1
|
Zhang S, Guo A, Wang H, Liu J, Dong C, Ren J, Wang G. Oncogenic MORC2 in cancer development and beyond. Genes Dis 2024; 11:861-873. [PMID: 37692502 PMCID: PMC10491978 DOI: 10.1016/j.gendis.2023.05.010] [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: 03/06/2023] [Revised: 05/19/2023] [Accepted: 05/25/2023] [Indexed: 09/12/2023] Open
Abstract
Microrchidia CW-type zinc finger 2 (MORC2) is a member of the MORC superfamily of nuclear proteins. Growing evidence has shown that MORC2 not only participates in gene transcription and chromatin remodeling but also plays a key in human disease and tumor development by regulating the expression of downstream oncogenes or tumor suppressors. The present review provides an updated overview of MORC2 in the aspect of cancer hallmark and therapeutic resistance and summarizes its upstream regulators and downstream target genes. This systematic review may provide a favorable theoretical basis for emerging players of MORC2 in tumor development and new insight into the potential clinical application of basic science discoveries in the future.
Collapse
Affiliation(s)
- Shan Zhang
- Key Laboratory of Cell Biology, Department of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Ayao Guo
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Huan Wang
- Key Laboratory of Cell Biology, Department of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Jia Liu
- Key Laboratory of Cell Biology, Department of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Chenshuang Dong
- Key Laboratory of Cell Biology, Department of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Junyi Ren
- Key Laboratory of Cell Biology, Department of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Guiling Wang
- Key Laboratory of Cell Biology, Department of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| |
Collapse
|
2
|
Zeng S, Yang H, Wang B, Xie Y, Xu K, Liu L, Cao W, Liu X, Tang B, Liu M, Zhang R. The MORC2 p.S87L mutation reduces proliferation of pluripotent stem cells derived from a patient with the spinal muscular atrophy-like phenotype by inhibiting proliferation-related signaling pathways. Neural Regen Res 2024; 19:205-211. [PMID: 37488868 PMCID: PMC10479865 DOI: 10.4103/1673-5374.375347] [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: 11/07/2022] [Revised: 03/04/2023] [Accepted: 03/29/2023] [Indexed: 07/26/2023] Open
Abstract
Mutations in the microrchidia CW-type zinc finger protein 2 (MORC2) gene are the causative agent of Charcot-Marie-Tooth disease type 2Z (CMT2Z), and the hotspot mutation p.S87L is associated with a more severe spinal muscular atrophy-like clinical phenotype. The aims of this study were to determine the mechanism of the severe phenotype caused by the MORC2 p.S87L mutation and to explore potential treatment strategies. Epithelial cells were isolated from urine samples from a spinal muscular atrophy (SMA)-like patient (MORC2 p.S87L), a CMT2Z patient (MORC2 p.Q400R), and a healthy control and induced to generate pluripotent stem cells, which were then differentiated into motor neuron precursor cells. Next-generation RNA sequencing followed by KEGG pathway enrichment analysis revealed that differentially expressed genes involved in the PI3K/Akt and MAPK/ERK signaling pathways were enriched in the p.S87L SMA-like patient group and were significantly downregulated in induced pluripotent stem cells. Reduced proliferation was observed in the induced pluripotent stem cells and motor neuron precursor cells derived from the p.S87L SMA-like patient group compared with the CMT2Z patient group and the healthy control. G0/G1 phase cell cycle arrest was observed in induced pluripotent stem cells derived from the p.S87L SMA-like patient. MORC2 p.S87L-specific antisense oligonucleotides (p.S87L-ASO-targeting) showed significant efficacy in improving cell proliferation and activating the PI3K/Akt and MAPK/ERK pathways in induced pluripotent stem cells. However, p.S87L-ASO-targeting did not rescue proliferation of motor neuron precursor cells. These findings suggest that downregulation of the PI3K/Akt and MAPK/ERK signaling pathways leading to reduced cell proliferation and G0/G1 phase cell cycle arrest in induced pluripotent stem cells might be the underlying mechanism of the severe p.S87L SMA-like phenotype. p.S87L-ASO-targeting treatment can alleviate disordered cell proliferation in the early stage of pluripotent stem cell induction.
Collapse
Affiliation(s)
- Sen Zeng
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Honglan Yang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Binghao Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yongzhi Xie
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Ke Xu
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Lei Liu
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Wanqian Cao
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xionghao Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan Province, China
| | - Beisha Tang
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan Province, China
| | - Mujun Liu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan Province, China
| | - Ruxu Zhang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
3
|
Müller I, Helin K. Keep quiet: the HUSH complex in transcriptional silencing and disease. Nat Struct Mol Biol 2024; 31:11-22. [PMID: 38216658 DOI: 10.1038/s41594-023-01173-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/23/2023] [Indexed: 01/14/2024]
Abstract
The human silencing hub (HUSH) complex is an epigenetic repressor complex whose role has emerged as an important guardian of genome integrity. It protects the genome from exogenous DNA invasion and regulates endogenous retroelements by recruiting histone methyltransferases catalyzing histone 3 lysine 9 trimethylation (H3K9me3) and additional proteins involved in chromatin compaction. In particular, its regulation of transcriptionally active LINE1 retroelements, by binding to and neutralizing LINE1 transcripts, has been well characterized. HUSH is required for mouse embryogenesis and is associated with disease, in particular cancer. Here we provide insights into the structural and biochemical features of the HUSH complex. Furthermore, we discuss the molecular mechanisms by which the HUSH complex is recruited to specific genomic regions and how it silences transcription. Finally, we discuss the role of HUSH complex members in mammalian development, antiretroviral immunity, and diseases such as cancer.
Collapse
Affiliation(s)
- Iris Müller
- Cell Biology Program and Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kristian Helin
- Cell Biology Program and Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- The Institute of Cancer Research, London, UK.
| |
Collapse
|
4
|
Thomas L, Chutani N, R K, Nair AS, Yellapu NK, Karyala P, Pakala SB. Microrchidia 2/histone deacetylase 1 complex regulates E-cadherin gene expression and function. Biochem J 2023; 480:1675-1691. [PMID: 37815456 DOI: 10.1042/bcj20230304] [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: 07/26/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/11/2023]
Abstract
Although Microrchidia 2 (MORC2) is widely overexpressed in human malignancies and linked to cancer cell proliferation, metabolism, and metastasis, the mechanism of action of MORC2 in cancer cell migration and invasion is yet undeciphered. Here, we identified for the first time that MORC2, a chromatin remodeler, regulates E-cadherin expression and, subsequently regulates breast cancer cell migration and invasion. We observed a negative correlation between the expression levels of MORC2 and E-cadherin in breast cancer. Furthermore, the overexpression of MORC2 resulted in decreased expression levels of E-cadherin. In addition, co-immunoprecipitation and chromatin immunoprecipitation assays revealed that MORC2 interacts with HDAC1 and gets recruited onto the E-cadherin promoter to inhibit its transcription, thereby suppress its expression. Consequently, knockdown of HDAC1 in MORC2-overexpressing cells led to reduced cancer cell migration and invasion. Interestingly, we noticed that MORC2-regulated glucose metabolism via c-Myc, and LDHA, also modulates the expression of E-cadherin. Collectively, these results demonstrate for the first time a mechanistic role for MORC2 as an upstream regulator of E-cadherin expression and its associated functions in breast cancer.
Collapse
Affiliation(s)
- Liz Thomas
- Biology Division, Indian Institute of Science Education and Research (IISER) Tirupati, Mangalam, Tirupati 517 507, India
| | - Namita Chutani
- Biology Division, Indian Institute of Science Education and Research (IISER) Tirupati, Mangalam, Tirupati 517 507, India
| | - Krishna R
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala 695 014, India
| | - Asha S Nair
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala 695 014, India
| | - Nanda Kumar Yellapu
- Department of Biostatistics & Data Science, University of Kansas Medical Centre, 3901 Rainbow Boulevard, Kansas City, KS 66160, U.S.A
| | - Prashanthi Karyala
- Department of Biotechnology, Faculty of Life and Allied Health Sciences, Ramaiah University of Applied Sciences, Bengaluru 560054, India
| | - Suresh B Pakala
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500 046, India
| |
Collapse
|
5
|
Chutani N, Ragula S, Syed K, Pakala SB. Novel Insights into the Role of Chromatin Remodeler MORC2 in Cancer. Biomolecules 2023; 13:1527. [PMID: 37892209 PMCID: PMC10605154 DOI: 10.3390/biom13101527] [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: 09/16/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
A newly discovered chromatin remodeler, MORC2, is a Microrchidia (MORC) family member. MORC2 acts as a chromatin remodeler by binding to the DNA and changing chromatin conformation using its ATPase domain. MORC2 is highly expressed in a variety of human cancers. It controls diverse signaling pathways essential for cancer development through its target genes and interacting partners. MORC2 promotes cancer cells' growth, invasion, and migration by regulating the expression of genes involved in these processes. MORC2 is localized primarily in the nucleus and is also found in the cytoplasm. In the cytoplasm, MORC2 interacts with adenosine triphosphate (ATP)-citrate lyase (ACLY) to promote lipogenesis and cholesterogenesis in cancer. In the nucleus, MORC2 interacts with the transcription factor c-Myc to control the transcription of genes involved in glucose metabolism to drive cancer cell migration and invasion. Furthermore, MORC2 recruits on to the promoters of tumor suppressor genes to repress their transcription and expression to promote oncogenesis. In addition to its crucial function in oncogenesis, it plays a vital role in DNA repair. Overall, this review concisely summarizes the current knowledge about MORC2-regulated molecular pathways involved in cancer.
