1
|
Wang L, Wang P, Chen X, Yang H, Song S, Song Z, Jia L, Chen H, Bao X, Guo N, Huan X, Xi Y, Shen Y, Yang X, Su Y, Sun Y, Gao Y, Chen Y, Ding J, Lang J, Miao Z, Zhang A, He J. Thioparib inhibits homologous recombination repair, activates the type I IFN response, and overcomes olaparib resistance. EMBO Mol Med 2023; 15:e16235. [PMID: 36652375 PMCID: PMC9994488 DOI: 10.15252/emmm.202216235] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 01/19/2023] Open
Abstract
Poly-ADP-ribose polymerase (PARP) inhibitors (PARPi) have shown great promise for treating BRCA-deficient tumors. However, over 40% of BRCA-deficient patients fail to respond to PARPi. Here, we report that thioparib, a next-generation PARPi with high affinity against multiple PARPs, including PARP1, PARP2, and PARP7, displays high antitumor activities against PARPi-sensitive and -resistant cells with homologous recombination (HR) deficiency both in vitro and in vivo. Thioparib treatment elicited PARP1-dependent DNA damage and replication stress, causing S-phase arrest and apoptosis. Conversely, thioparib strongly inhibited HR-mediated DNA repair while increasing RAD51 foci formation. Notably, the on-target inhibition of PARP7 by thioparib-activated STING/TBK1-dependent phosphorylation of STAT1, triggered a strong induction of type I interferons (IFNs), and resulted in tumor growth retardation in an immunocompetent mouse model. However, the inhibitory effect of thioparib on tumor growth was more pronounced in PARP1 knockout mice, suggesting that a specific PARP7 inhibitor, rather than a pan inhibitor such as thioparib, would be more relevant for clinical applications. Finally, genome-scale CRISPR screening identified PARP1 and MCRS1 as genes capable of modulating thioparib sensitivity. Taken together, thioparib, a next-generation PARPi acting on both DNA damage response and antitumor immunity, serves as a therapeutic potential for treating hyperactive HR tumors, including those resistant to earlier-generation PARPi.
Collapse
Affiliation(s)
- Li‐Min Wang
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Pingyuan Wang
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- Pharm‐X Center, School of PharmacyShanghai Jiao Tong UniversityShanghaiChina
- Institute of Evolution and Marine BiodiversityOcean University of ChinaQingdaoChina
| | - Xiao‐Min Chen
- University of Chinese Academy of SciencesBeijingChina
- The CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Hui Yang
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Shan‐Shan Song
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Zilan Song
- Pharm‐X Center, School of PharmacyShanghai Jiao Tong UniversityShanghaiChina
| | - Li Jia
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Hua‐Dong Chen
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xu‐Bin Bao
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Ne Guo
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xia‐Juan Huan
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yong Xi
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yan‐Yan Shen
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xin‐Ying Yang
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yi Su
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yi‐Ming Sun
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Ying‐Lei Gao
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yi Chen
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jian Ding
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jing‐Yu Lang
- University of Chinese Academy of SciencesBeijingChina
- The CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and HealthUniversity of Chinese Academy of Sciences, Chinese Academy of SciencesShanghaiChina
| | - Ze‐Hong Miao
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Ao Zhang
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
- Pharm‐X Center, School of PharmacyShanghai Jiao Tong UniversityShanghaiChina
| | - Jin‐Xue He
- State Key Laboratory of Drug Research, Cancer Research Center, Shanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
2
|
Huang CJ, Lyu X, Kang J. The molecular characteristics and functional roles of microspherule protein 1 (MCRS1) in gene expression, cell proliferation, and organismic development. Cell Cycle 2023; 22:619-632. [PMID: 36384428 PMCID: PMC9980701 DOI: 10.1080/15384101.2022.2145816] [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: 06/01/2022] [Revised: 09/21/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
Abstract
Accurate spatial and temporal regulation of cell cycle progression is essential for cell proliferation and organismic development. This review demonstrates the role of microspherule protein 58kD, commonly known as MCRS1, as a key cell cycle regulator of higher eukaryotic organisms. We discuss the isoforms and functional domains of MCRS1 as well as their subcellular localization at specific stages of the cell cycle. These molecular characteristics reveal MCRS1's dynamic regulatory role in gene expression, genome stability, cell proliferation, and organismic development. Furthermore, we discuss the molecular details of its seemingly opposite, tumor-suppressive or tumor-promoting, role in different types of cancer.
Collapse
Affiliation(s)
| | - Xiaoai Lyu
- Arts and Science, New York University Shanghai, Shanghai, China
- Graduate School of Arts and Science, New York University, New York, USA
| | - Jungseog Kang
- Arts and Science, New York University Shanghai, Shanghai, China
- NYU-ECNU Center for Computational Chemistry, New York University Shanghai, Shanghai, China
| |
Collapse
|
3
|
Liu B, Jing Z, Zhang X, Chen Y, Mao S, Kaundal R, Zou Y, Wei G, Zang Y, Wang X, Lin W, Di M, Sun Y, Chen Q, Li Y, Xia J, Sun J, Lin CP, Huang X, Chi T. Large-scale multiplexed mosaic CRISPR perturbation in the whole organism. Cell 2022; 185:3008-3024.e16. [PMID: 35870449 DOI: 10.1016/j.cell.2022.06.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/23/2022] [Accepted: 06/20/2022] [Indexed: 12/13/2022]
Abstract
Here, we report inducible mosaic animal for perturbation (iMAP), a transgenic platform enabling in situ CRISPR targeting of at least 100 genes in parallel throughout the mouse body. iMAP combines Cre-loxP and CRISPR-Cas9 technologies and utilizes a germline-transmitted transgene carrying a large array of individually floxed, tandemly linked gRNA-coding units. Cre-mediated recombination triggers expression of all the gRNAs in the array but only one of them per cell, converting the mice to mosaic organisms suitable for phenotypic characterization and also for high-throughput derivation of conventional single-gene perturbation lines via breeding. Using gRNA representation as a readout, we mapped a miniature Perturb-Atlas cataloging the perturbations of 90 genes across 39 tissues, which yields rich insights into context-dependent gene functions and provides a glimpse of the potential of iMAP in genome decoding.
Collapse
Affiliation(s)
- Bo Liu
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zhengyu Jing
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaoming Zhang
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yuxin Chen
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Shaoshuai Mao
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ravinder Kaundal
- Department of Immunobiology, Yale University Medical School, New Haven, CT 06520, USA
| | - Yan Zou
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ge Wei
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ying Zang
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xinxin Wang
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wenyang Lin
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Minghui Di
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yiwen Sun
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qin Chen
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yongqin Li
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jing Xia
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jianlong Sun
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Chao-Po Lin
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xingxu Huang
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Tian Chi
- Gene Editing Center, School of Life Sciences and Technology, ShanghaiTech University, Shanghai 201210, China; Department of Immunobiology, Yale University Medical School, New Haven, CT 06520, USA.
| |
Collapse
|
4
|
Ermis M, Antmen E, Kuren O, Demirci U, Hasirci V. A Cell Culture Chip with Transparent, Micropillar-Decorated Bottom for Live Cell Imaging and Screening of Breast Cancer Cells. MICROMACHINES 2022; 13:mi13010093. [PMID: 35056257 PMCID: PMC8779566 DOI: 10.3390/mi13010093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 12/31/2021] [Accepted: 01/04/2022] [Indexed: 12/30/2022]
Abstract
In the recent years, microfabrication technologies have been widely used in cell biology, tissue engineering, and regenerative medicine studies. Today, the implementation of microfabricated devices in cancer research is frequent and advantageous because it enables the study of cancer cells in controlled microenvironments provided by the microchips. Breast cancer is one of the most common cancers in women, and the way breast cancer cells interact with their physical microenvironment is still under investigation. In this study, we developed a transparent cell culture chip (Ch-Pattern) with a micropillar-decorated bottom that makes live imaging and monitoring of the metabolic, proliferative, apoptotic, and morphological behavior of breast cancer cells possible. The reason for the use of micropatterned surfaces is because cancer cells deform and lose their shape and acto-myosin integrity on micropatterned substrates, and this allows the quantification of the changes in morphology and through that identification of the cancerous cells. In the last decade, cancer cells were studied on micropatterned substrates of varying sizes and with a variety of biomaterials. These studies were conducted using conventional cell culture plates carrying patterned films. In the present study, cell culture protocols were conducted in the clear-bottom micropatterned chip. This approach adds significantly to the current knowledge and applications by enabling low-volume and high-throughput processing of the cell behavior, especially the cell–micropattern interactions. In this study, two different breast cancer cell lines, MDA-MB-231 and MCF-7, were used. MDA-MB-231 cells are invasive and metastatic, while MCF-7 cells are not metastatic. The nuclei of these two cell types deformed to distinctly different levels on the micropatterns, had different metabolic and proliferation rates, and their cell cycles were affected. The Ch-Pattern chips developed in this study proved to have significant advantages when used in the biological analysis of live cells and highly beneficial in the study of screening breast cancer cell–substrate interactions in vitro.
