ANP and β‐MHC overexpression. These findings suggest that G9α histone methyltransferase‐mediated imbalance in histone H3K9me3 methylation plays a critical role in alcohol‐induced abnormal expression cardiomyogenesis‐related genes during pregnancy. Therefore, G9α histone methyltransferase may be an intervention target for congenital heart disease.
Collapse
Affiliation(s)
- Bohui Peng
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiao Han
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chang Peng
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaomei Luo
- Department of Physiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi, China
| | - Ling Deng
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lixin Huang
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
15
|
Zheng Y, Li B, Wang J, Xiong Y, Wang K, Qi Y, Sun H, Wu L, Yang L. Identification of SUV39H2 as a potential oncogene in lung adenocarcinoma. Clin Epigenetics 2018; 10:129. [PMID: 30348215 PMCID: PMC6198372 DOI: 10.1186/s13148-018-0562-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/09/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND SUV39H2 (suppressor of variegation 3-9 homolog 2), which introduces H3K9me3 to induce transcriptional repression, has been reported to play critical roles in heterochromatin maintenance, DNA repair, and recently, carcinogenesis. Dysregulation of SUV39H2 expression has been observed in several types of cancers. However, neither the genomic landscape nor the clinical significance of SUV39H2 in lung adenocarcinoma has been probed comprehensively. METHODS In this research, we conducted bioinformatics analysis to primarily sort out potential genes with dysregulated expressions. After we identified SUV39H2, RNA-seq was performed for a high-throughput evaluation of altered gene expression and dysregulated pathways, followed by a series of validations via RT-qPCR and bioinformatics analyses. Finally, to assess the potential oncogenic role of SUV39H2, we employed the invasion assay and clone formation assay in vitro and tumorigenesis assays in mouse models in vivo. RESULTS Through bioinformatics analyses, we found that SUV39H2 underwent a severe upregulation in the tumor tissue, which was also confirmed in the surgically removed tissues. Overexpression of SUV39H2 was mainly associated with its amplification and with shorter patient overall survival. Then, the RNA-seq demonstrated that TPM4, STOM, and OPTN might be affected by the loss of function of SUV39H2. Finally, in vitro and in vivo experiments with SUV39H2 knockdown all suggested a potential role of SUV39H2 in both carcinogenesis and metastasis. CONCLUSIONS SUV39H2 expression was elevated in lung adenocarcinoma. TPM4, OPTN, and STOM were potentially regulated by SUV39H2. SUV39H2 might be a potential oncogene in lung adenocarcinoma, mediating tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Yu Zheng
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Baihui Li
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Jian Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Yanjuan Xiong
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Kaiyuan Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Ying Qi
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Houfang Sun
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Lei Wu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Lili Yang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China. .,National Clinical Research Center for Cancer, Tianjin, China. .,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China. .,Tianjin's Clinical Research Center for Cancer, Tianjin, China. .,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.