Collapse
Affiliation(s)
- Namita Chutani
- Biology Division, Indian Institute of Science Education and Research (IISER) Tirupati, Mangalam, Tirupati 517 507, India;
| | - Sandhya Ragula
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500 046, India;
| | - Khajamohiddin Syed
- Department of Biochemistry and Microbiology, Faculty of Science, Agriculture and Engineering, University of Zululand, KwaDlangezwa 3886, South Africa;
| | - Suresh B. Pakala
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500 046, India;
- Department of Biochemistry and Microbiology, Faculty of Science, Agriculture and Engineering, University of Zululand, KwaDlangezwa 3886, South Africa;
| |
Collapse
|
6
|
Zhao P, Ning J, Huang J, Wei B, Wang Z, Huang X. High Expression of MORC2 is Associated with Poor Clinical Outcomes and Immune Infiltrates in Colon Adenocarcinoma. Int J Gen Med 2023; 16:4595-4615. [PMID: 37850194 PMCID: PMC10577261 DOI: 10.2147/ijgm.s420715] [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: 05/20/2023] [Accepted: 09/07/2023] [Indexed: 10/19/2023] Open
Abstract
Purpose Microrchidia 2 (MORC2) is a universally expressed molecule that has recently been identified as a chromatin modulator and elevated in many malignancies. However, its prognostic value and immunological role of MORC2 in colon adenocarcinoma (COAD) have never been illustrated. Methods The clinical parameters and MORC2 expression datasets of COAD patients were obtained from The Cancer Genome Atlas (TCGA). Cancer and adjacent tissue specimens from surgically resected COAD patients were collected, and quantitative real-time PCR was used to detect MORC2 expression. Differentially expressed genes related to MORC2 were discovered and used for functional enrichment analysis. The diagnostic and prognostic values of MORC2 in COAD were conducted using receiver operating characteristics (ROC), Kaplan-Meier survival curve analysis, PrognoScan, Gene Expression Profiling Interactive Analysis (GEPIA) public databases and nomograms. Eventually, the association of MORC2 with tumor microenvironment was analyzed by using TIMER and GSVA package of R (v3.6.3). Results MORC2 expression was upregulated in COAD tissues, and the RT-qPCR results further verified the reliability of our differential analysis at the transcriptional level. Additionally, higher expression of MORC2 was correlated to a poor prognosis for COAD patients. MORC2 was an independent prognostic factor for COAD and could be a diagnostic factor for early COAD. Furthermore, MORC2 expression was positively correlated with immune cells such as NK cells, TFH cells and so on. Conclusion The findings demonstrated that overexpression of MORC2 was correlated with worse prognosis and immune infiltrates of COAD. MORC2 can serve as a reliable diagnostic and prognostic biomarker and a target of immunotherapy for COAD patients.
Collapse
Affiliation(s)
- Peizhuang Zhao
- Department of Geriatrics and Gastroenterology, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Jiajia Ning
- Department of Geriatrics and Gastroenterology, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Jun Huang
- Department of Geriatrics and Gastroenterology, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Binqian Wei
- Department of Geriatrics and Gastroenterology, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Zhen Wang
- Department of Geriatrics and Gastroenterology, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Xue Huang
- Department of Geriatrics and Gastroenterology, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| |
Collapse
|
7
|
Badie A, Gaiddon C, Mellitzer G. Histone Deacetylase Functions in Gastric Cancer: Therapeutic Target? Cancers (Basel) 2022; 14:5472. [PMID: 36358890 PMCID: PMC9659209 DOI: 10.3390/cancers14215472] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 09/05/2023] Open
Abstract
Gastric cancer (GC) is one of the most aggressive cancers. Therapeutic treatments are based on surgery combined with chemotherapy using a combination of platinum-based agents. However, at metastatic stages of the disease, survival is extremely low due to late diagnosis and resistance mechanisms to chemotherapies. The development of new classifications has not yet identified new prognostic markers for clinical use. The studies of epigenetic processes highlighted the implication of histone acetylation status, regulated by histone acetyltransferases (HATs) and by histone deacetylases (HDACs), in cancer development. In this way, inhibitors of HDACs (HDACis) have been developed and some of them have already been clinically approved to treat T-cell lymphoma and multiple myeloma. In this review, we summarize the regulations and functions of eighteen HDACs in GC, describing their known targets, involved cellular processes, associated clinicopathological features, and impact on survival of patients. Additionally, we resume the in vitro, pre-clinical, and clinical trials of four HDACis approved by Food and Drug Administration (FDA) in cancers in the context of GC.
Collapse
Affiliation(s)
| | | | - Georg Mellitzer
- Laboratoire Streinth, Université de Strasbourg, Inserm UMR_S 1113 IRFAC, 67200 Strasbourg, France
| |
Collapse
|
8
|
Gupta S, Panda PK, Luo W, Hashimoto RF, Ahuja R. Network analysis reveals that the tumor suppressor lncRNA GAS5 acts as a double-edged sword in response to DNA damage in gastric cancer. Sci Rep 2022; 12:18312. [PMID: 36316351 PMCID: PMC9622883 DOI: 10.1038/s41598-022-21492-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/28/2022] [Indexed: 11/14/2022] Open
Abstract
The lncRNA GAS5 acts as a tumor suppressor and is downregulated in gastric cancer (GC). In contrast, E2F1, an important transcription factor and tumor promoter, directly inhibits miR-34c expression in GC cell lines. Furthermore, in the corresponding GC cell lines, lncRNA GAS5 directly targets E2F1. However, lncRNA GAS5 and miR-34c remain to be studied in conjunction with GC. Here, we present a dynamic Boolean network to classify gene regulation between these two non-coding RNAs (ncRNAs) in GC. This is the first study to show that lncRNA GAS5 can positively regulate miR-34c in GC through a previously unknown molecular pathway coupling lncRNA/miRNA. We compared our network to several in-vivo/in-vitro experiments and obtained an excellent agreement. We revealed that lncRNA GAS5 regulates miR-34c by targeting E2F1. Additionally, we found that lncRNA GAS5, independently of p53, inhibits GC proliferation through the ATM/p38 MAPK signaling pathway. Accordingly, our results support that E2F1 is an engaging target of drug development in tumor growth and aggressive proliferation of GC, and favorable results can be achieved through tumor suppressor lncRNA GAS5/miR-34c axis in GC. Thus, our findings unlock a new avenue for GC treatment in response to DNA damage by these ncRNAs.
Collapse
Affiliation(s)
- Shantanu Gupta
- grid.11899.380000 0004 1937 0722Departamento de Ciência da Computação, Instituto de Matemática e Estatística, Universidade de São Paulo, Rua do Matão 1010, São Paulo, SP 05508-090 Brasil
| | - Pritam Kumar Panda
- grid.8993.b0000 0004 1936 9457Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, 751 20 Uppsala, Sweden
| | - Wei Luo
- grid.8993.b0000 0004 1936 9457Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, 751 20 Uppsala, Sweden
| | - Ronaldo F. Hashimoto
- grid.11899.380000 0004 1937 0722Departamento de Ciência da Computação, Instituto de Matemática e Estatística, Universidade de São Paulo, Rua do Matão 1010, São Paulo, SP 05508-090 Brasil
| | - Rajeev Ahuja
- grid.8993.b0000 0004 1936 9457Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, 751 20 Uppsala, Sweden ,grid.462391.b0000 0004 1769 8011Department of Physics, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001 India
| |
Collapse
|
9
|
Cao Y, Ning B, Tian Y, Lan T, Chu Y, Ren F, Wang Y, Meng Q, Li J, Jia B, Chang Z. CREPT Disarms the Inhibitory Activity of HDAC1 on Oncogene Expression to Promote Tumorigenesis. Cancers (Basel) 2022; 14:cancers14194797. [PMID: 36230720 PMCID: PMC9562184 DOI: 10.3390/cancers14194797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/31/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary It has been proposed that highly expressed HDAC1 (histone deacetylases 1) removes the acetyl group from the histones at the promoter regions of tumor suppressor genes to block their expression in tumors. We here revealed the underlying mechanism that HDAC1 differentially regulates the expression of oncogenes and tumor suppressors. In detail, we found that HDAC1 is unable to occupy the promoters of oncogenes but maintains its occupancy with the tumor suppressors due to its interaction with an oncoprotein, CREPT (cell cycle-related and expression-elevated protein in tumor). Abstract Histone deacetylases 1 (HDAC1), an enzyme that functions to remove acetyl molecules from ε-NH3 groups of lysine in histones, eliminates the histone acetylation at the promoter regions of tumor suppressor genes to block their expression during tumorigenesis. However, it remains unclear why HDAC1 fails to impair oncogene expression. Here we report that HDAC1 is unable to occupy at the promoters of oncogenes but maintains its occupancy with the tumor suppressors due to its interaction with CREPT (cell cycle-related and expression-elevated protein in tumor, also named RPRD1B), an oncoprotein highly expressed in tumors. We observed that CREPT competed with HDAC1 for binding to oncogene (such as CCND1, CLDN1, VEGFA, PPARD and BMP4) promoters but not the tumor suppressor gene (such as p21 and p27) promoters by a chromatin immunoprecipitation (ChIP) qPCR experiment. Using immunoprecipitation experiments, we deciphered that CREPT specifically occupied at the oncogene promoter via TCF4, a transcription factor activated by Wnt signaling. In addition, we performed a real-time quantitative PCR (qRT-PCR) analysis on cells that stably over-expressed CREPT and/or HDAC1, and we propose that HDAC1 inhibits CREPT to activate oncogene expression under Wnt signaling activation. Our findings revealed that HDAC1 functions differentially on tumor suppressors and oncogenes due to its interaction with the oncoprotein CREPT.