Collapse
Affiliation(s)
- Menekse Ermis
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara 06800, Turkey; (M.E.); (E.A.); (O.K.)
| | - Ezgi Antmen
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara 06800, Turkey; (M.E.); (E.A.); (O.K.)
| | - Ozgur Kuren
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara 06800, Turkey; (M.E.); (E.A.); (O.K.)
| | - Utkan Demirci
- Canary Center for Cancer Early Detection, Department of Radiology, Electrical Engineering Department, Stanford University, Palo Alto, CA 94305, USA;
| | - Vasif Hasirci
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara 06800, Turkey; (M.E.); (E.A.); (O.K.)
- Department of Medical Engineering, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey
- ACU Biomaterials Center, Acibadem Mehmet Ali Aydinlar University, Istanbul 34684, Turkey
- Correspondence:
| |
Collapse
|
5
|
Cui W, Cheong A, Wang Y, Tsuchida Y, Liu Y, Tremblay KD, Mager J. MCRS1 is essential for epiblast development during early mouse embryogenesis. Reproduction 2020; 159:1-13. [PMID: 31671403 DOI: 10.1530/rep-19-0334] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/29/2019] [Indexed: 12/11/2022]
Abstract
Microspherule protein 1 (MCRS1, also known as MSP58) is an evolutionarily conserved protein that has been implicated in various biological processes. Although a variety of functions have been attributed to MCRS1 in vitro, mammalian MCRS1 has not been studied in vivo. Here we report that MCRS1 is essential during early murine development. Mcrs1 mutant embryos exhibit normal morphology at the blastocyst stage but cannot be recovered at gastrulation, suggesting an implantation failure. Outgrowth (OG) assays reveal that mutant blastocysts do not form a typical inner cell mass (ICM) colony, the source of embryonic stem cells (ESCs). Surprisingly, cell death and histone H4 acetylation analysis reveal that apoptosis and global H4 acetylation are normal in mutant blastocysts. However, analysis of lineage specification reveals that while the trophoblast and primitive endoderm are properly specified, the epiblast lineage is compromised and exhibits a severe reduction in cell number. In summary, our study demonstrates the indispensable role of MCRS1 in epiblast development during early mammalian embryogenesis.
Collapse
Affiliation(s)
- Wei Cui
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA.,Animal Models Core Facility, Institute for Applied Life Sciences (IALS), University of Massachusetts, Amherst, Massachusetts, USA
| | - Agnes Cheong
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Yongsheng Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Yuran Tsuchida
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Yong Liu
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, Fuyang Normal University, Fuyang, Anhui, People's Republic of China
| | - Kimberly D Tremblay
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Jesse Mager
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| |
Collapse
|
6
|
Expression of Rta in B Lymphocytes during Epstein-Barr Virus Latency. J Mol Biol 2020; 432:5227-5243. [PMID: 32710985 DOI: 10.1016/j.jmb.2020.07.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/29/2020] [Accepted: 07/20/2020] [Indexed: 11/21/2022]
Abstract
Rta of Epstein-Barr virus (EBV) is thought to be expressed only during the lytic cycle to promote the transcription of lytic genes. However, we found that Rta is expressed in EBV-infected B cells during viral latency, at levels detectable by immunoblot analysis. Latent Rta expression cannot be attributed to spontaneous lytic activation, as we observed that more than 90% of Akata, P3HR1, and 721 cells latently infected by EBV express Rta. We further found that Rta is sequestered in the nucleolus during EBV latency through its interaction with MCRS2, a nucleolar protein. When Rta is sequestered in the nucleolus, it no longer activates RNA polymerase II-driven transcription, thus explaining why Rta expression during latency does not transactivate EBV lytic genes. Additional experiments showed that Rta can bind to 18S rRNA and become incorporated into ribosomes, and a transient transfection experiment showed that Rta promotes translation from an mRNA reporter. These findings reveal that Rta has novel functions beyond transcriptional activation during EBV latency and may have interesting implications for the concept of EBV latency.
Collapse
|
7
|
Xia Y, Wang L, Ma X, Li X. Investigation on the Genomic Characterization of Uterine Sarcoma for rAd- p53 Combined with Chemotherapy Treatment. Hum Gene Ther 2020; 31:881-890. [PMID: 32013587 DOI: 10.1089/hum.2019.305] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The aim is to investigate the genomic characterization of uterine sarcoma for rAd-p53 (Gendicine®) combined with chemotherapy treatment. We recently published an article on 12 cases of uterine sarcomas, which were treated with rAd-p53 combined with chemotherapy. We found that rAd-p53 combined with chemotherapy is effective for various uterine sarcomas. Pretreatment pathological specimens of four uterine sarcoma patients were collected from the above recent clinical research and numbered 1-4A/B. Tumor samples were subjected to targeted sequencing by using a 416 genes panel. We profiled the mutation spectrum and tumor mutation burden in the tumors, identified mutated genes, and explored their gene function. We also verified the p53 protein expression using immunohistochemistry. We identified a total of 30 mutated genes that were found from the next-generation sequencing test results. The average number of mutated genes was up to seven in the five samples. TP53 gene was mutated in two of the four patients, No. 1 and No. 4B. They are c.C833G (p.P278R) missense mutation and a point mutation (C141*) that result in a premature stop codon. We did not find a mutated TP53 gene in the other two cases, but we identified mutated genes, including CREBBP, LYN, CDKN2A, and JAK2, which were located upstream of the TP53 gene; they may have an impact on TP53. We also identified 11 additional genes which are involved in p53-related signaling pathways or have interaction with p53. Compared to solid tumor mutational burden (TMB) distribution, none of their TMB was ranking in the top 25%. Mutant p53 protein expression was positive in two specimens. Our results demonstrated that the TP53 signaling pathway plays an important role in uterine sarcoma tumorigenesis. TP53 and the upstream genes such as CREBBP, LYN, CDKN2A, and JAK2 may be involved in the genomic characterization for rAd-p53 (Gendicine) combined with chemotherapy in uterine sarcoma. Besides, the average amount of mutated genes from every patient is large.
Collapse
Affiliation(s)
- Yu Xia
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lei Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaolin Ma
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiuqin Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
8
|
Park HJ, Ko HL, Won DH, Hwang DB, Shin YS, Kwak HW, Kim HJ, Yun JW, Nam JH. Comprehensive Analysis of the Safety Profile of a Single-Stranded RNA Nano-Structure Adjuvant. Pharmaceutics 2019; 11:E464. [PMID: 31500241 PMCID: PMC6781302 DOI: 10.3390/pharmaceutics11090464] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 01/01/2023] Open
Abstract
Adjuvants enhance the efficacy of vaccines by stimulating immune response-related gene expression and pathways. Although some adjuvants have been approved for commercial use in human vaccines (e.g., Alum, MF59, and AS03), they might elicit adverse side effects, such as autoimmune diseases. Recently, we developed a novel single-stranded RNA (ssRNA) nano-structure adjuvant, which can stimulate both Th1 and Th2 responses. In this study, we evaluated the safety and toxicological profiles of this ssRNA nano-structure adjuvant in vitro and in vivo. Mice were intramuscularly immunized with the ssRNA nano-structure adjuvant three times, once every 2 weeks. The results indicate no significant differences in hematological and serum biochemistry parameters between the ssRNA-treated groups and the control group. From a histopathological perspective, no evidence of tissue damage was found in any group. The levels of IgE and anti-nuclear antibodies, which are markers of autoimmune disease, were not different between the ssRNA-treated groups and the control group. The findings of this study suggest that the ssRNA nano-structure can be used as a safe adjuvant to increase vaccine efficacies.