| |
Collapse
|
16
|
Katoh M. Multi‑layered prevention and treatment of chronic inflammation, organ fibrosis and cancer associated with canonical WNT/β‑catenin signaling activation (Review). Int J Mol Med 2018; 42:713-725. [PMID: 29786110 PMCID: PMC6034925 DOI: 10.3892/ijmm.2018.3689] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/16/2018] [Indexed: 12/13/2022] Open
Abstract
β-catenin/CTNNB1 is an intracellular scaffold protein that interacts with adhesion molecules (E-cadherin/CDH1, N-cadherin/CDH2, VE-cadherin/CDH5 and α-catenins), transmembrane-type mucins (MUC1/CD227 and MUC16/CA125), signaling regulators (APC, AXIN1, AXIN2 and NHERF1/EBP50) and epigenetic or transcriptional regulators (BCL9, BCL9L, CREBBP/CBP, EP300/p300, FOXM1, MED12, SMARCA4/BRG1 and TCF/LEF). Gain-of-function CTTNB1 mutations are detected in bladder cancer, colorectal cancer, gastric cancer, liver cancer, lung cancer, pancreatic cancer, prostate cancer and uterine cancer, whereas loss-of-function CTNNB1 mutations are also detected in human cancer. ABCB1, ALDH1A1, ASCL2, ATF3, AXIN2, BAMBI, CCND1, CD44, CLDN1, CTLA4, DKK1, EDN1, EOMES, FGF18, FGF20, FZD7, IL10, JAG1, LEF1, LGR5, MITF, MSX1, MYC, NEUROD1, NKD1, NODAL, NOTCH2, NOTUM, NRCAM, OPN, PAX3, PPARD, PTGS2, RNF43, SNAI1, SP5, TCF7, TERT, TNFRSF19, VEGFA and ZNRF3 are representative β-catenin target genes. β-catenin signaling is involved in myofibroblast activation and subsequent pulmonary fibrosis, in addition to other types of fibrosis. β-catenin and NF-κB signaling activation are involved in field cancerization in the stomach associated with Helicobacter pylori (H. pylori) infection and in the liver associated with hepatitis C virus (HCV) infection and other etiologies. β-catenin-targeted therapeutics are functionally classified into β-catenin inhibitors targeting upstream regulators (AZ1366, ETC-159, G007-LK, GNF6231, ipafricept, NVP-TNKS656, rosmantuzumab, vantictumab, WNT-C59, WNT974 and XAV939), β-catenin inhibitors targeting protein-protein interactions (CGP049090, CWP232228, E7386, ICG-001, LF3 and PRI-724), β-catenin inhibitors targeting epigenetic regulators (PKF118-310), β-catenin inhibitors targeting mediator complexes (CCT251545 and cortistatin A) and β-catenin inhibitors targeting transmembrane-type transcriptional outputs, including CD44v6, FZD7 and LGR5. Eradicating H. pylori and HCV is the optimal approach for the first-line prevention of gastric cancer and hepatocellular carcinoma (HCC), respectively. However, β-catenin inhibitors may be applicable for the prevention of organ fibrosis, second-line HCC prevention and treating β-catenin-driven cancer. The multi-layered prevention and treatment strategy of β-catenin-related human diseases is necessary for the practice of personalized medicine and implementation of precision medicine.
Collapse
Affiliation(s)
- Masaru Katoh
- Department of Omics Network, National Cancer Center, Chuo Ward, Tokyo 104‑0045, Japan
| |
Collapse
|
17
|
Katoh M. Combination immuno-oncology therapy with immune checkpoint blockers targeting PD-L1, PD-1 or CTLA4 and epigenetic drugs targeting MYC and immune evasion for precision medicine. J Thorac Dis 2018; 10:1294-1299. [PMID: 29708143 DOI: 10.21037/jtd.2018.03.62] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Masaru Katoh
- Department of Omics Network, National Cancer Center Tokyo, Japan
| |
Collapse
|
18
|
Abstract
The ARF and INK4a genes are located in the same CDKN2a locus, both showing its tumor suppressive activity. ARF has been shown to detect potentially harmful oncogenic signals, making incipient cancer cells undergo senescence or apoptosis. INK4a, on the other hand, responds to signals from aging in a variety of tissues including islets of Langerhans, neuronal cells, and cancer stem cells in general. It also detects oncogenic signals from incipient cancer cells to induce them senescent to prevent neoplastic transformation. Both of these genes are inactivated by gene deletion, promoter methylation, frame shift, and aberrant splicing although mutations changing the amino acid sequences affect only the latter. Recent studies indicated that polycomb gene products EZH2 and BMI1 repressed p16INK4a expression in primary cells, but not in cells deficient for pRB protein function. It was also reported that that p14ARF inhibits the stability of the p16INK4a protein in human cancer cell lines and mouse embryonic fibroblasts through its interaction with regenerating islet-derived protein 3γ. Overexpression of INK4a is associated with better prognosis of cancer when it is associated with human papilloma virus infection. However, it has a worse prognostic value in other tumors since it is an indicator of pRB loss. The p16INK4a tumor suppressive protein can thus be used as a biomarker to detect early stage cancer cells as well as advanced tumor cells with pRB inactivation since it is not expressed in normal cells.