Collapse
Affiliation(s)
- Yajun Cao
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Bobin Ning
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing 100039, China
| | - Ye Tian
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Tingwei Lan
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yunxiang Chu
- Department of Gastroenterology, Emergency General Hospital, Beijing 100028, China
| | - Fangli Ren
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yinyin Wang
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Qingyu Meng
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing 100039, China
| | - Jun Li
- Qingda Cell Biotech Inc., Beijing 100084, China
| | - Baoqing Jia
- Department of General Surgery, The First Medical Centre, Chinese PLA General Hospital, Beijing 100039, China
- Correspondence: (B.J.); (Z.C.); Tel.: +86-(10)-62773624 (B.J.); +86-(10)-62785076 (Z.C.)
| | - Zhijie Chang
- State Key Laboratory of Membrane Biology, School of Medicine, Tsinghua University, Beijing 100084, China
- Correspondence: (B.J.); (Z.C.); Tel.: +86-(10)-62773624 (B.J.); +86-(10)-62785076 (Z.C.)
| |
Collapse
|
10
|
Saroha HS, Kumar Guddeti R, Jacob JP, Kumar Pulukuri K, Karyala P, Pakala SB. MORC2/β-catenin signaling axis promotes proliferation and migration of breast cancer cells. Med Oncol 2022; 39:135. [PMID: 35727356 DOI: 10.1007/s12032-022-01728-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/05/2022] [Indexed: 02/06/2023]
Abstract
Although Microrchidia 2 (MORC2) is overexpressed in many types of human cancer, its role in breast cancer progression remains unknown. Here, we report that the chromatin remodeler MORC2 expression positively correlates with β-catenin expression in breast cancer cell lines and patients. Overexpression of MORC2 augmented the expression of β-catenin and its target genes, cyclin D1 and c-Myc. Consistent with these results, we found MORC2 knockdown resulted in decreased expression of β-catenin and its target genes. Surprisingly, we observed that c-Myc, the target gene of β-catenin, regulated the MORC2-β-catenin signaling axis through a feedback mechanism. We demonstrated that MORC2 regulates β-catenin expression and function by modulating the phosphorylation of AKT. In addition, we observed reduced proliferation and migration of MORC2 overexpressing breast cancer cells upon β-catenin inhibition. Overall, our results demonstrate that MORC2 promotes breast cancer cell proliferation and migration by regulating β-catenin signaling.
Collapse
Affiliation(s)
- Himanshu Singh Saroha
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Karakambadi Road, Mangalam, Tirupati, Andhra Pradesh, 517507, India
| | - Rohith Kumar Guddeti
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Karakambadi Road, Mangalam, Tirupati, Andhra Pradesh, 517507, India
| | - Jasmine P Jacob
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Tirupati, Karakambadi Road, Mangalam, Tirupati, Andhra Pradesh, 517507, India
| | - Kiran Kumar Pulukuri
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Tirupati, Karakambadi Road, Mangalam, Tirupati, Andhra Pradesh, 517507, India
| | - Prashanthi Karyala
- Department of Biotechnology, Faculty of Life and Allied Health Sciences, Ramaiah University of Applied Sciences, Bengaluru, 560054, India
| | - Suresh B Pakala
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Karakambadi Road, Mangalam, Tirupati, Andhra Pradesh, 517507, India.
| |
Collapse
|
11
|
Jacquier A, Roubille S, Lomonte P, Schaeffer L. Microrchidia CW-Type Zinc Finger 2, a Chromatin Modifier in a Spectrum of Peripheral Neuropathies. Front Cell Neurosci 2022; 16:896854. [PMID: 35722617 PMCID: PMC9203694 DOI: 10.3389/fncel.2022.896854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/11/2022] [Indexed: 12/12/2022] Open
Abstract
Microrchidia CW-type zinc finger 2 (MORC2) gene encodes a protein expressed in all tissues and enriched in the brain. MORC2 protein is composed of a catalytic ATPase domain, three coil-coiled domains allowing dimerization or protein complex interaction, a zinc-finger CW domain allowing DNA interaction, and a CHROMO-like (CHRromatin Organization Modifier) domain. Recently, de novo or dominantly inherited heterozygous mutations have been associated with a spectrum of disorders affecting the peripheral nervous system such as the Charcot-Marie-Tooth disease, spinal muscular atrophy-like phenotype disorder, or a neurodevelopmental syndrome associated with developmental delay, impaired growth, dysmorphic facies, and axonal neuropathy (DIGFAN). In this review, we detail the various mutations of MORC2 and their consequences on clinical manifestations. Possible genotype-phenotype correlations as well as intra and inter-family variability are discussed. MORC2 molecular functions such as transcriptional modulation, DNA damage repair, and lipid metabolism are then reviewed. We further discuss the impact of MORC2 mutations on the epigenetic landscape in the neuromuscular system and hypothesize probable pathophysiological mechanisms underlying the phenotypic variability observed.
Collapse
Affiliation(s)
- Arnaud Jacquier
- INMG-Pathophysiology and Genetics of Neuron and Muscle, CNRS UMR 5261, INSERM U1315, Université Claude Bernard Lyon 1, Faculté de Médecine Lyon Est, Lyon, France
- Hospices Civils de Lyon, Groupement Est, Bron, France
| | - Simon Roubille
- INMG-Pathophysiology and Genetics of Neuron and Muscle, CNRS UMR 5261, INSERM U1315, Université Claude Bernard Lyon 1, Faculté de Médecine Lyon Est, Lyon, France
| | - Patrick Lomonte
- INMG-Pathophysiology and Genetics of Neuron and Muscle, CNRS UMR 5261, INSERM U1315, Université Claude Bernard Lyon 1, Faculté de Médecine Lyon Est, Lyon, France
| | - Laurent Schaeffer
- INMG-Pathophysiology and Genetics of Neuron and Muscle, CNRS UMR 5261, INSERM U1315, Université Claude Bernard Lyon 1, Faculté de Médecine Lyon Est, Lyon, France
- Hospices Civils de Lyon, Groupement Est, Bron, France
| |
Collapse
|
12
|
Chutani N, Singh AK, Kadumuri RV, Pakala SB, Chavali S. Structural and Functional Attributes of Microrchidia Family of Chromatin Remodelers. J Mol Biol 2022; 434:167664. [PMID: 35659506 DOI: 10.1016/j.jmb.2022.167664] [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: 01/24/2022] [Revised: 05/10/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022]
Abstract
Chromatin remodelers affect the spatio-temporal dynamics of global gene-expression by structurally modulating and/or reorganizing the chromatin. Microrchidia (MORC) family is a relatively new addition to the four well studied families of chromatin remodeling proteins. In this review, we discuss the current understanding of the structural aspects of human MORCs as well as their epigenetic functions. From a molecular and systems-level perspective, we explore their participation in phase-separated structures, possible influence on various biological processes through protein-protein interactions, and potential extra-nuclear roles. We describe how dysregulation/dysfunction of MORCs can lead to various pathological conditions. We conclude by emphasizing the importance of undertaking integrated efforts to obtain a holistic understanding of the various biological roles of MORCs.
Collapse
Affiliation(s)
- Namita Chutani
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517 507, Andhra Pradesh, India. https://twitter.com/ChutaniNamita
| | - Anjali Kumari Singh
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517 507, Andhra Pradesh, India. https://twitter.com/anjali_k_s
| | - Rajashekar Varma Kadumuri
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517 507, Andhra Pradesh, India
| | - Suresh B Pakala
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517 507, Andhra Pradesh, India.
| | - Sreenivas Chavali
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517 507, Andhra Pradesh, India.
| |
Collapse
|
13
|
Anti-Colon Cancer Activity of Novel Peptides Isolated from In Vitro Digestion of Quinoa Protein in Caco-2 Cells. Foods 2022; 11:foods11020194. [PMID: 35053925 PMCID: PMC8774364 DOI: 10.3390/foods11020194] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 12/31/2021] [Accepted: 01/07/2022] [Indexed: 02/07/2023] Open
Abstract
Quinoa peptides are the bioactive components obtained from quinoa protein digestion, which have been proved to possess various biological activities. However, there are few studies on the anticancer activity of quinoa peptides, and the mechanism has not been clarified. In this study, the novel quinoa peptides were obtained from quinoa protein hydrolysate and identified by liquid chromatography-tandem mass spectrometry (LC-MS/MS). The anticancer activity of these peptides was predicted by PeptideRanker and evaluated using an antiproliferative assay in colon cancer Caco-2 cells. Combined with the result of histone deacetylase 1 (HDAC1) inhibitory activity assay, the highly anticancer activity peptides FHPFPR, NWFPLPR, and HYNPYFPG were screened and further investigated. Molecular docking was used to analyze the binding site between peptides and HDAC1, and results showed that three peptides were bound in the active pocket of HDAC1. Moreover, real-time quantitative polymerase chain reaction (RT-qPCR), and Western blot showed that the expression of HDAC1, NFκB, IL-6, IL-8, Bcl-2 was significantly decreased, whereas caspase3 expression showed a remarkable evaluation. In conclusion, quinoa peptides may have the potential to protect against cancer development by inhibiting HDAC1 activity and regulating the expression of the cancer-related genes, which indicates that these peptides could be explored as functional foods to alleviate colon cancer.