Collapse
Affiliation(s)
- Hyeong-Jun Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Hae Li Ko
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Dong-Hoon Won
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Da-Bin Hwang
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Yoo-Sub Shin
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Hye-Won Kwak
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Hye-Jung Kim
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Jun-Won Yun
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Jae-Hwan Nam
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| |
Collapse
|
9
|
Abstract
Renal cell carcinomas (RCCs) are a diverse set of malignancies that have recently been shown to harbour mutations in a number of chromatin modifier genes - including PBRM1, SETD2, BAP1, KDM5C, KDM6A, and MLL2 - through high-throughput sequencing efforts. Current research focuses on understanding the biological activities that chromatin modifiers employ to suppress tumorigenesis and on developing clinical approaches that take advantage of this knowledge. Unsurprisingly, several common themes unify the functions of these epigenetic modifiers, particularly regulation of histone post-translational modifications and nucleosome organization. Furthermore, chromatin modifiers also govern processes crucial for DNA repair and maintenance of genomic integrity as well as the regulation of splicing and other key processes. Many chromatin modifiers have additional non-canonical roles in cytoskeletal regulation, which further contribute to genomic stability, expanding the repertoire of functions that might be essential in tumorigenesis. Our understanding of how mutations in chromatin modifiers contribute to tumorigenesis in RCC is improving but remains an area of intense investigation. Importantly, elucidating the activities of chromatin modifiers offers intriguing opportunities for the development of new therapeutic interventions in RCC.
Collapse
Affiliation(s)
- Aguirre A de Cubas
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - W Kimryn Rathmell
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
10
|
Tan P, Guo YH, Zhan JK, Long LM, Xu ML, Ye L, Ma XY, Cui XJ, Wang HQ. LncRNA-ANRIL inhibits cell senescence of vascular smooth muscle cells by regulating miR-181a/Sirt1. Biochem Cell Biol 2019; 97:571-580. [PMID: 30789795 DOI: 10.1139/bcb-2018-0126] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cardiovascular disease is one of the major threats to human life and health, and vascular aging is an important cause of its occurrence. Antisense non-coding RNA in the INK4 locus (ANRIL) is a kind of long non-coding RNA (lncRNA) that plays important roles in cell senescence. However, the role and mechanism of ANRIL in senescence of vascular smooth muscle cells (VSMCs) are unclear. METHODS Cell viability and cell cycle were evaluated using an MTT assay and flow cytometry analysis, respectively. Senescence-associated (SA)-β-galactosidase (gal) staining was used to determine cell senescence. Dual luciferase reporter assays were conducted to confirm the binding of ANRIL and miR-181a, as well as miR-181a and Sirt1. The expression of ANRIL, miR-181a, and Sirt1 was determined using qRT-PCR and protein levels of SA-β-gal and p53-p21 pathway-related proteins were evaluated by Western blotting. RESULTS ANRIL and Sirt1 were down-regulated, whereas miR-181a was up-regulated in aging VSMCs. In young and aging VSMCs, over-expression of ANRIL could down-regulate miR-181a and up-regulate Sirt1. MTT and SA-β-gal staining assays showed that over-expression of ANRIL and inhibition of miR-181a promoted cell viability and inhibited VSMC senescence. The dual-luciferase reporter assay determined that miR-181a directly targets ANRIL and the 3'-UTR of Sirt1. Furthermore, over-expression of ANRIL inhibited cell cycle arrest and the p53-p21 pathway. CONCLUSION ANRIL promotes cell viability and inhibits senescence in VSMCs, possibly by regulating miR-181a/Sirt1, and alleviating cell cycle arrest by inhibiting the p53-p21 pathway. This study provides novel insights for the role of ANRIL in the development of cell senescence.
Collapse
Affiliation(s)
- Pan Tan
- Department of Geriatrics, the Second Xiangya Hospital of Central South University, Changsha 410011, P.R. China
| | - Yong-Hong Guo
- Department of Geriatrics, the Second Xiangya Hospital of Central South University, Changsha 410011, P.R. China
| | - Jun-Kun Zhan
- Department of Geriatrics, the Second Xiangya Hospital of Central South University, Changsha 410011, P.R. China
| | - Li-Min Long
- Department of Geriatrics, the Second Xiangya Hospital of Central South University, Changsha 410011, P.R. China
| | - Mei-Li Xu
- Department of Geriatrics, the Second Xiangya Hospital of Central South University, Changsha 410011, P.R. China
| | - Ling Ye
- Department of Geriatrics, the Second Xiangya Hospital of Central South University, Changsha 410011, P.R. China
| | - Xin-Yu Ma
- Department of Anesthesiology, the Second Xiangya Hospital of Central South University, Changsha 410011, P.R. China
| | - Xing-Jun Cui
- Department of Geriatrics, the Second Xiangya Hospital of Central South University, Changsha 410011, P.R. China
| | - Hai-Qin Wang
- Department of Geriatrics, the Second Xiangya Hospital of Central South University, Changsha 410011, P.R. China
| |
Collapse
|
11
|
Yang H, Zhang F, Huang CJ, Liao J, Han Y, Hao P, Chu Y, Lu X, Li W, Yu H, Kang J. Mps1 regulates spindle morphology through MCRS1 to promote chromosome alignment. Mol Biol Cell 2019; 30:1060-1068. [PMID: 30785839 PMCID: PMC6724509 DOI: 10.1091/mbc.e18-09-0546] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Accurate partitioning of chromosomes during mitosis is essential for genetic stability and requires the assembly of the dynamic mitotic spindle and proper kinetochore–microtubule attachment. The spindle assembly checkpoint (SAC) monitors the incompleteness and errors in kinetochore–microtubule attachment and delays anaphase. The SAC kinase Mps1 regulates the recruitment of downstream effectors to unattached kinetochores. Mps1 also actively promotes chromosome alignment during metaphase, but the underlying mechanism is not completely understood. Here, we show that Mps1 regulates chromosome alignment through MCRS1, a spindle assembly factor that controls the dynamics of the minus end of kinetochore microtubules. Mps1 binds and phosphorylates MCRS1. This mechanism enables KIF2A localization to the minus end of spindle microtubules. Thus, our study reveals a novel role of Mps1 in regulating the dynamics of the minus end of microtubules and expands the functions of Mps1 in genome maintenance.
Collapse
Affiliation(s)
- Hongdan Yang
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China
| | - Fengxia Zhang
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China
| | - Ching-Jung Huang
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China
| | - Jun Liao
- School of Life Science and Technology, Shanghaitech University, Shanghai 201210, China
| | - Ying Han
- School of Life Science and Technology, Shanghaitech University, Shanghai 201210, China
| | - Piliang Hao
- School of Life Science and Technology, Shanghaitech University, Shanghai 201210, China
| | - Youjun Chu
- School of Life Science and Technology, Shanghaitech University, Shanghai 201210, China
| | - Xiaoai Lu
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China
| | - Wenshu Li
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China
| | - Hongtao Yu
- Howard Hughes Medical Institute, Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jungseog Kang
- College of Arts and Science, New York University at Shanghai, Shanghai 200122, China.,NYU-ECNU Center for Computational Chemistry, New York University at Shanghai, Shanghai 200062, China
| |
Collapse
|
12
|
Epigenetic Regulation of Organ Regeneration in Zebrafish. J Cardiovasc Dev Dis 2018; 5:jcdd5040057. [PMID: 30558240 PMCID: PMC6306890 DOI: 10.3390/jcdd5040057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 02/06/2023] Open
Abstract
The zebrafish is broadly used for investigating de novo organ regeneration, because of its strong regenerative potential. Over the past two decades of intense study, significant advances have been made in identifying both the regenerative cell sources and molecular signaling pathways in a variety of organs in adult zebrafish. Epigenetic regulation has gradually moved into the center-stage of this research area, aided by comprehensive work demonstrating that DNA methylation, histone modifications, chromatin remodeling complexes, and microRNAs are essential for organ regeneration. Here, we present a brief review of how these epigenetic components are induced upon injury, and how they are involved in sophisticated organ regeneration. In addition, we highlight several prospective research directions and their potential implications for regenerative medicine.