Collapse
Affiliation(s)
- Kazushi Inoue
- The Department of Pathology, Wake Forest University Health Sciences, Winston-Salem, NC 27157
| | - Elizabeth A Fry
- The Department of Pathology, Wake Forest University Health Sciences, Winston-Salem, NC 27157
| |
Collapse
|
19
|
Zhou X, Gu Y, Han Q, Soliman M, Song C, Ge Z. Coexistence of EZH2, NOTCH1, IL7R, and PHF6 Mutations in Adult T-cell Acute Lymphoblastic Leukemia. Turk J Haematol 2017; 34:366-368. [PMID: 28747286 PMCID: PMC5774360 DOI: 10.4274/tjh.2017.0194] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Affiliation(s)
- Xilian Zhou
- Zhongda Hospital, Medical School of Southeast University Department of Hematology Nanjing, China
| | - Yan Gu
- Zhongda Hospital, Medical School of Southeast University Department of Hematology Nanjing, China
| | - Qi Han
- Zhongda Hospital, Medical School of Southeast University Department of Hematology Nanjing, China
| | - Mario Soliman
- Pennsylvania State University, Department of Pediatrics, Pennsylvania, USA
| | - Chunhua Song
- Pennsylvania State University, Department of Pediatrics, Pennsylvania, USA
| | - Zheng Ge
- Zhongda Hospital, Medical School of Southeast University Department of Hematology Nanjing, China
| |
Collapse
|
20
|
Katoh M, Katoh M. Molecular genetics and targeted therapy of WNT-related human diseases (Review). Int J Mol Med 2017; 40:587-606. [PMID: 28731148 PMCID: PMC5547940 DOI: 10.3892/ijmm.2017.3071] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 07/12/2017] [Indexed: 12/15/2022] Open
Abstract
Canonical WNT signaling through Frizzled and LRP5/6 receptors is transduced to the WNT/β-catenin and WNT/stabilization of proteins (STOP) signaling cascades to regulate cell fate and proliferation, whereas non-canonical WNT signaling through Frizzled or ROR receptors is transduced to the WNT/planar cell polarity (PCP), WNT/G protein-coupled receptor (GPCR) and WNT/receptor tyrosine kinase (RTK) signaling cascades to regulate cytoskeletal dynamics and directional cell movement. WNT/β-catenin signaling cascade crosstalks with RTK/SRK and GPCR-cAMP-PKA signaling cascades to regulate β-catenin phosphorylation and β-catenin-dependent transcription. Germline mutations in WNT signaling molecules cause hereditary colorectal cancer, bone diseases, exudative vitreoretinopathy, intellectual disability syndrome and PCP-related diseases. APC or CTNNB1 mutations in colorectal, endometrial and prostate cancers activate the WNT/β-catenin signaling cascade. RNF43, ZNRF3, RSPO2 or RSPO3 alterations in breast, colorectal, gastric, pancreatic and other cancers activate the WNT/β-catenin, WNT/STOP and other WNT signaling cascades. ROR1 upregulation in B-cell leukemia and solid tumors and ROR2 upregulation in melanoma induce invasion, metastasis and therapeutic resistance through Rho-ROCK, Rac-JNK, PI3K-AKT and YAP signaling activation. WNT signaling in cancer, stromal and immune cells dynamically orchestrate immune evasion and antitumor immunity in a cell context-dependent manner. Porcupine (PORCN), RSPO3, WNT2B, FZD5, FZD10, ROR1, tankyrase and β-catenin are targets of anti-WNT signaling therapy, and ETC-159, LGK974, OMP-18R5 (vantictumab), OMP-54F28 (ipafricept), OMP-131R10 (rosmantuzumab), PRI-724 and UC-961 (cirmtuzumab) are in clinical trials for cancer patients. Different classes of anti-WNT signaling therapeutics are necessary for the treatment of APC/CTNNB1-, RNF43/ZNRF3/RSPO2/RSPO3- and ROR1-types of human cancers. By contrast, Dickkopf-related protein 1 (DKK1), SOST and glycogen synthase kinase 3β (GSK3β) are targets of pro-WNT signaling therapy, and anti-DKK1 (BHQ880 and DKN-01) and anti-SOST (blosozumab, BPS804 and romosozumab) monoclonal antibodies are being tested in clinical trials for cancer patients and osteoporotic post-menopausal women. WNT-targeting therapeutics have also been applied as reagents for in vitro stem-cell processing in the field of regenerative medicine.