Collapse
|
14
|
MORC protein family-related signature within human disease and cancer. Cell Death Dis 2021; 12:1112. [PMID: 34839357 PMCID: PMC8627505 DOI: 10.1038/s41419-021-04393-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 10/06/2021] [Accepted: 11/03/2021] [Indexed: 01/03/2023]
Abstract
The microrchidia (MORC) family of proteins is a highly conserved nuclear protein superfamily, whose members contain common domain structures (GHKL-ATPase, CW-type zinc finger and coiled-coil domain) yet exhibit diverse biological functions. Despite the advancing research in previous decades, much of which focuses on their role as epigenetic regulators and in chromatin remodeling, relatively little is known about the role of MORCs in tumorigenesis and pathogenesis. MORCs were first identified as epigenetic regulators and chromatin remodelers in germ cell development. Currently, MORCs are regarded as disease genes that are involved in various human disorders and oncogenes in cancer progression and are expected to be the important biomarkers for diagnosis and treatment. A new paradigm of expanded MORC family function has raised questions regarding the regulation of MORCs and their biological role at the subcellular level. Here, we systematically review the progress of researching MORC members with respect to their domain architectures, diverse biological functions, and distribution characteristics and discuss the emerging roles of the aberrant expression or mutation of MORC family members in human disorders and cancer development. Furthermore, the illustration of related mechanisms of the MORC family has made MORCs promising targets for developing diagnostic tools and therapeutic treatments for human diseases, including cancers.
Collapse
|
15
|
MORC2 Interactome: Its Involvement in Metabolism and Cancer. Biophys Rev 2021; 13:507-514. [PMID: 34471435 DOI: 10.1007/s12551-021-00812-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/31/2021] [Indexed: 12/21/2022] Open
Abstract
Microrchidia 2 (MORC2) is an emerging chromatin modifier with a role in chromatin remodeling and epigenetic regulation. MORC2 is found to be upregulated in most cancers, playing a significant role in tumorigenesis and tumor metastasis. Recent studies have demonstrated that MORC2 is a scaffolding protein, which interacts with the proteins involved in DNA repair, chromatin remodeling, lipogenesis, and glucose metabolism. In this review, we discuss the domain architecture and cellular and subcellular localization of MORC2. Further, we highlight MORC2-specific interacting partners involved in metabolic reprogramming and other pathological functions such as cancer progression and metastasis.
Collapse
|
16
|
Regan JL, Schumacher D, Staudte S, Steffen A, Lesche R, Toedling J, Jourdan T, Haybaeck J, Mumberg D, Henderson D, Győrffy B, Regenbrecht CRA, Keilholz U, Schäfer R, Lange M. RNA sequencing of long-term label-retaining colon cancer stem cells identifies novel regulators of quiescence. iScience 2021; 24:102618. [PMID: 34142064 PMCID: PMC8185225 DOI: 10.1016/j.isci.2021.102618] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/23/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Recent data suggest that therapy-resistant quiescent cancer stem cells (qCSCs) are the source of relapse in colon cancer. Here, using colon cancer patient-derived organoids and xenografts, we identify rare long-term label-retaining qCSCs that can re-enter the cell cycle to generate new tumors. RNA sequencing analyses demonstrated that these cells display the molecular hallmarks of quiescent tissue stem cells, including expression of p53 signaling genes, and are enriched for transcripts common to damage-induced quiescent revival stem cells of the regenerating intestine. In addition, we identify negative regulators of cell cycle, downstream of p53, that we show are indicators of poor prognosis and may be targeted for qCSC abolition in both p53 wild-type and mutant tumors. These data support the temporal inhibition of downstream targets of p53 signaling, in combination with standard-of-care treatments, for the elimination of qCSCs and prevention of relapse in colon cancer. Colon tumors contain therapy-resistant quiescent cancer stem cells (qCSCs) qCSC gene expression mirrors that of quiescent stem cells of the regenerating gut qCSCs are enriched for p53 signaling genes qCSC elimination may be achieved by inhibiting downstream targets of p53 signaling
Collapse
Affiliation(s)
- Joseph L Regan
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany.,Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Dirk Schumacher
- Laboratory of Molecular Tumor Pathology, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany.,German Cancer Consortium (DKTK), DKFZ, 69120 Heidelberg, Germany
| | - Stephanie Staudte
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany.,German Cancer Consortium (DKTK), DKFZ, 69120 Heidelberg, Germany.,Department of Radiation Oncology and Radiotherapy, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Andreas Steffen
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany
| | - Ralf Lesche
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany.,Nuvisan ICB GmbH, 13353 Berlin, Germany
| | - Joern Toedling
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany.,Nuvisan ICB GmbH, 13353 Berlin, Germany
| | - Thibaud Jourdan
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany
| | - Johannes Haybaeck
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, A-6020 Innsbruck, Austria.,Diagnostic & Research Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, 8036 Graz, Austria
| | - Dominik Mumberg
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany
| | - David Henderson
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, 1094 Budapest, Hungary.,TTK Cancer Biomarker Research Group, Institute of Enzymology, 1117 Budapest, Hungary
| | - Christian R A Regenbrecht
- Laboratory of Molecular Tumor Pathology, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany.,CELLphenomics GmbH, 13125 Berlin, Germany.,Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Reinhold Schäfer
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany.,Laboratory of Molecular Tumor Pathology, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany.,German Cancer Consortium (DKTK), DKFZ, 69120 Heidelberg, Germany
| | - Martin Lange
- Bayer AG, Research & Development, Pharmaceuticals, 13342 Berlin, Germany.,Nuvisan ICB GmbH, 13353 Berlin, Germany
| |
Collapse
|
17
|
Guddeti RK, Thomas L, Kannan A, Karyala P, Pakala SB. The chromatin modifier MORC2 affects glucose metabolism by regulating the expression of lactate dehydrogenase A through a feed forward loop with c-Myc. FEBS Lett 2021; 595:1289-1302. [PMID: 33626175 DOI: 10.1002/1873-3468.14062] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 02/02/2021] [Accepted: 02/15/2021] [Indexed: 12/26/2022]
Abstract
Microrchidia family CW-type zinc finger 2 (MORC2) is a recently identified chromatin modifier with an emerging role in cancer metastasis. However, its role in glucose metabolism, a hallmark of malignancy, remains to be explored. We found that MORC2 is a glucose-inducible gene and a target of c-Myc. Our meta-analysis revealed that MORC2 expression is positively correlated with the expression of enzymes involved in glucose metabolism in breast cancer patients. Furthermore, overexpression of MORC2 in MCF-7 and BT-549 cells augmented the expression and activity of a key glucose metabolism enzyme, lactate dehydrogenase A (LDHA). Conversely, selective knockdown of MORC2 by siRNA markedly decreased LDHA expression and activity and in turn reduced cancer cell migration. Collectively, these findings provide evidence that MORC2, a glucose-inducible gene, modulates the migration of breast cancer cells through the MORC2-c-Myc-LDHA axis.
Collapse
Affiliation(s)
- Rohith Kumar Guddeti
- Biology Division, Indian Institute of Science Education and Research (IISER) Tirupati, India
| | - Liz Thomas
- Biology Division, Indian Institute of Science Education and Research (IISER) Tirupati, India
| | - Anbarasu Kannan
- Department of Protein Chemistry and Technology, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - Prashanthi Karyala
- Department of Biochemistry, Indian Academy Degree College Autonomous, Bengaluru, India
| | - Suresh B Pakala
- Biology Division, Indian Institute of Science Education and Research (IISER) Tirupati, India
| |
Collapse
|
18
|
Fukuda K, Shinkai Y. SETDB1-Mediated Silencing of Retroelements. Viruses 2020; 12:E596. [PMID: 32486217 PMCID: PMC7354471 DOI: 10.3390/v12060596] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/28/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022] Open
Abstract
SETDB1 (SET domain bifurcated histone lysine methyltransferase 1) is a protein lysine methyltransferase and methylates histone H3 at lysine 9 (H3K9). Among other H3K9 methyltransferases, SETDB1 and SETDB1-mediated H3K9 trimethylation (H3K9me3) play pivotal roles for silencing of endogenous and exogenous retroelements, thus contributing to genome stability against retroelement transposition. Furthermore, SETDB1 is highly upregulated in various tumor cells. In this article, we describe recent advances about how SETDB1 activity is regulated, how SETDB1 represses various types of retroelements such as L1 and class I, II, and III endogenous retroviruses (ERVs) in concert with other epigenetic factors such as KAP1 and the HUSH complex and how SETDB1-mediated H3K9 methylation can be maintained during replication.
Collapse
Affiliation(s)
- Kei Fukuda
- Cellular Memory Laboratory, RIKEN Cluster for Pioneering Research, RIKEN, Wako 351-0198, Japan
| | - Yoichi Shinkai
- Cellular Memory Laboratory, RIKEN Cluster for Pioneering Research, RIKEN, Wako 351-0198, Japan
| |
Collapse
|
19
|
Cheng Y, He C, Wang M, Ma X, Mo F, Yang S, Han J, Wei X. Targeting epigenetic regulators for cancer therapy: mechanisms and advances in clinical trials. Signal Transduct Target Ther 2019; 4:62. [PMID: 31871779 PMCID: PMC6915746 DOI: 10.1038/s41392-019-0095-0] [Citation(s) in RCA: 590] [Impact Index Per Article: 118.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 10/16/2019] [Accepted: 10/24/2019] [Indexed: 02/05/2023] Open
Abstract
Epigenetic alternations concern heritable yet reversible changes in histone or DNA modifications that regulate gene activity beyond the underlying sequence. Epigenetic dysregulation is often linked to human disease, notably cancer. With the development of various drugs targeting epigenetic regulators, epigenetic-targeted therapy has been applied in the treatment of hematological malignancies and has exhibited viable therapeutic potential for solid tumors in preclinical and clinical trials. In this review, we summarize the aberrant functions of enzymes in DNA methylation, histone acetylation and histone methylation during tumor progression and highlight the development of inhibitors of or drugs targeted at epigenetic enzymes.