Collapse
|
13
|
mTORC1 Inactivation Promotes Colitis-Induced Colorectal Cancer but Protects from APC Loss-Dependent Tumorigenesis. Cell Metab 2018; 27:118-135.e8. [PMID: 29275959 DOI: 10.1016/j.cmet.2017.11.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/21/2017] [Accepted: 11/15/2017] [Indexed: 01/17/2023]
Abstract
Dietary habits that can induce inflammatory bowel disease (IBD) are major colorectal cancer (CRC) risk factors, but mechanisms linking nutrients, IBD, and CRC are unknown. Using human data and mouse models, we show that mTORC1 inactivation-induced chromosomal instability impairs intestinal crypt proliferation and regeneration, CDK4/6 dependently. This triggers interleukin (IL)-6-associated reparative inflammation, inducing crypt hyper-proliferation, wound healing, and CRC. Blocking IL-6 signaling or reactivating mTORC1 reduces inflammation-induced CRC, so mTORC1 activation suppresses tumorigenesis in IBD. Conversely, mTORC1 inactivation is beneficial in APC loss-dependent CRC. Thus, IL-6 blockers or protein-rich-diet-linked mTORC1 activation may prevent IBD-associated CRC. However, abolishing mTORC1 can mitigate CRC in predisposed patients with APC mutations. Our work reveals mTORC1 oncogenic and tumor-suppressive roles in intestinal epithelium and avenues to optimized and personalized therapeutic regimens for CRC.
Collapse
|
14
|
Tong Y, Hao J, Tu Q, Yu H, Yan L, Li Y, Lv L, Wang F, Iavarone A, Zhao X. A tree shrew glioblastoma model recapitulates features of human glioblastoma. Oncotarget 2017; 8:17897-17907. [PMID: 28199986 PMCID: PMC5392295 DOI: 10.18632/oncotarget.15225] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 01/16/2017] [Indexed: 01/09/2023] Open
Abstract
Tupaia belangeri (tree shrew), an animal species whose genome has significantly higher similarity to primates than rodents, has been used in biomedical research. To generate animal models that reproduce the human tumors more faithfully than rodents, we present the first report of a cancer model mimicking human tumor genetics in tree shrew. By engineering a lentiviral system for the transduction of mutant H-Ras and a shRNA against tree shrew p53, we successfully generated malignant glioma in tree shrew. The tree shrew glioma exhibited aggressive behavior and a relatively short latency, and markedly reduced animal survival. Remarkably, the biological features of human high-grade glioma (necrosis, microvascular proliferation, pseudopalisading) were all present in tree shrew glioma. Furthermore, genetic analysis of tree shrew glioma revealed that the tumors were clustered within the mesenchymal subgroup of human glioblastoma multiforme. Compared with the corresponding mouse glioma, tree shrew gliomas were markedly more similar to human glioblastoma at gene expression profile. The tree shrew glioma model provides colleagues working in the field of gliomas and cancer in general with a more accurate animal model.
Collapse
Affiliation(s)
- Yaohui Tong
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China.,Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, Yunnan, China
| | - Junjun Hao
- State Key Lab of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Qiu Tu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Hualin Yu
- Department of Neurological Surgery, The First Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Lanzhen Yan
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, Yunnan, China.,Kunming Primate Research Center, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Yuan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China.,Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Longbao Lv
- Kunming Primate Research Center, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Fei Wang
- Department of Neurological Surgery, The First Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Antonio Iavarone
- Institute for Cancer Genetics, Columbia University, New York, New York 10032, USA
| | - Xudong Zhao
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming 650223, Yunnan, China.,Kunming Primate Research Center, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| |
Collapse
|
15
|
He L, Chen Y, Feng J, Sun W, Li S, Ou M, Tang L. Cellular senescence regulated by SWI/SNF complex subunits through p53/p21 and p16/pRB pathway. Int J Biochem Cell Biol 2017; 90:29-37. [PMID: 28716547 DOI: 10.1016/j.biocel.2017.07.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 07/02/2017] [Accepted: 07/13/2017] [Indexed: 01/16/2023]
Abstract
SWI/SNF complex is an evolutionarily well-conserved chromatin-remodeling complex, which is implicated in the nucleosomes removing or sliding, impacting on the DNA repair, replication and genes expression regulation. The SWI/SNF complex consists up to 12 protein subunits. The catalytic subunits are BRG1 or BRM, which are exclusive ATPase subunits. BRG1 has been reported to play an important role in cellular senescence. However, The function of non-catalytic subunits involved in cellular senescence is rarely investigated. Therefore, we focused on the senescence regulation roles of SWI/SNF non-catalytic subunits in cellular senescent model induced by H2O2. H2O2 treatment was used to induce cellular senescence models in vitro. Screening the candidate subunits involved in this process by comparing the expression levels of SWI/SNF subunits with/without H2O2 treatment. Over-expression and knockdown the candidate subunits were utilized to investigate the functions and mechanism of the subunits involved in senescence regulation. The expressions of BAF57, BAF60a and SNF5 were changed significantly after H2O2 treatment. Overexpression of the three subunits separately induced cell growth arrest in both HaCaT and GLL19 cells, while knockdown of the subunits separately eased the senescence induced by H2O2 treatment. Results further showed that BAF57, BAF60a and SNF5 regulated cellular senescence via both p53/p21 and p16/pRB pathways, and the three subunits all had a directly interaction with p53. These results indicated that BAF57, BAF60a and SNF5 might act as novel pro-senescence factors in both normal and tumor human skin cells. Therefore, inhibiting expression of the three factors might delay the cellular senescence process.
Collapse
Affiliation(s)
- Ling He
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Ying Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Jianguo Feng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China; Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, China
| | - Weichao Sun
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Shun Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Mengting Ou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.
| |
Collapse
|
16
|
Wang G, Fu Y, Hu F, Lan J, Xu F, Yang X, Luo X, Wang J, Hu J. Loss of BRG1 induces CRC cell senescence by regulating p53/p21 pathway. Cell Death Dis 2017; 8:e2607. [PMID: 28182012 PMCID: PMC5386468 DOI: 10.1038/cddis.2017.1] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 11/21/2016] [Accepted: 11/29/2016] [Indexed: 12/18/2022]
Abstract
Brahma-related gene-1 (BRG1) is the specific ATPase of switch/sucrose nonfermentable chromatin-remodeling complex that is aberrantly expressed or mutated in various cancers. However, the exact role of BRG1 in oncogenesis remains unknown. In this study, we demonstrate that the knockdown (KD) of BRG1 promotes cellular senescence by influencing the SIRT1/p53/p21 signal axis in colorectal cancer (CRC). In particular, we reveal that the expression level of BRG1 is inversely correlated with p21, one of the classic senescence regulators, and is decreased in senescent CRC cells. KD of BRG1 promoting senescence is indicated by the increase of senescence-associated β-galactosidase (SA-β-gal) activity, inhibition of cell proliferation, induction of cell cycle arrest, and formation of senescence-associated heterochromatin foci. BRG1 binds to SIRT1 and interferes with SIRT1-mediated deacetylation of p53 at K382. Rescue experiments by co-silencing p53 or treatment with EX527, a SIRT1-specific inhibitor, abrogated the cellular senescence induced by KD of BRG1. BRG1 KD cells resulted in smaller tumor formation than that in control cells in vivo. Collectively, our study shows that BRG1 has an important role in cellular senescence and tumor growth. The BRG1/SIRT1/p53 signal axis is a novel mechanism of cell senescence in CRC and is a new potential target for cancer therapy.
Collapse
Affiliation(s)
- Guihua Wang
- Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yinjia Fu
- Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Fuqing Hu
- Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jinqing Lan
- Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Xu
- Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Yang
- Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xuelai Luo
- Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junbo Hu
- Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Hou H, Xing W, Li W. Brahma-related gene 1 induces apoptosis in a p53-dependent manner in human rheumatoid fibroblast-like synoviocyte MH7A. Medicine (Baltimore) 2016; 95:e5241. [PMID: 28002318 PMCID: PMC5181802 DOI: 10.1097/md.0000000000005241] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Blocked apoptosis and aggressive inflammatory responses occur in fibroblast-like synoviocyte (FLS) of rheumatoid arthritis (RA) patients. Although Brahma-related gene 1 (BRG1) is considered as a tumor suppressor, few research covers its role in RA. This study aims to reveal effects and potential mechanisms of BRG1 in human FLS cell line MH7A.BRG1 expression in MH7A cells was altered by transfection of overexpression vectors or short hairpin RNAs (shRNAs). Cell viability and apoptosis were detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and flow cytometry after transfection. Factors involved in inflammation and apoptosis were quantified by qPCR and Western blot. The interaction between BRG1 and p53 was assessed by immunoprecipitation (IP).Results showed that BRG1 overexpression significantly suppressed MH7A cell viability and induced apoptosis (P < 0.01), and its knockdown had opposite effects. BRG1 reduced mRNA levels of matrix metallopeptidase 3, TIMP metallopeptidase inhibitor 2, cyclooxygenase 2, and interleukin 6, implying its suppressive effects on inflammation. BRG1 interacted with and promoted p53 (P < 0.05). B-cell chronic lymphocytic leukemia/lymphoma 2 was suppressed (P < 0.05), while cytochrome c, caspase 3 (CASP3) and CASP9 were activated (P < 0.01) by BRG1. However, the regulation on these factors was abrogated by p53 knockdown (P < 0.01).These findings suggest that BRG1 may induce apoptosis and suppress inflammation in MH7A cells. Potential functional mechanisms involve the regulation of apoptotic factors by BRG1, which may depend on the recruitment and promotion of p53. This study provides the essential proof for applying BRG1 to the molecular therapy of RA.