Collapse
Affiliation(s)
| | - Masaru Katoh
- Department of Omics Network, National Cancer Center, Tokyo 104-0045, Japan
| |
Collapse
|
21
|
Kurmasheva RT, Sammons M, Favours E, Wu J, Kurmashev D, Cosmopoulos K, Keilhack H, Klaus CR, Houghton PJ, Smith MA. Initial testing (stage 1) of tazemetostat (EPZ-6438), a novel EZH2 inhibitor, by the Pediatric Preclinical Testing Program. Pediatr Blood Cancer 2017; 64:10.1002/pbc.26218. [PMID: 27555605 PMCID: PMC5584632 DOI: 10.1002/pbc.26218] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 07/05/2016] [Accepted: 07/26/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND Tazemetostat (EPZ-6438) is a selective inhibitor of the histone methyltransferase EZH2 and currently in clinical development for non-Hodgkin lymphoma and genetically defined tumors. PROCEDURES Tazemetostat was tested against the Pediatric Preclinical Testing Program (PPTP) solid tumor xenografts using a dose of 400 mg/kg administered twice daily by oral gavage for 28 days. H3K27me3:H3 ratios were determined in control and treated tumors. RESULTS Tazemetostat induced significant differences in event-free survival (EFS) distribution compared with control in nine of 30 (30%) of the xenografts studied. Significant differences in EFS distribution were observed in five of seven (71%) rhabdoid tumor xenograft lines compared with four of 23 (17%) nonrhabdoid xenograft lines (chi-square [χ2 ] test P = 0.006). Tazemetostat induced tumor growth inhibition meeting criteria for intermediate and high EFS treated-to-control (T/C) activity in two of 25 (8%) and one of 25 (4%) xenografts, respectively. Intermediate and high activity for the EFS T/C metric was observed exclusively among rhabdoid tumor xenografts (three of five rhabdoid tumor vs 0 of 22 nonrhabdoid tumors (χ² test P < 0.001). One rhabdoid tumor xenograft (G401) showed stable disease. For one rhabdoid tumor (G401), delayed tumor regression to tazemetostat was noted following 1 week of tumor growth. Tazemetostat induced significant reduction of H3K27me3 levels in the majority of tumors compared with controls. CONCLUSIONS Tazemetostat demonstrated significant antitumor activity in rhabdoid tumor models but showed no consistent activity against any other histology. Tazemetostat reduced H3K27me3 levels irrespective of tumor response. Further preclinical testing to evaluate tazemetostat in combination with other anticancer agents is warranted.