Collapse
Affiliation(s)
- Yuan Cheng
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Cai He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Fei Mo
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Shengyong Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Junhong Han
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Al Bitar S, Gali-Muhtasib H. The Role of the Cyclin Dependent Kinase Inhibitor p21 cip1/waf1 in Targeting Cancer: Molecular Mechanisms and Novel Therapeutics. Cancers (Basel) 2019; 11:cancers11101475. [PMID: 31575057 PMCID: PMC6826572 DOI: 10.3390/cancers11101475] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/26/2019] [Accepted: 07/30/2019] [Indexed: 12/15/2022] Open
Abstract
p21cip1/waf1 mediates various biological activities by sensing and responding to multiple stimuli, via p53-dependent and independent pathways. p21 is known to act as a tumor suppressor mainly by inhibiting cell cycle progression and allowing DNA repair. Significant advances have been made in elucidating the potential role of p21 in promoting tumorigenesis. Here, we discuss the involvement of p21 in multiple signaling pathways, its dual role in cancer, and the importance of understanding its paradoxical functions for effectively designing therapeutic strategies that could selectively inhibit its oncogenic activities, override resistance to therapy and yet preserve its tumor suppressive functions.
Collapse
Affiliation(s)
- Samar Al Bitar
- Department of Biology, and Center for Drug Discovery, American University of Beirut, Beirut 1103, Lebanon.
| | - Hala Gali-Muhtasib
- Department of Biology, and Center for Drug Discovery, American University of Beirut, Beirut 1103, Lebanon.
| |
Collapse
|
21
|
Wu Y, Xing Y, Zou D. Study of the relationship between how ethanol affects learning and memory and the expression of p21 WAF1/CIP1 in the female mouse hippocampus. Neurosci Lett 2019; 708:134354. [PMID: 31254559 DOI: 10.1016/j.neulet.2019.134354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/24/2019] [Accepted: 06/25/2019] [Indexed: 01/27/2023]
Abstract
The purpose of this study was to investigate the effects of different concentrations of ethanol on learning and memory in female mice and the corresponding interaction with histone deacetylase 1(HDAC1), estrogen receptor α(ERα) and p21 WAF1/CIP1. Data from the Morris water maze test showed that mice in the 50% ethanol group might experience cognitive impairment, while mice in the 2% ethanol group might experience enhanced cognitive capabilities. The number of damaged neurons in the hippocampal CA1 area in the 50% ethanol group was higher than the numbers observed in other groups. The expression of HDAC1 and ERα proteins was lower in the 50% ethanol group than they were in the control group, while p21 WAF1/CIP1 expression was increased. The expression of these proteins in the 2% ethanol group was completely reversed when compared to the 50% ethanol group. p21 WAF1/CIP1 was involved in the cognitive change induced by ethanol. The f2 (-400 bp to -800 bp) and f7 (-2400 bp to -2800 bp) fragments in the p21 WAF1/CIP1 promoter region were functionally active regions that experienced binding relating to HDAC1 and ERα.
Collapse
Affiliation(s)
- Yi Wu
- Department of Pathophysiology, Shenyang Medical College, 146 Huang He North Road, Shenyang Liaoning 110034, PR China.
| | - Yang Xing
- Zhengzhou Yihe Hospital Affiliated of Henan University, Zhengzhou, Henan, 450002, PR China.
| | - Dan Zou
- Department of Pathophysiology, Shenyang Medical College, 146 Huang He North Road, Shenyang Liaoning 110034, PR China.
| |
Collapse
|
22
|
Zhang L, Bu L, Hu J, Xu Z, Ruan L, Fang Y, Wang P. HDAC1 knockdown inhibits invasion and induces apoptosis in non-small cell lung cancer cells. Biol Chem 2019. [PMID: 29537214 DOI: 10.1515/hsz-2017-0306] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Non-small cell lung cancer (NSCLC) is a common malignant tumor. Although the abnormal expression and potential clinical prognostic value of histone deacetylase 1 (HDAC1) were recently discovered in many kinds of cancer, the roles and molecular mechanisms of HDAC1 in NSCLC is still limited. The CCK-8 assay is used to evaluate the viability of NSCLC cells. Downregulation of HDAC1 by shRNA. The TUNEL assay was used to evaluate the role of HDAC1 in NSCLC apoptosis. To evaluate the role of HDAC1 in NSCLC cells migration, the Boyden chamber transwell assay and wound healing assay were used. To evaluate the cells invasion, the matrigel precoated Transwell assay was used. Enzyme-linked immunosorbent assays (ELISAs) were used to detect the level of vascular endothelial growth factor (VEGF) and IL-8 in NSCLC. To investigate the role of HDAC1 in angiogenesis, the tube formation assay was investigated. In this study, we showed that HDAC1 expression was elevated in NSCLC lines compared to that in normal liver cells LO2. Furthermore, downregulation of HDAC1 inhibited cell proliferation, prevented cell migration, decreased cell invasion, reduced tumor angiogenesis and induced cell apoptosis. In summary, HDAC1 may be regarded as a potential indicator for NSCLC patient treatment.
Collapse
Affiliation(s)
- Libin Zhang
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, No.157 Jinbi Road, Kunming City, 650032 Yunnan Province, China
| | - Liang Bu
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, No.157 Jinbi Road, Kunming City, 650032 Yunnan Province, China
| | - Jiang Hu
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, No.157 Jinbi Road, Kunming City, 650032 Yunnan Province, China
| | - Zheyuan Xu
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, No.157 Jinbi Road, Kunming City, 650032 Yunnan Province, China
| | - Libo Ruan
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, No.157 Jinbi Road, Kunming City, 650032 Yunnan Province, China
| | - Yan Fang
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, No.157 Jinbi Road, Kunming City, 650032 Yunnan Province, China
| | - Ping Wang
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, No.157 Jinbi Road, Kunming City, 650032 Yunnan Province, China
| |
Collapse
|
23
|
MORC2 regulates C/EBPα-mediated cell differentiation via sumoylation. Cell Death Differ 2019; 26:1905-1917. [PMID: 30644437 PMCID: PMC6748086 DOI: 10.1038/s41418-018-0259-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 11/25/2018] [Accepted: 11/28/2018] [Indexed: 12/29/2022] Open
Abstract
The expression and activity of CCAAT/enhancer-binding protein α (C/EBPα) are involved in sumoylation modification, which is critical to divert normal cells from differentiation to proliferation. However, the role and underlying mechanism of C/EBPα in cancer is poorly understood. Human MORC2 (microrchidia family CW-type zinc-finger 2), is a member of the MORC proteins family containing a CW-type zinc-finger domain. Here, we found that MORC2 interacted with TE-III domain of C/EBPα, and the overexpression of MORC2 promoted wild-type C/EBPα sumoylation and its subsequent degradation, which didn’t significantly observe in mutant C/EBPα-K161R. Furthermore, the overexpression of MORC2 inhibited C/EBPα-mediated C2C12 cell differentiation to maintain cell cycle progression. Moreover, the striking correlation between the decreased C/EBPα expression and the increased MORC2 expression was also observed in the poor differentiation status of gastric cancer tissues. Most notably, the high expression of MORC2 is correlated with an aggressive phenotype of clinical gastric cancer and shorter overall survival of patients. Taken together, our findings demonstrated that MORC2 expression regulated C/EBPα-mediated the axis of differentiation/proliferation via sumoylation modification, and affected its protein stability, causing cell proliferation and tumorigenesis.