Collapse
|
18
|
Yang CP, Kuo YL, Lee YC, Lee KH, Chiang CW, Wang JM, Hsu CC, Chang WC, Lin DY. RINT-1 interacts with MSP58 within nucleoli and plays a role in ribosomal gene transcription. Biochem Biophys Res Commun 2016; 478:873-80. [PMID: 27530925 DOI: 10.1016/j.bbrc.2016.08.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 08/07/2016] [Indexed: 11/27/2022]
Abstract
The nucleolus is the cellular site of ribosomal (r)DNA transcription and ribosome biogenesis. The 58-kDa microspherule protein (MSP58) is a nucleolar protein involved in rDNA transcription and cell proliferation. However, regulation of MSP58-mediated rDNA transcription remains unknown. Using a yeast two-hybrid system with MSP58 as bait, we isolated complementary (c)DNA encoding Rad50-interacting protein 1 (RINT-1), as a MSP58-binding protein. RINT-1 was implicated in the cell cycle checkpoint, membrane trafficking, Golgi apparatus and centrosome dynamic integrity, and telomere length control. Both in vitro and in vivo interaction assays showed that MSP58 directly interacts with RINT-1. Interestingly, microscopic studies revealed the co-localization of MSP58, RINT-1, and the upstream binding factor (UBF), a rRNA transcription factor, in the nucleolus. We showed that ectopic expression of MSP58 or RINT-1 resulted in decreased rRNA expression and rDNA promoter activity, whereas knockdown of MSP58 or RINT-1 by siRNA exerted the opposite effect. Coexpression of MSP58 and RINT-1 robustly decreased rRNA synthesis compared to overexpression of either protein alone, whereas depletion of RINT-1 from MSP58-transfected cells enhanced rRNA synthesis. We also found that MSP58, RINT-1, and the UBF were associated with the rDNA promoter using a chromatin immunoprecipitation assay. Because aberrant ribosome biogenesis contributes to neoplastic transformation, our results revealed a novel protein complex involved in the regulation of rRNA gene expression, suggesting a role for MSP58 and RINT-1 in cancer development.
Collapse
Affiliation(s)
- Chuan-Pin Yang
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan, ROC
| | - Yu-Liang Kuo
- Department of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung, 40201, Taiwan, ROC; Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung, 40201, Taiwan, ROC
| | - Yi-Chao Lee
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan, ROC
| | - Kuen-Haur Lee
- Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan, ROC
| | - Chi-Wu Chiang
- Infectious Diseases and Signaling Research Center, National Cheng Kung University, Tainan, 70101, Taiwan, ROC; Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC
| | - Ju-Ming Wang
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan, ROC; Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan, ROC; Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC; Infectious Diseases and Signaling Research Center, National Cheng Kung University, Tainan, 70101, Taiwan, ROC
| | - Che-Chia Hsu
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan, ROC
| | - Wen-Chang Chang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan, ROC; Infectious Diseases and Signaling Research Center, National Cheng Kung University, Tainan, 70101, Taiwan, ROC; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan, ROC; Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, 11031, Taiwan, ROC.
| | - Ding-Yen Lin
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan, ROC; Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan, ROC; Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC; Infectious Diseases and Signaling Research Center, National Cheng Kung University, Tainan, 70101, Taiwan, ROC; Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan, ROC.
| |
Collapse
|
19
|
Li C, Chen M, Zhao P, Ayana DA, Wang L, Jiang Y. Expression of MCRS1 and MCRS2 and their correlation with serum carcinoembryonic antigen in colorectal cancer. Exp Ther Med 2016; 12:589-596. [PMID: 27446248 PMCID: PMC4950148 DOI: 10.3892/etm.2016.3424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 03/03/2016] [Indexed: 12/16/2022] Open
Abstract
Cancer-associated genes serve a crucial role in carcinogenesis. The present study aimed to investigate the mRNA expression levels of microspherule protein 1 (MCRS1) and MCRS2 in colorectal cancer (CRC) and their association with clinical variables. The mRNA expression levels of MCRS1 and MCRS2 were assessed by semi-quantitative reverse transcription polymerase chain reaction in the tumor and corresponding non-tumor tissues of 54 newly-diagnosed CRC patients, as well as in the normal colonic mucosa tissue of 19 age/gender-matched healthy controls. Immunofluorescence was also employed to identify the expression of MCRS1 in CRC tissues, while the concentration of serum carcinoembryonic antigen (CEA) was determined by an enzyme-linked immunoassay. The results identified a negative correlation between MCRS1 and MCRS2 expression levels (r=-0.3018, P=0.0266). MCRS1 mRNA expression was significantly increased and MCRS2 mRNA expression was decreased in CRC tissues compared with the levels in the corresponding normal tissues (both P<0.001). An increase in MCRS1 expression and a decrease in MCRS2 expression was detected in advanced stage when compared with early stage CRC patients. Immunofluorescence analysis revealed increased expression of MCRS1 in CRC patients. Furthermore, the expression levels of MCRS1 displayed positive correlation, whilst those of MCRS2 displayed negative correlation, with the serum CEA level in patients with CRC. The results suggest that increased MCRS1 and decreased MCRS2 expression appeared to be involved in the pathogenesis of CRC. The present study provides evidence suggesting that MCRS1 and MCRS2 may identify CRC patients at a risk of disease relapse, and thus, may be potential tools for monitoring disease activity and act as novel diagnostic markers in the treatment of CRC.
Collapse
Affiliation(s)
- Chenguang Li
- Department of Colorectal and Anal Surgery, The First Hospital, Jilin University, Changchun, Jilin 130032, P.R. China; Key Laboratory of Zoonosis Research, Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130032, P.R. China
| | - Mingxiao Chen
- Department of Colorectal and Anal Surgery, The First Hospital, Jilin University, Changchun, Jilin 130032, P.R. China
| | - Pingwei Zhao
- Department of Colorectal and Anal Surgery, The First Hospital, Jilin University, Changchun, Jilin 130032, P.R. China
| | - Desalegn Admassu Ayana
- Department of Medical Laboratory Sciences, Haramaya University, Dire Dawa 3000, Ethiopia
| | - Lei Wang
- Department of Colorectal and Anal Surgery, The First Hospital, Jilin University, Changchun, Jilin 130032, P.R. China
| | - Yanfang Jiang
- Key Laboratory of Zoonosis Research, Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130032, P.R. China
| |
Collapse
|
20
|
Liu M, Zhou K, Huang Y, Cao Y. The candidate oncogene (MCRS1) promotes the growth of human lung cancer cells via the miR-155-Rb1 pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:121. [PMID: 26467212 PMCID: PMC4606992 DOI: 10.1186/s13046-015-0235-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 10/05/2015] [Indexed: 12/23/2022]
Abstract
Background Microspherule protein 1 (MCRS1) is a candidate oncogene and participates in various cellular processes, including growth, migration, senescence and transformation. MCRS1 is overexpressed in non-small cell lung cancer (NSCLC) and promotes the growth of cancer cells. However, the mechanisms driving these processes are not fully understood. Methods Retrovirus-mediated RNA interference was employed to knockdown MCRS1 expression in cell lines. Cell proliferation assays and animal experiments were respectively performed to evaluate the growth of NSCLC cells in vitro and in vivo. Microarray analysis was carried out for mRNA profiling. Luciferase reporter assay and microRNA (miRNA) transfection were used to investigate the interaction between miRNA and gene. Results Stably knocking down MCRS1 expression inhibited the proliferation of NSCLC cells in vitro and in vivo. By comparing the mRNA expression profiles of NSCLC cells with or without MCRS1 silencing, we found that MCRS1 regulated expressions of various genes related to cell proliferation, including Rb1, TP53, cell cycle-related genes, MYC, E2F2, PCNA, and Ki67. However, MCRS1 did not directly bind to these differentially expressed genes. Here, we confirmed that Rb1, an important tumor suppression gene (TSG), is a direct target of miR-155 which is directly up-regulated by MCRS1. Furthermore, the level of Rb1 expression in NSCLC tissues was inversely correlated with those of miR-155 and MCRS1, and MCRS1 regulated expression of Rb1 via miR-155. Additionally, we found that the DNA copy number of the MCRS1 gene played a role in MCRS1 overexpression in NSCLCs. Conclusion MCRS1 overexpression induced NSCLC proliferation through the miR-155–Rb1 pathway and DNA copy-number amplification is one of the mechanisms underlying MCRS1 overexpression in NSCLC. Moreover, we put forward the hypothesis that there are regulatory relationships between oncogenes and TSGs apart from the functional synergy of both; the oncogene-miRNA-TSG networks are one of mechanisms among the regulatory relationships; the regulatory relationships and the networks might play active roles in the development and progression of cancer. Electronic supplementary material The online version of this article (doi:10.1186/s13046-015-0235-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Minxia Liu
- Laboratory of Molecular and Experimental Pathology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China. .,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China.