Collapse
Affiliation(s)
| | | | | | - Jianwrong Wu
- St. Jude Children’s Research Hospital, Memphis, TN
| | | | | | | | | | | | | |
Collapse
|
22
|
Therapeutics Targeting FGF Signaling Network in Human Diseases. Trends Pharmacol Sci 2016; 37:1081-1096. [DOI: 10.1016/j.tips.2016.10.003] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/06/2016] [Accepted: 10/06/2016] [Indexed: 12/14/2022]
|
23
|
Yao Y, Hu H, Yang Y, Zhou G, Shang Z, Yang X, Sun K, Zhan S, Yu Z, Li P, Pan G, Sun L, Zhu X, He S. Downregulation of Enhancer of Zeste Homolog 2 (EZH2) is essential for the Induction of Autophagy and Apoptosis in Colorectal Cancer Cells. Genes (Basel) 2016; 7:E83. [PMID: 27706111 PMCID: PMC5083922 DOI: 10.3390/genes7100083] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/26/2016] [Indexed: 11/17/2022] Open
Abstract
Increasing evidence indicates that elevated expression of enhancer of zeste homolog 2 gene (EZH2) in many human malignant tumors acts a significant role in the oncogenic process. However, the underlying molecular mechanism is still unclarified. It is evident that apoptosis and autophagy of tumor cells is crucial for the tumorigenesis and progression of cancer, however, the exact role of EZH2 plays in apoptosis and autophagy has not been fully elucidated in colorectal cancer (CRC). Our previous study found that the expression level of EZH2 was higher in CRC tumor tissues than in the paired normal tissues using immunohistochemical analysis. We also recently found that the autophagy-related gene-related protein Ambra1 plays an important role in the autophagy pathway in CRC cells. In this study, mRNA and protein expression of EZH2 in four CRC cell lines were tested at first and RKO and HCT116 cells showed the highest levels among them. Here we transfected with EZH2-shRNA, or added DZNep (an EZH2 inhibitor) to RKO and HCT116 cells in order to detect the effect of EZH2 on autophagy via determining the change of the protein expression of LC3 and Ambra1. The outcome indicated an obvious decrease of autophagy level in cells transfected with EZH2-shRNA or DZNep. We also found the apoptotic rate of cells was elevated significantly after downregulation of EZH2. In addition, compared to control group, CRC cells transfected with EZH2-shRNA or added DZNep revealed a significantly increased G1 cell cycle rate and an obvious decrease in the G2 cell cycle rate. Further analysis showed that knockdown of EZH2 induced cell-cycle arrest in CRC cells. Meanwhile, downregulation of EZH2 in CRC cells induces autophagy and apoptosis. Taken together, our results suggest that EZH2 plays a critical role in autophagy and apoptosis in the progression of CRC, which potentially facilitates the development of an ideal strategy for combating colorectal cancer.
Collapse
Affiliation(s)
- Yizhou Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Hao Hu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Yong Yang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Guoqiang Zhou
- Department of Gastrointestinal Surgery, Changshu No.2 Hospital, Suzhou 215500, China.
| | - Zengfu Shang
- Department of Radiation Medicine, Medical College of Soochow University, Suzhou 215006, China.
| | - Xiaodong Yang
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Kang Sun
- Department of General Surgery, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Shenghua Zhan
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Zhengyuan Yu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Peiyao Li
- Medical Engineering and Maintenance Center, Chinese PLA General Hospital, Beijing 100853, China.
| | - Guofeng Pan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Liang Sun
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Xinguo Zhu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Songbing He
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| |
Collapse
|
24
|
Dysregulation of histone methyltransferases in breast cancer - Opportunities for new targeted therapies? Mol Oncol 2016; 10:1497-1515. [PMID: 27717710 DOI: 10.1016/j.molonc.2016.09.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/14/2016] [Accepted: 09/14/2016] [Indexed: 01/24/2023] Open
Abstract
Histone methyltransferases (HMTs) catalyze the methylation of lysine and arginine residues on histone tails and non-histone targets. These important post-translational modifications are exquisitely regulated and affect chromatin compaction and transcriptional programs leading to diverse biological outcomes. There is accumulating evidence that genetic alterations of several HMTs impinge on oncogenic or tumor-suppressor functions and influence both cancer initiation and progression. HMTs therefore represent an opportunity for therapeutic targeting in those patients with tumors in which HMTs are dysregulated, to reverse the histone marks and transcriptional programs associated with aggressive tumor behavior. In this review, we describe the known histone methyltransferases and their emerging roles in breast cancer tumorigenesis.
Collapse
|