Collapse
|
24
|
Liu J, Shao Y, He Y, Ning K, Cui X, Liu F, Wang Z, Li F. MORC2 promotes development of an aggressive colorectal cancer phenotype through inhibition of NDRG1. Cancer Sci 2018; 110:135-146. [PMID: 30407715 PMCID: PMC6317918 DOI: 10.1111/cas.13863] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 11/01/2018] [Accepted: 11/03/2018] [Indexed: 01/19/2023] Open
Abstract
MORC2 (microrchidia family CW‐type zinc finger 2) is a newly identified chromatin remodeling protein that functions in diverse biological processes including gene transcription. NDRG1 is a metastasis suppressor and a prognostic biomarker for colorectal cancer (CRC). However, the relationship between MORC2 and NDRG1 transcriptional regulation and the roles of MORC2 in CRC remain elusive. Here, we showed that MORC2 downregulated NDRG1 mRNA, protein levels, and promoter activity in CRC cells. We also found that MORC2 bound to the −446 to −213 bp region of the NDRG1 promoter. Mechanistically, histone deacetylase sirtuin 1 (SIRT1) was involved in NDRG1 transcriptional regulation. MORC2 was able to interact with SIRT1 and inhibit NDRG1 promoter activity cumulatively with SIRT1. MORC2 overexpression led to a decrease of H3Ac and H4Ac of the NDRG1 promoter. Importantly, we showed that NDRG1 was essential in MORC2‐mediated promotion of CRC cell migration and invasion in vitro, as well as lung metastasis of CRC cells in vivo. Moreover, MORC2 expression correlated negatively with NDRG1 expression in CRC patients. High expression of MORC2 was significantly associated with lymph node metastasis (P = 0.019) and poor pTNM stage (P = 0.02) and the expression of MORC2 correlated with poor prognosis in colon cancer patients. Our findings thus contribute to the knowledge of the regulatory mechanism of MORC2 in downregulating NDRG1, and suggest MORC2 as a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Jiao Liu
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, China
| | - Yangguang Shao
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, China
| | - Yuxin He
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, China
| | - Ke Ning
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, China
| | - Xi Cui
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, China
| | - Furong Liu
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, China
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, China
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC, and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, China
| |
Collapse
|
25
|
Ding QS, Zhang L, Wang BC, Zeng Z, Zou XQ, Cao PB, Zhou GM, Tang M, Wu L, Wu LL, Yu HG, Guo Y, Zhou FX. Aberrant high expression level of MORC2 is a common character in multiple cancers. Hum Pathol 2018; 76:58-67. [PMID: 29555576 DOI: 10.1016/j.humpath.2018.03.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 02/28/2018] [Accepted: 03/09/2018] [Indexed: 12/26/2022]
|
26
|
Fukuda K, Okuda A, Yusa K, Shinkai Y. A CRISPR knockout screen identifies SETDB1-target retroelement silencing factors in embryonic stem cells. Genome Res 2018; 28:846-858. [PMID: 29728365 PMCID: PMC5991520 DOI: 10.1101/gr.227280.117] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 04/26/2018] [Indexed: 12/16/2022]
Abstract
In mouse embryonic stem cells (mESCs), the expression of provirus and endogenous retroelements is epigenetically repressed. Although many cellular factors involved in retroelement silencing have been identified, the complete molecular mechanism remains elusive. In this study, we performed a genome-wide CRISPR screen to advance our understanding of retroelement silencing in mESCs. The Moloney murine leukemia virus (MLV)–based retroviral vector MSCV-GFP, which is repressed by the SETDB1/TRIM28 pathway in mESCs, was used as a reporter provirus, and we identified more than 80 genes involved in this process. In particular, ATF7IP and the BAF complex components are linked with the repression of most of the SETDB1 targets. We characterized two factors, MORC2A and RESF1, of which RESF1 is a novel molecule in retroelement silencing. Although both factors are recruited to repress provirus, their roles in repression are different. MORC2A appears to function dependent on repressive epigenetic modifications, while RESF1 regulates repressive epigenetic modifications associated with SETDB1. Our genome-wide CRISPR screen cataloged genes which function at different levels in silencing of SETDB1-target retroelements and provides a useful resource for further molecular studies.
Collapse
Affiliation(s)
- Kei Fukuda
- Cellular Memory Laboratory, Cluster for Pioneering Research, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Akihiko Okuda
- Division of Developmental Biology, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane Hidaka Saitama 350-1241, Japan
| | - Kosuke Yusa
- Wellcome Sanger Institute, Hinxton, Cambridge, CB10 1SA, United Kingdom
| | - Yoichi Shinkai
- Cellular Memory Laboratory, Cluster for Pioneering Research, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
27
|
Yin X, Wu Q, Monga J, Xie E, Wang H, Wang S, Zhang H, Wang ZY, Zhou T, Shi Y, Rogers J, Lin H, Min J, Wang F. HDAC1 Governs Iron Homeostasis Independent of Histone Deacetylation in Iron-Overload Murine Models. Antioxid Redox Signal 2018; 28:1224-1237. [PMID: 29113455 DOI: 10.1089/ars.2017.7161] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
AIMS Iron-overload disorders are common and could lead to significant morbidity and mortality worldwide. Due to limited treatment options, there is a great need to develop novel strategies to remove the excess body iron. To discover potential epigenetic modulator in hepcidin upregulation and subsequently decreasing iron burden, we performed an epigenetic screen. The in vivo effects of the identified compounds were further tested in iron-overload mouse models, including Hfe-/-, Hjv-/-, and hepatocyte-specific Smad4 knockout (Smad4fl/fl;Alb-Cre+) mice. RESULTS Entinostat (MS-275), the clinical used histone deacetylase 1 (HDAC1) inhibitor, was identified the most potent hepcidin agonist. Consistently, Hdac1-deficient mice also presented higher hepcidin levels than wild-type controls. Notably, the long-term treatment with entinostat in Hfe-/- mice significantly alleviated iron overload through upregulating hepcidin transcription. In contrast, entinostat showed no effect on hepcidin expression and iron levels in Smad4fl/fl;Alb-Cre+ mice. Further mechanistic studies revealed that HDAC1 suppressed expression of hepcidin through interacting with SMAD4 rather than deacetylation of SMAD4 or histone-H3 on the hepcidin promoter. INNOVATION The findings uncovered HDAC1 as a novel hepcidin suppressor through complexing with SMAD4 but not deacetylation of either histone 3 or SMAD4. In addition, our study suggested a novel implication of entinostat in treating iron-overload disorders. CONCLUSIONS Based on our results, we conclude that entinostat strongly activated hepcidin in vivo and in vitro. HDAC1 could serve as a novel hepcidin suppressor by binding to SMAD4, effect of which is independent of BMP/SMAD1/5/8 signaling. Antioxid. Redox Signal. 28, 1224-1237.
Collapse
Affiliation(s)
- Xiangju Yin
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China .,2 Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University , Zhengzhou, China .,3 Institute of Resources and Environment, Henan Polytechnic University , Jiaozuo, China
| | - Qian Wu
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China .,2 Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University , Zhengzhou, China
| | - Jitender Monga
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Enjun Xie
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Hao Wang
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China .,2 Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University , Zhengzhou, China
| | - Shufen Wang
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Huizhen Zhang
- 2 Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University , Zhengzhou, China
| | - Zhan-You Wang
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Tianhua Zhou
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Yujun Shi
- 4 Laboratory of Pathology, West China Hospital, Sichuan University , Chengdu, China
| | - Jack Rogers
- 5 Neurochemistry Laboratory, Departments of Psychiatry and Pediatrics, Massachusetts General Hospital and Harvard Medical School , Charlestown, Massachusetts
| | - Hening Lin
- 6 Department of Chemistry and Chemical Biology, Howard Hughes Medical Institute, Cornell University , Ithaca, New York
| | - Junxia Min
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China
| | - Fudi Wang
- 1 College of Life and Health Sciences, Northeastern University ; The First Affiliated Hospital, Institute of Translational Medicine, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Public Health, School of Medicine, Zhejiang University , Hangzhou, China .,2 Department of Nutrition, Precision Nutrition Innovation Center, School of Public Health, Zhengzhou University , Zhengzhou, China
| |
Collapse
|
28
|
Pan Z, Ding Q, Guo Q, Guo Y, Wu L, Wu L, Tang M, Yu H, Zhou F. MORC2, a novel oncogene, is upregulated in liver cancer and contributes to proliferation, metastasis and chemoresistance. Int J Oncol 2018; 53:59-72. [PMID: 29620211 PMCID: PMC5958890 DOI: 10.3892/ijo.2018.4333] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/22/2018] [Indexed: 12/20/2022] Open
Abstract
Microrchidia 2 (MORC2) is important in DNA damage repair and lipogenesis, however, the clinical and functional role of MORC2 in liver cancer remains to be fully elucidated. The aim the present study was to clarify the role of MORC2 in liver cancer. Expression profile analysis, immunohistochemical staining, reverse transcription-quantitative polymerase chain reaction analysis and western blot analysis were performed to evaluate the levels of MORC2 in liver cancer patient specimens and cell lines; subsequently the expression of MORC2 was suppressed or increased in liver cancer cells and the effects of MORC2 on the cancerous transformation of liver cancer cells were examined in vitro and in vivo. MORC2 was upregulated in liver cancer tissues, and the upregulation was associated with certain clinicopathologic features of patients with liver cancer. MORC2 knockdown caused marked inhibition of liver cancer cell proliferation and clonogenicity, whereas the overexpression of MORC2 substantially promoted liver cancer cell proliferation. In addition, the knockdown of MORC2 inhibited the migratory and invasive ability of liver cancer cells, whereas increased migration and invasion rates were observed in cells with ectopic expression of MORC2. In a model of nude mice, the overexpression of MORC2 promoted tumorigenicity and markedly enhanced pulmonary metastasis of liver cancer. Furthermore, MORC2 regulated apoptosis and its expression level had an effect on the sensitivity of liver cancer cells to doxorubicin, 5-fluorouracil and cisplatin. Mechanically, MORC2 modulated the mitochondrial apoptotic pathway, possibly in a p53-dependent manner, and its dysregulation also resulted in the abnormal activation of the Hippo pathway. For the first time, to the best of our knowledge, the present study confirmed that MORC2 was a novel oncogene in liver cancer. These results provide useful insight into the mechanism underlying the tumorigenesis and progression of liver cancer, and offers clues into potential novel liver cancer therapies.