| | - Kecheng Zhou
- Laboratory of Molecular and Experimental Pathology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China. .,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, China.
| | - Yunchao Huang
- Department of Thoracic and Cardiovascular Surgery, The Third Affiliated Hospital of Kunming Medical University (Yunnan Tumor Hospital), Kunming, China.
| | - Yi Cao
- Laboratory of Molecular and Experimental Pathology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
| |
Collapse
|
21
|
Petrik D, Latchney SE, Masiulis I, Yun S, Zhang Z, Wu JI, Eisch AJ. Chromatin Remodeling Factor Brg1 Supports the Early Maintenance and Late Responsiveness of Nestin-Lineage Adult Neural Stem and Progenitor Cells. Stem Cells 2015; 33:3655-65. [PMID: 26418130 DOI: 10.1002/stem.2215] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 09/14/2015] [Indexed: 12/25/2022]
Abstract
Insights from embryonic development suggest chromatin remodeling is important in adult neural stem cells (aNSCs) maintenance and self-renewal, but this concept has not been fully explored in the adult brain. To assess the role of chromatin remodeling in adult neurogenesis, we inducibly deleted Brg1--the core subunit of SWI/SNF-like Brg1/Brm-associated factor chromatin remodeling complexes--in nestin-expressing aNSCs and their progeny in vivo and in culture. This resulted in abnormal adult neurogenesis in the hippocampus, which initially reduced hippocampal aNSCs and progenitor maintenance, and later reduced its responsiveness to physiological stimulation. Mechanistically, deletion of Brg1 appeared to impair cell cycle progression, which is partially due to elevated p53 pathway and p21 expression. Knockdown of p53 rescued the neurosphere growth defects caused by Brg1 deletion. Our results show that epigenetic chromatin remodeling (via a Brg1 and p53/p21-dependent process) determines the aNSCs and progenitor maintenance and responsiveness of neurogenesis.
Collapse
Affiliation(s)
- David Petrik
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sarah E Latchney
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Irene Masiulis
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sanghee Yun
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Zilai Zhang
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jiang I Wu
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Amelia J Eisch
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
22
|
Peng J, Ma J, Li W, Mo R, Zhang P, Gao K, Jin X, Xiao J, Wang C, Fan J. Stabilization of MCRS1 by BAP1 prevents chromosome instability in renal cell carcinoma. Cancer Lett 2015; 369:167-74. [PMID: 26300492 DOI: 10.1016/j.canlet.2015.08.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/17/2015] [Accepted: 08/17/2015] [Indexed: 01/21/2023]
Abstract
Characterization of the exome and genome of carcinoma (ccRCC) by next-generation sequencing identified numerous genetic alternations. BRCA1-associated protein-1 (BAP1) was identified as one of the most frequently mutated genes in ccRCC, suggesting that BAP1 is a potential key driver for ccRCC cancer initiation and progression. However, how BAP1 mutations contribute to ccRCC remains to be elucidated. BAP1 is a nuclear de-ubiquitinating enzyme and cleaves the ubiquitin chain from the substrates. Here, we identified MCRS1 as a bona fide substrate for BAP1. MCRS1 is a component of the centrosome proteins, and plays an essential role in spindle assembly. BAP1 binds to MCRS1 and stabilizes MCRS1 by de-ubiquitination. BAP1 contributes to chromosome stability partially via MCRS1. A positive correlation was identified between BAP1 and MCRS1 expression in ccRCC tissues. Both BAP1 loss and MCRS1 down-regulation in ccRCC were associated with adverse clinicopathological features. This study revealed a novel mechanism for BAP1 involved in MCRS1 stability regulation, and provided insight in understanding the relationship between BAP1 mutations and chromosome instability in ccRCC.
Collapse
Affiliation(s)
- Jingtao Peng
- Department of Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 100 Haining Road, Shanghai 200080, China
| | - Jian Ma
- Department of Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 100 Haining Road, Shanghai 200080, China
| | - Weiguo Li
- Department of Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 100 Haining Road, Shanghai 200080, China
| | - Ren Mo
- Department of Urology, Inner Mongolia Autonomous Region Peoples Hospital, 20 Zhaowuda Road, Hohhot 010017, China
| | - Pingzhao Zhang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai 200433, China
| | - Kun Gao
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai 200433, China
| | - Xiaofeng Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai 200433, China
| | - Jiantao Xiao
- Department of Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 100 Haining Road, Shanghai 200080, China
| | - Chenji Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, 2005 Songhu Road, Shanghai 200433, China.
| | - Jie Fan
- Department of Urology, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, 100 Haining Road, Shanghai 200080, China.
| |
Collapse
|
23
|
Yang CP, Chiang CW, Chen CH, Lee YC, Wu MH, Tsou YH, Yang YS, Chang WC, Lin DY. Identification and characterization of nuclear and nucleolar localization signals in 58-kDa microspherule protein (MSP58). J Biomed Sci 2015; 22:33. [PMID: 25981436 PMCID: PMC4434885 DOI: 10.1186/s12929-015-0136-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 04/21/2015] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND MSP58 is a nucleolar protein associated with rRNA transcription and cell proliferation. Its mechanism of translocation into the nucleus or the nucleolus, however, is not entirely known. In order to address this lack, the present study aims to determine a crucial part of this mechanism: the nuclear localization signal (NLS) and the nucleolar localization signal (NoLS) associated with the MSP58 protein. RESULTS We have identified and characterized two NLSs in MSP58. The first is located between residues 32 and 56 (NLS1) and constitutes three clusters of basic amino acids (KRASSQALGTIPKRRSSSRFIKRKK); the second is situated between residues 113 and 123 (NLS2) and harbors a monopartite signal (PGLTKRVKKSK). Both NLS1 and NLS2 are highly conserved among different vertebrate species. Notably, one bipartite motif within the NLS1 (residues 44-56) appears to be absolutely necessary for MSP58 nucleolar localization. By yeast two-hybrid, pull-down, and coimmunoprecipitation analysis, we show that MSP58 binds to importin α1 and α6, suggesting that nuclear targeting of MSP58 utilizes a receptor-mediated and energy-dependent import mechanism. Functionally, our data show that both nuclear and nucleolar localization of MSP58 are crucial for transcriptional regulation on p21 and ribosomal RNA genes, and context-dependent effects on cell proliferation. CONCLUSIONS Results suggest that MSP58 subnuclear localization is regulated by two nuclear import signals, and that proper subcellular localization of MSP58 is critical for its role in transcriptional regulation. Our study reveals a molecular mechanism that controls nuclear and nucleolar localization of MSP58, a finding that might help future researchers understand the MSP58 biological signaling pathway.
Collapse
Affiliation(s)
- Chuan-Pin Yang
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan, ROC.
| | - Chi-Wu Chiang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC.
- Infectious Diseases and Signaling Research Center, National Cheng Kung University, Tainan, 70101, Taiwan, ROC.
| | - Chang-Han Chen
- Center for Translational Research in Biomedical Sciences, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 83301, Taiwan, ROC.
- Department of Applied Chemistry, National Chi Nan University, Puli, Nantou, 54561, Taiwan, ROC.
| | - Yi-Chao Lee
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, 11031, Taiwan, ROC.
- Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan, ROC.
| | - Mei-Hsiang Wu
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan, ROC.
| | - Yi-Huan Tsou
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC.
| | - Yu-San Yang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC.
| | - Wen-Chang Chang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC.
- Infectious Diseases and Signaling Research Center, National Cheng Kung University, Tainan, 70101, Taiwan, ROC.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan, ROC.