Collapse
Affiliation(s)
- Zhihong Pan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qianshan Ding
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qian Guo
- Hepatic Disease Institute, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430061, P.R. China
| | - Yong Guo
- College of Biotechnology, Guilin Medical University, Guilin, Guanxi 541004, P.R. China
| | - Lianlian Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Lu Wu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Meng Tang
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Clinical Cancer Study Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Honggang Yu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Fuxiang Zhou
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Clinical Cancer Study Center, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
29
|
Wang T, Qin ZY, Wen LZ, Guo Y, Liu Q, Lei ZJ, Pan W, Liu KJ, Wang XW, Lai SJ, Sun WJ, Wei YL, Liu L, Guo L, Chen YQ, Wang J, Xiao HL, Bian XW, Chen DF, Wang B. Epigenetic restriction of Hippo signaling by MORC2 underlies stemness of hepatocellular carcinoma cells. Cell Death Differ 2018; 25:2086-2100. [PMID: 29555977 DOI: 10.1038/s41418-018-0095-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/14/2018] [Accepted: 02/22/2018] [Indexed: 12/12/2022] Open
Abstract
The evolutionarily conserved Hippo signaling pathway is a key regulator of stem cell self-renewal, differentiation, and organ size. While alterations in Hippo signaling are causally linked to uncontrolled cell growth and a broad range of malignancies, genetic mutations in the Hippo pathway are uncommon and it is unclear how the tumor suppressor function of the Hippo pathway is disrupted in human cancers. Here, we report a novel epigenetic mechanism of Hippo inactivation in the context of hepatocellular carcinoma (HCC). We identify a member of the microrchidia (MORC) protein family, MORC2, as an inhibitor of the Hippo pathway by controlling upstream Hippo regulators, neurofibromatosis 2 (NF2) and kidney and brain protein (KIBRA). Mechanistically, MORC2 forms a complex with DNA methyltransferase 3A (DNMT3A) at the promoters of NF2 and KIBRA, leading to their DNA hyper-methylation and transcriptional repression. As a result, NF2 and KIBRA are crucial targets of MORC2 to regulate confluence-induced activation of Hippo signaling and contact inhibition of cell growth under both physiological and pathological conditions. The MORC2-NF2/KIBRA axis is critical for maintaining self-renewal, sorafenib resistance, and oncogenicity of HCC cells in vitro and in nude mice. Furthermore, MORC2 expression is elevated in HCC tissues, associated with stem-like properties of cancer cells, and disease progression in patients. Collectively, MORC2 promotes cancer stemness and tumorigenesis by facilitating DNA methylation-dependent silencing of Hippo signaling and could be a potential molecular target for cancer therapeutics.
Collapse
Affiliation(s)
- Tao Wang
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Zhong-Yi Qin
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Liang-Zhi Wen
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Yan Guo
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Qin Liu
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Zeng-Jie Lei
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, Jiangsu Province, China
| | - Wei Pan
- Department of Medical Genetics, Second Military Medical University (Navy Medical University), 200433, Shanghai, China
| | - Kai-Jun Liu
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Xing-Wei Wang
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Shu-Jie Lai
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Wen-Jing Sun
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Yan-Ling Wei
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Lei Liu
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Ling Guo
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Yu-Qin Chen
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Jun Wang
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Hua-Liang Xiao
- Department of Pathology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Dong-Feng Chen
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China.
| | - Bin Wang
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China.
| |
Collapse
|
30
|
Shi B, Xue J, Zhou J, Kasowitz SD, Zhang Y, Liang G, Guan Y, Shi Q, Liu M, Sha J, Huang X, Wang PJ. MORC2B is essential for meiotic progression and fertility. PLoS Genet 2018; 14:e1007175. [PMID: 29329290 PMCID: PMC5785033 DOI: 10.1371/journal.pgen.1007175] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 01/25/2018] [Accepted: 12/29/2017] [Indexed: 12/11/2022] Open
Abstract
The microrchidia (MORC) family proteins are chromatin-remodelling factors and function in diverse biological processes such as DNA damage response and transposon silencing. Here, we report that mouse Morc2b encodes a functional germ cell-specific member of the MORC protein family. Morc2b arose specifically in the rodent lineage through retrotransposition of Morc2a during evolution. Inactivation of Morc2b leads to meiotic arrest and sterility in both sexes. Morc2b-deficient spermatocytes and oocytes exhibit failures in chromosomal synapsis, blockades in meiotic recombination, and increased apoptosis. Loss of MORC2B causes mis-regulated expression of meiosis-specific genes. Furthermore, we find that MORC2B interacts with MORC2A, its sequence paralogue. Our results demonstrate that Morc2b, a relatively recent gene, has evolved an essential role in meiosis and fertility. In sexually reproducing organisms, meiosis, a process unique to germ cells, produces haploid gametes. Abnormalities in meiosis can lead to infertility, loss of pregnancy, or genetic diseases such as Down syndrome. The meiotic processes are tightly regulated by a large number of genes including many meiosis-specific ones. The majority of meiosis-specific factors are conserved, however, species-specific factors have evolved. Here we report functional studies of a rodent lineage–specific gene named Morc2b. Morc2b belongs to a family of chromatin-remodelling factors. Morc2b is specifically expressed in germ cells. Disruption of Morc2b causes meiotic arrest and infertility in both sexes. Notably, MORC2B regulates the expression of a number of meiosis-specific genes. Interestingly, MORC2B interacts with its sequence homologue MORC2A. These functional studies have uncovered a new protein complex in the regulation of key meiotic processes and suggested the presence of continued selection pressure for evolution of new meiosis-specific factors.
Collapse
Affiliation(s)
- Baolu Shi
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jiangyang Xue
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Jian Zhou
- Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Seth D. Kasowitz
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
| | - Yuanwei Zhang
- USTC-SJH Joint Center for Human Reproduction and Genetics, School of Life Sciences, University of Science and Technology of China, Hefei,Anhui, China
| | - Guanxiang Liang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Yongjuan Guan
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
| | - Qinghua Shi
- USTC-SJH Joint Center for Human Reproduction and Genetics, School of Life Sciences, University of Science and Technology of China, Hefei,Anhui, China
| | - Mingxi Liu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Jiahao Sha
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Xiaoyan Huang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
- * E-mail: (XH); (PJW)
| | - P. Jeremy Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail: (XH); (PJW)
| |
Collapse
|
31
|
HDAC1 promoted migration and invasion binding with TCF12 by promoting EMT progress in gallbladder cancer. Oncotarget 2017; 7:32754-64. [PMID: 27092878 PMCID: PMC5078048 DOI: 10.18632/oncotarget.8740] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/28/2016] [Indexed: 12/29/2022] Open
Abstract
The identification of prognostic markers for gallbladder cancer is needed for clinical practice. Histone deacetylases (HDACs) play an important role in tumor development and progression by modifying histone and non-histone proteins. However, the expression of HDAC1 in patients with gallbladder cancer is still unknown. Here, we reported that HDAC1 expression was elevated in cancerous tissue and correlated with lymph node metastasis and poorer overall survival in patients with GBC. Knockdown of HDAC1 using lentivirus delivery of HDAC1-specific shRNA abrogated the migration and invasion of GBC cells in vitro. TCF-12, as the HDAC1 binding protein, has also correlates with poor prognosis in GBC patients. And there is a positive correlation between HDAC1 and TCF-12 which leading the high invasion and migration ability of GBC cells. Taken together, our data suggested that HDAC1 and TCF-12 are a potential prognostic maker and may be a molecular target for inhibiting invasion and metastasis in GBC.
Collapse
|
32
|
Chromatin remodeling protein MORC2 promotes breast cancer invasion and metastasis through a PRD domain-mediated interaction with CTNND1. Oncotarget 2017; 8:97941-97954. [PMID: 29228664 PMCID: PMC5716704 DOI: 10.18632/oncotarget.18556] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 06/05/2017] [Indexed: 11/25/2022] Open
Abstract
MORC family CW-type zinc finger 2 (MORC2) is a newly identified chromatin remodeling protein with emerging roles in the regulation of DNA damage response and gene transcription, but its mechanistic role in breast cancer development and progression remains unexplored. Here, we show that MORC2 promoted breast cancer invasion and metastasis and these effects depended on a proline-rich domain (PRD) within its carboxy-terminal region spanning residues 601–734. Induced expression of wild-type MORC2 did not significantly affect cell proliferation and cell-cycle progression, but promoted breast cancer cell migration and invasion in vitro and metastatic lung colonization in vivo. The PRD domain was dispensable for the protein stability and subcellular localization of MORC2, but depletion of the PRD domain substantially suppressed MORC2-mediated migration, invasion, and metastasis. Proteomic and biochemical analyses further demonstrated that wild-type MORC2, but not PRD deletion mutant, interacted with catenin delta 1 (CTNND1), a cadherin-associated protein that participates in tumor invasion and metastasis. Moreover, knockdown of endogenous CTNND1 by short hairpin RNAs suppressed the migratory and invasive potential of MORC2-expressing cells. Taken together, these results suggest that MORC2 promotes breast cancer invasion and metastasis through its PRD domain-mediated interaction with CTNND1.
Collapse
|
33
|
Koch A, Kang HG, Steinbrenner J, Dempsey DA, Klessig DF, Kogel KH. MORC Proteins: Novel Players in Plant and Animal Health. FRONTIERS IN PLANT SCIENCE 2017; 8:1720. [PMID: 29093720 PMCID: PMC5651269 DOI: 10.3389/fpls.2017.01720] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 09/20/2017] [Indexed: 05/02/2023]
Abstract
Microrchidia (MORC) proteins comprise a family of proteins that have been identified in prokaryotes and eukaryotes. They are defined by two hallmark domains: a GHKL-type ATPase and an S5 fold. MORC proteins in plants were first discovered via a genetic screen for Arabidopsis mutants compromised for resistance to a viral pathogen. Subsequent studies expanded their role in plant immunity and revealed their involvement in gene silencing and transposable element repression. Emerging data suggest that MORC proteins also participate in pathogen-induced chromatin remodeling and epigenetic gene regulation. In addition, biochemical analyses recently demonstrated that plant MORCs have topoisomerase II (topo II)-like DNA modifying activities that may be important for their function. Interestingly, animal MORC proteins exhibit many parallels with their plant counterparts, as they have been implicated in disease development and gene silencing. In addition, human MORCs, like plant MORCs, bind salicylic acid and this inhibits some of their topo II-like activities. In this review, we will focus primarily on plant MORCs, although relevant comparisons with animal MORCs will be provided.
Collapse
Affiliation(s)
- Aline Koch
- Centre for BioSystems, Land Use and Nutrition, Institute for Phytopathology, Justus Liebig University Giessen, Giessen, Germany
| | - Hong-Gu Kang
- Department of Biology, Texas State University, San Marcos, TX, United States
| | - Jens Steinbrenner
- Centre for BioSystems, Land Use and Nutrition, Institute for Phytopathology, Justus Liebig University Giessen, Giessen, Germany
| | | | - Daniel F. Klessig
- Boyce Thompson Institute for Plant Research, Ithaca, NY, United States
- *Correspondence: Daniel F. Klessig
| | - Karl-Heinz Kogel
- Centre for BioSystems, Land Use and Nutrition, Institute for Phytopathology, Justus Liebig University Giessen, Giessen, Germany
- Karl-Heinz Kogel
| |
Collapse
|
34
|
Hong G, Qiu H, Wang C, Jadhav G, Wang H, Tickner J, He W, Xu J. The Emerging Role of MORC Family Proteins in Cancer Development and Bone Homeostasis. J Cell Physiol 2016; 232:928-934. [PMID: 27791268 DOI: 10.1002/jcp.25665] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 10/26/2016] [Indexed: 01/13/2023]
Abstract
Microrchidia (MORC or MORC family CW-type zinc finger protein), a highly conserved nuclear protein superfamily, is an interesting new player in signaling-dependent chromatin remodeling and epigenetic regulation. MORC family proteins consist of MORC1, MORC2, MORC3, and MORC4 which display common structural determinants such as CW-type zinc finger and coiled-coil domains. They also exhibit unique structural motifs and tissue-specific expression profiles. MORC1 was first discovered as a key regulator for male meiosis and spermatogenesis. Accumulating biochemical and functional analyses unveil MORC proteins as key regulators for cancer development. More recently, using an ENU mutagenesis mouse model, MORC3 was found to play a role in regulating bone and calcium homeostasis. Here we discuss recent research progress on the emerging role of MORC proteins in cancer development and bone metabolism. Unravelling the cellular and molecular mechanisms by which MORC proteins carry out their functions in a tissue specific manner are important subjects for future investigation. J. Cell. Physiol. 232: 928-934, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Guoju Hong
- The National Key Discipline and the Orthopedic Laboratory, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,School of Pathology Laboratory Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Heng Qiu
- School of Pathology Laboratory Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Chao Wang
- School of Pathology Laboratory Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Gaurav Jadhav
- School of Pathology Laboratory Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Haibin Wang
- The National Key Discipline and the Orthopedic Laboratory, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Jennifer Tickner
- School of Pathology Laboratory Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Wei He
- The National Key Discipline and the Orthopedic Laboratory, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China
| | - Jiake Xu
- The National Key Discipline and the Orthopedic Laboratory, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People's Republic of China.,School of Pathology Laboratory Medicine, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
35
|
Hyun YS, Hong YB, Choi BO, Chung KW. Clinico-genetics in Korean Charcot-Marie-Tooth disease type 2Z with MORC2 mutations. Brain 2016; 139:e40. [PMID: 27105897 DOI: 10.1093/brain/aww082] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Young Se Hyun
- Department of Biological Sciences, Kongju National University, Gongju, Korea
| | - Young Bin Hong
- Stem Cell and Regenerative Medicine Centre and Neuroscience Centre, Samsung Medical Centre, Seoul, Korea
| | - Byung-Ok Choi
- Stem Cell and Regenerative Medicine Centre and Neuroscience Centre, Samsung Medical Centre, Seoul, Korea Department of Neurology, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, Korea Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science and Tech, Sungkyunkwan University, Seoul, Korea
| | - Ki Wha Chung
- Department of Biological Sciences, Kongju National University, Gongju, Korea
| |
Collapse
|
36
|
Ye X, Zhang Y, Wang X, Li Y, Gao Y. Tumor-suppressive functions of long-chain acyl-CoA synthetase 4 in gastric cancer. IUBMB Life 2016; 68:320-7. [PMID: 26949059 DOI: 10.1002/iub.1486] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/01/2016] [Indexed: 12/19/2022]
Abstract
Long chain acyl CoA synthetase 4 (ACSL4) is a key enzyme in fatty acid metabolism with marked preference for arachidonic acid (AA). Recent reports have implicated its crucial roles in tumorigenesis. However in gastric cancer (GC), the expression and function of ACSL4 remain unclear. In the present study, we identified ACSL4 as a potential tumor suppressor in GC. The ACSL4 expression in GC samples was evaluated by real-time PCR and immunohistochemistry. The results indicated that the mRNA and protein levels of ACSL4 were frequently downregulated in cancer tissues compared with the adjacent non-cancerous mucosa control tissues. Cell-based functional assays exhibited that ectopic expression of ACSL4 inhibits cell growth, colony formation and cell migration, whereas ACSL4 knockdown enhanced these effects. In a nude mice model, ACSL4 knockdown also promoted subcutaneous xenografts' growth in vivo. Moreover, western blot analysis revealed that ACSL4 expression had a significant effect on FAK and P21 protein level. These findings suggest that ACSL4 plays a tumor-suppressive role and could be a potential therapeutic target in GC.
Collapse
Affiliation(s)
- Xiaojuan Ye
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yi Zhang
- Department of Oncology, East Hospital, Dalian Medical University, Shanghai, 200120, China
| | - Xiao Wang
- Department of Oncology, East Hospital, Dalian Medical University, Shanghai, 200120, China
| | - Yandong Li
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yong Gao
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| |
Collapse
|
37
|
Albulym OM, Kennerson ML, Harms MB, Drew AP, Siddell AH, Auer-Grumbach M, Pestronk A, Connolly A, Baloh RH, Zuchner S, Reddel SW, Nicholson GA. MORC2 mutations cause axonal Charcot-Marie-Tooth disease with pyramidal signs. Ann Neurol 2016; 79:419-27. [PMID: 26659848 DOI: 10.1002/ana.24575] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/02/2015] [Accepted: 12/04/2015] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To use linkage analysis and whole exome sequencing to identify the genetic mutation in a multigenerational Australian family with Charcot-Marie-Tooth disease type 2 (CMT2) and pyramidal signs. METHODS Genome-wide linkage analysis was performed to map the locus. Whole exome sequencing was undertaken on selected individuals (3 affected, 1 normal), and segregation analysis and mutation screening were carried out using high-resolution melt analysis. The GEM.app database was queried to identify additional families with mutations. RESULTS Significant linkage (2-point LOD score ≥ +3) and haplotype analysis mapped a new locus for CMT2 and pyramidal signs to a 6.6Mb interval on chromosome 22q12.1-q12.3. Whole exome sequencing identified a novel mutation (p.R252W) in the microrchidia CW-type zinc finger 2 (MORC2) gene mapping within the linkage region. The mutation fully segregated with the disease phenotype in the family. Screening additional families and querying unsolved CMT2 exomes, we identified the p.R252W mutation in 2 unrelated early onset CMT2 families and a second mutation p.E236G in 2 unrelated CMT2 families. Both the mutations occurred at highly conserved amino acid residues and were absent in the normal population. INTERPRETATION We have identified a new locus in which MORC2 mutations are the likely pathogenic cause of CMT2 and pyramidal signs in these families. MORC2 encodes the human CW-type zinc finger 2 protein, which is a chromatin modifier involved in the regulation of DNA repair as well as gene transcription.
Collapse
Affiliation(s)
- Obaid M Albulym
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Marina L Kennerson
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Molecular Medicine Laboratory, Concord Hospital, Concord, NSW, Australia
| | - Matthew B Harms
- Department of Neurology, Washington University School of Medicine, St Louis, MO
| | - Alexander P Drew
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Anna H Siddell
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | | | - Alan Pestronk
- Department of Neurology, Washington University School of Medicine, St Louis, MO
| | - Anne Connolly
- Department of Neurology, Washington University School of Medicine, St Louis, MO
| | - Robert H Baloh
- Department of Neurology, Cedars Sinai Medical Center, Los Angeles, CA
| | - Stephan Zuchner
- Dr John T. MacDonald Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL
| | - Stephen W Reddel
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Molecular Medicine Laboratory, Concord Hospital, Concord, NSW, Australia
| | - Garth A Nicholson
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Molecular Medicine Laboratory, Concord Hospital, Concord, NSW, Australia
| |
Collapse
|
38
|
Tong Y, Li Y, Gu H, Wang C, Liu F, Shao Y, Li J, Cao L, Li F. Microchidia protein 2, MORC2, downregulates the cytoskeleton adapter protein, ArgBP2, via histone methylation in gastric cancer cells. Biochem Biophys Res Commun 2015; 467:821-7. [PMID: 26476214 DOI: 10.1016/j.bbrc.2015.10.059] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 10/10/2015] [Indexed: 11/18/2022]
Abstract
ArgBP2 is an adapter protein that plays an important role in actin-dependent processes such as cell adhesion and migration. However, its function and regulation mechanisms in gastric cancer have not yet been investigated. Here, we showed the low expression of ArgBP2 mRNA level in gastric tumor samples and its repressive function in the proliferation, migration, and invasion of gastric cancer cells. Then, we cloned and identified ArgBP2 promoter and verified that MORC2 bound to the promoter. Moreover, we demonstrated that MORC2 enhanced the recruitment of EZH2, which promoted the tri-methylation of H3K27, leading to the transcriptional repression of ArgBP2. Our results might thus contribute to understanding the molecular mechanisms of ArgBP2 regulation and suggesting ArgBP2 as a potential therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Yuxin Tong
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Yan Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Hui Gu
- Department of Key Laboratory of Health Ministry for Congenital Malformation Shengjing Hospital, China Medical University, Shenyang, Liaoning 110004, China
| | - Chunyu Wang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Funan Liu
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yangguang Shao
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Jiabin Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Liu Cao
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|