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, 11031, Taiwan, ROC.
| | - Ding-Yen Lin
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, 70101, Taiwan, ROC.
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan, ROC.
- Infectious Diseases and Signaling Research Center, National Cheng Kung University, Tainan, 70101, Taiwan, ROC.
- Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan, ROC.
| |
Collapse
|
24
|
Fawal MA, Brandt M, Djouder N. MCRS1 binds and couples Rheb to amino acid-dependent mTORC1 activation. Dev Cell 2015; 33:67-81. [PMID: 25816988 DOI: 10.1016/j.devcel.2015.02.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 12/11/2014] [Accepted: 02/10/2015] [Indexed: 12/31/2022]
Abstract
Ras homolog enriched in brain (Rheb) is critical for mechanistic target of rapamycin complex 1 (mTORC1) activation in response to growth factors and amino acids (AAs). Whereas growth factors inhibit the tuberous sclerosis complex (TSC1-TSC2), a negative Rheb regulator, the role of AAs in Rheb activation remains unknown. Here, we identify microspherule protein 1 (MCRS1) as the essential link between Rheb and mTORC1 activation. MCRS1, in an AA-dependent manner, maintains Rheb at lysosome surfaces, connecting Rheb to mTORC1. MCRS1 suppression in human cancer cells using small interference RNA or mouse embryonic fibroblasts using an inducible-Cre/Lox system reduces mTORC1 activity. MCRS1 depletion promotes Rheb/TSC2 interaction, rendering Rheb inactive and delocalizing it from lysosomes to recycling endocytic vesicles, leading to mTORC1 inactivation. These findings have important implications for signaling mechanisms in various pathologies, including diabetes mellitus and cancer.
Collapse
Affiliation(s)
- Mohamad-Ali Fawal
- Cancer Cell Biology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, CNIO, 28029 Madrid, Spain
| | - Marta Brandt
- Cancer Cell Biology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, CNIO, 28029 Madrid, Spain
| | - Nabil Djouder
- Cancer Cell Biology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, CNIO, 28029 Madrid, Spain.
| |
Collapse
|
25
|
Yeh CH, Kuo PL, Wang YY, Wu YY, Chen MF, Lin DY, Lai TH, Chiang HS, Lin YH. SEPT12/SPAG4/LAMINB1 complexes are required for maintaining the integrity of the nuclear envelope in postmeiotic male germ cells. PLoS One 2015; 10:e0120722. [PMID: 25775403 PMCID: PMC4361620 DOI: 10.1371/journal.pone.0120722] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 01/26/2015] [Indexed: 11/19/2022] Open
Abstract
Male infertility affects approximately 50% of all infertile couples. The male-related causes of intracytoplasmic sperm injection failure include the absence of sperm, immotile or immature sperm, and sperm with structural defects such as those caused by premature chromosomal condensation and DNA damage. Our previous studies based on a knockout mice model indicated that SEPT12 proteins are critical for the terminal morphological formation of sperm. SEPT12 mutations in men result in teratozospermia and oligozospermia. In addition, the spermatozoa exhibit morphological defects of the head and tail, premature chromosomal condensation, and nuclear damage. However, the molecular functions of SEPT12 during spermatogenesis remain unclear. To determine the molecular functions of SEPT12, we applied a yeast 2-hybrid system to identify SEPT12 interactors. Seven proteins that interact with SEPT12 were identified: SEPT family proteins (SEPT4 and SEPT6), nuclear or nuclear membrane proteins (protamine 2, sperm-associated antigen 4, and NDC1 transmembrane nucleoproine), and sperm-related structural proteins (pericentriolar material 1 and obscurin-like 1). Sperm-associated antigen 4 (SPAG4; also known as SUN4) belongs to the SUN family of proteins and acts as a linker protein between nucleoskeleton and cytoskeleton proteins and localizes in the nuclear membrane. We determined that SEPT12 interacts with SPAG4 in a male germ cell line through coimmunoprecipitation. During human spermiogenesis, SEPT12 is colocalized with SPAG4 near the nuclear periphery in round spermatids and in the centrosome region in elongating spermatids. Furthermore, we observed that SEPT12/SPAG4/LAMINB1 formed complexes and were coexpressed in the nuclear periphery of round spermatids. In addition, mutated SEPT12, which was screened from an infertile man, affected the integration of these nuclear envelope complexes through coimmunoprecipitation. This was the first study that suggested that SEPT proteins link to the SUN/LAMIN complexes during the formation of nuclear envelopes and are involved in the development of postmeiotic germ cells.
Collapse
Affiliation(s)
- Chung-Hsin Yeh
- Division of Urology, Department of Surgery, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Pao-Lin Kuo
- Department of Obstetrics & Gynecology, National Cheng Kung University, College of Medicine, Tainan, Taiwan
| | - Ya-Yun Wang
- Department of Obstetrics & Gynecology, National Cheng Kung University, College of Medicine, Tainan, Taiwan
| | - Ying-Yu Wu
- Graduate Institute of Basic Medicine, Fu Jen Catholic University, College of Medicine, New Taipei City, Taiwan
| | - Mei-Feng Chen
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Tao-Yuan, Taiwan
| | - Ding-Yen Lin
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Tsung-Hsuan Lai
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Department of Obstetrics and Gynecology, Cathay General Hospital, Taipei City, Taiwan
- Institute of Systems Biology and Bioinformatics, National Central University, Zhongli City, Taoyuan County, Taiwan
| | - Han-Sun Chiang
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Basic Medicine, Fu Jen Catholic University, College of Medicine, New Taipei City, Taiwan
| | - Ying-Hung Lin
- Graduate Institute of Basic Medicine, Fu Jen Catholic University, College of Medicine, New Taipei City, Taiwan
| |
Collapse
|
26
|
Zhang L, Nemzow L, Chen H, Hu JJ, Gong F. Whole genome expression profiling shows that BRG1 transcriptionally regulates UV inducible genes and other novel targets in human cells. PLoS One 2014; 9:e105764. [PMID: 25157878 PMCID: PMC4144907 DOI: 10.1371/journal.pone.0105764] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/19/2014] [Indexed: 12/16/2022] Open
Abstract
UV irradiation is known to cause cyclobutane pyrimidine dimers (CPDs) and pyrimidine (6–4) pyrimidone photoproducts (6-4PPs), and plays a large role in the development of cancer. Tumor suppression, through DNA repair and proper cell cycle regulation, is an integral factor in maintaining healthy cells and preventing development of cancer. Transcriptional regulation of the genes involved in the various tumor suppression pathways is essential for them to be expressed when needed and to function properly. BRG1, an ATPase catalytic subunit of the SWI/SNF chromatin remodeling complex, has been identified as a tumor suppressor protein, as it has been shown to play a role in Nucleotide Excision Repair (NER) of CPDs, suppress apoptosis, and restore checkpoint deficiency, in response to UV exposure. Although BRG1 has been shown to regulate transcription of some genes that are instrumental in proper DNA damage repair and cell cycle maintenance in response to UV, its role in transcriptional regulation of the whole genome in response to UV has not yet been elucidated. With whole genome expression profiling in SW13 cells, we show that upon UV induction, BRG1 regulates transcriptional expression of many genes involved in cell stress response. Additionally, our results also highlight BRG1's general role as a master regulator of the genome, as it transcriptionally regulates approximately 4.8% of the human genome, including expression of genes involved in many pathways. RT-PCR and ChIP were used to validate our genome expression analysis. Importantly, our study identifies several novel transcriptional targets of BRG1, such as ATF3. Thus, BRG1 has a larger impact on human genome expression than previously thought, and our studies will provide inroads for future analysis of BRG1's role in gene regulation.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Leah Nemzow
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Hua Chen
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Jennifer J. Hu
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Feng Gong
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
27
|
Hsu CC, Chen CH, Hsu TI, Hung JJ, Ko JL, Zhang B, Lee YC, Chen HK, Chang WC, Lin DY. The 58-kda microspherule protein (MSP58) represses human telomerase reverse transcriptase (hTERT) gene expression and cell proliferation by interacting with telomerase transcriptional element-interacting factor (TEIF). BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1843:565-579. [PMID: 24361335 DOI: 10.1016/j.bbamcr.2013.12.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 11/13/2013] [Accepted: 12/04/2013] [Indexed: 11/21/2022]
Abstract
58-kDa microspherule protein (MSP58) plays an important role in a variety of cellular processes including transcriptional regulation, cell proliferation and oncogenic transformation. Currently, the mechanisms underlying the oncogenic effect of MSP58 are not fully understood. The human telomerase reverse transcriptase (hTERT) gene, which encodes an essential component for telomerase activity that is involved in cellular immortalization and transformation, is strictly regulated at the gene transcription level. Our previous study revealed a novel function of MSP58 in cellular senescence. Here we identify telomerase transcriptional element-interacting factor (TEIF) as a novel MSP58-interacting protein and determine the effect of MSP58 on hTERT transcription. This study thus provides evidence showing MSP58 to be a negative regulator of hTERT expression and telomerase activity. Luciferase reporter assays indicated that MSP58 could suppress the transcription ofhTERTpromoter. Additionally, stable overexpression of MSP58 protein in HT1080 and 293T cells decreased both endogenous hTERT expression and telomerase activity. Conversely, their upregulation was induced by MSP58 silencing. Chromatin immunoprecipitation assays showed that MSP58 binds to the hTERT proximal promoter. Furthermore, overexpression of MSP58 inhibited TEIF-mediated hTERT transactivation, telomerase activation, and cell proliferation promotion. The inhibitory effect of MSP58 occurred through inhibition of TEIF binding to DNA. Ultimately, the HT1080-implanted xenograft mouse model confirmed these cellular effects. Together, our findings provide new insights into both the biological function of MSP58 and the regulation of telomerase/hTERT expression.
Collapse
|
28
|
Udden SMN, Morita-Fujimura Y, Satake M, Ikawa S. c-ABL tyrosine kinase modulates p53-dependent p21 induction and ensuing cell fate decision in response to DNA damage. Cell Signal 2013; 26:444-52. [PMID: 24177958 DOI: 10.1016/j.cellsig.2013.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 10/11/2013] [Accepted: 10/21/2013] [Indexed: 02/05/2023]
Abstract
The c-ABL non-receptor tyrosine kinase and the p53 tumor suppressor protein are pivotal modulators of cellular responses to DNA damage. However, a comprehensive understanding of the role of c-ABL kinase in p53-dependent transcription of p21(CIP1/WAF1) and ensuing cell fate decision is still obscure. Here, we demonstrate that c-ABL tyrosine kinase regulates p53-dependent induction of p21. As a result, it modulates cell fate decision by p53 in response to DNA damage differently according to the extent of DNA damage. When human cancer cells were treated with DNA damaging agent, adriamycin (0.08 μg/ml), p21 was induced following p53 induction. Owing largely to p21, a substantial fraction of cells treated with adriamycin were blocked at the G2 phase of the cell cycle and most cells eventually became senescent. When these cells were simultaneously treated with a c-ABL kinase inhibitor, STI571, or a c-ABL-specific siRNA along with adriamycin, the p53-dependent p21 induction was dramatically diminished, even though p53 is substantially induced. Accordingly, G2-arrest, and cellular senescence largely dependent on p21 were substantially abrogated. On the contrary, when cells were treated with a relatively high dose of adriamycin (0.4 μg/ml) cells became apoptotic, and the simultaneous presence of a c-ABL kinase inhibitor STI571 augmented the extent of apoptosis. We speculate this is due to abrogation of p53-dependent p21 induction, which leads to elimination of anti-apoptotic function of p21. In summary, c-ABL appears to promote senescence or inhibit apoptosis, depending on the extent of DNA damage. These findings suggest that the combined use of ABL kinase inhibitor and DNA damaging drug in chemotherapy against tumors retaining wild type p53 should be carefully designed.
Collapse
Affiliation(s)
- S M Nashir Udden
- Center for Interdisciplinary Research, Tohoku University, Sendai 980-8578, Japan; Department of Project Programs, Tohoku University, Sendai 980-8575, Japan; Department of Molecular Immunology, Institute of Development, Aging, and Cancer, Tohoku University, Sendai 980-8575, Japan; Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Yuiko Morita-Fujimura
- Center for Interdisciplinary Research, Tohoku University, Sendai 980-8578, Japan; Department of Project Programs, Tohoku University, Sendai 980-8575, Japan; International Advanced Research and Education Organization, Tohoku University, Sendai 980-8578, Japan
| | - Masanobu Satake
- Department of Molecular Immunology, Institute of Development, Aging, and Cancer, Tohoku University, Sendai 980-8575, Japan; Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Shuntaro Ikawa
- Center for Interdisciplinary Research, Tohoku University, Sendai 980-8578, Japan; Department of Project Programs, Tohoku University, Sendai 980-8575, Japan.
| |
Collapse
|
29
|
Lin W, Li XM, Zhang J, Huang Y, Wang J, Zhang J, Jiang XF, Fei Z. Increased expression of the 58-kD microspherule protein (MSP58) is correlated with poor prognosis in glioma patients. Med Oncol 2013; 30:677. [PMID: 23996240 DOI: 10.1007/s12032-013-0677-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 07/21/2013] [Indexed: 10/26/2022]
Abstract
The pathological grading system for human gliomas is usually used to evaluate the prognosis of glioma patients. However, some glioma patients with similar grades have obvious discrepancies in survival. It is therefore necessary to identify some new certain tumor biomarkers that are more suitable for the prognostic assessment of gliomas than the grading system. The 58-kD microspherule protein (MSP58) is an evolutionarily conserved nuclear protein and plays an important role in the regulation of cell proliferation and malignant transformation. However, whether MSP58 can be used as a biomarker to evaluate the malignancy and the prognosis of glioma patients is unknown. In the present study, we performed immunohistochemical analysis to evaluate MSP58 protein expression in 158 specimens of human gliomas and 34 normal control brain tissues. Compared with the control tissues, MSP58 expression was not only significantly higher in the glioma tissues (P < 0.05), but also increased with the increasing pathological grade (P < 0.001). Furthermore, the Kaplan-Meier analysis showed that high expression of MSP58 could predict poor survival in glioma patients (P < 0.001). In the multivariate analysis, high expression of MSP58 was also an independent unfavorable prognostic factor for the overall survival in glioma patients (P < 0.001, hazard ratio, 8.177, 95% CI 2.571-26.008). In conclusion, the increased expression of MSP58 is correlated with a higher malignant grade and poor prognosis in glioma patients. MSP58 is valuable both as an indicator of the malignancy of gliomas and as a prognostic factor for the clinical outcome of glioma patients.
Collapse
Affiliation(s)
- Wei Lin
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Liang Y, Liu M, Wang P, Ding X, Cao Y. Analysis of 20 genes at chromosome band 12q13:RACGAP1andMCRS1overexpression in nonsmall-cell lung cancer. Genes Chromosomes Cancer 2012; 52:305-15. [DOI: 10.1002/gcc.22030] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Revised: 10/16/2012] [Accepted: 10/16/2012] [Indexed: 11/11/2022] Open
|
31
|
The novel interaction between microspherule protein Msp58 and ubiquitin E3 ligase EDD regulates cell cycle progression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1833:21-32. [PMID: 23069210 DOI: 10.1016/j.bbamcr.2012.10.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Revised: 09/18/2012] [Accepted: 10/04/2012] [Indexed: 11/20/2022]
Abstract
Microspherule protein Msp58 (or MCRS1) plays a role in numerous cellular processes including transcriptional regulation and cell proliferation. It is not well understood either how Msp58 mediates its myriad functions or how it is itself regulated. Here, by immunoprecipitation, we identify EDD (E3 identified by differential display) as a novel Msp58-interacting protein. EDD, also called UBR5, is a HECT-domain (homologous to E6-AP carboxy-terminus) containing ubiquitin ligase that plays a role in cell proliferation, differentiation and DNA damage response. Both in vitro and in vivo binding assays show that Msp58 directly interacts with EDD. Microscopy studies reveal that these two proteins co-localize in the nucleus. We have also found that depletion of EDD leads to an increase of Msp58 protein level and extends the half-life of Msp58, demonstrating that EDD negatively regulates Msp58's protein stability. Furthermore, we show that Msp58 is upregulated in multiple different cell lines upon the treatment with proteasome inhibitor MG132 and exogenously expressed Msp58 is ubiquitinated, suggesting that Msp58 is degraded by the ubiquitin-proteasome pathway. Finally, knockdown of either Msp58 or EDD in human lung fibroblast WI-38 cells affects the levels of cyclins B, D and E, as well as cell cycle progression. Together, these results suggest a role for the Msp58/EDD interaction in controlling cell cycle progression. Given that both Msp58 and EDD are often aberrantly expressed in various human cancers, our findings open a new direction to elucidate Msp58 and EDD's roles in cell proliferation and tumorigenesis.
Collapse
|