1
|
Chu X, Ge S, Wu B, Zuo Y, Xu T, Yu J, Yin L, Qu H, Li J, Xiao Y, Wang Z. ASFV p30 interacts with CCAR2 and MATR3 to promote ASFV replication. Vet Microbiol 2025; 302:110416. [PMID: 39919500 DOI: 10.1016/j.vetmic.2025.110416] [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: 11/12/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/09/2025]
Abstract
African swine fever (ASF) is a highly contagious and lethal disease caused by the African swine fever virus (ASFV). Currently, effective vaccines are not available for the prevention and control of ASF. ASFV is susceptible to mutations as it has a large genome and encodes numerous proteins. In addition to evading the host immune response, ASFV utilizes host proteins to regulate its replication. The ASFV p30 protein is involved in virus internalization into the host cell and is expressed throughout the viral replication cycle, influencing viral replication. This study identified the host proteins that interact with p30 using mass spectrometry analysis. Immunoprecipitation analysis confirmed that the ASFV p30 protein interacted with the host proteins CCAR2 and MATR3, co-localizing with them in the cytoplasm. CCAR2 and MATR3 promoted ASFV replication. Conversely, ASFV infection upregulated the expression of CCAR2 and MATR3 in the host. Thus, the ASFV p30 protein regulates ASFV replication by interacting with CCAR2 and MATR3.
Collapse
Affiliation(s)
- Xuefei Chu
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China; College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Tai'an, Shandong Province 271018, China.
| | - Shengqiang Ge
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China; Qingdao Key Laboratory of Modern Bioengineering and Animal Disease Research, Qingdao 266032, China; Key Laboratory of Animal Biosafety Risk Warning Prevention and Control (South China), Ministry of Agriculture and Rural Affairs, Qingdao, Shandong 266032, China.
| | - Bingrong Wu
- College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Tai'an, Shandong Province 271018, China.
| | - Yuanyuan Zuo
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China.
| | - Tiangang Xu
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China.
| | - Jiarong Yu
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China.
| | - Lei Yin
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China.
| | - Hailong Qu
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China.
| | - Jinming Li
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China.
| | - Yihong Xiao
- College of Animal Science and Technology, Shandong Agricultural University, 61 Daizong Street, Tai'an, Shandong Province 271018, China.
| | - Zhiliang Wang
- China Animal Health and Epidemiology Center, No. 369 Nanjing Road, Qingdao 266032, China.
| |
Collapse
|
2
|
Mohan N, Johnson GS, Tovar Perez JE, Dashwood WM, Rajendran P, Dashwood RH. Alternative splicing of BAZ1A in colorectal cancer disrupts the DNA damage response and increases chemosensitization. Cell Death Dis 2024; 15:570. [PMID: 39112459 PMCID: PMC11306231 DOI: 10.1038/s41419-024-06954-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
Bromodomain Adjacent to Zinc Finger Domain 1A (BAZ1A) is a critical regulator of chromatin remodeling. We sought to clarify the roles of BAZ1A in the etiology of colorectal cancer, including the mechanisms of its alternatively spliced variants. Public databases were examined and revealed high BAZ1A expression in the majority of colorectal cancer patients, which was corroborated in a panel of human colon cancer cell lines. BAZ1A silencing reduced cell viability and increased markers of DNA damage, apoptosis, and senescence, along with the downregulation of Wnt/β-catenin signaling. The corresponding molecular changes resulted in tumor growth inhibition when BAZ1A-knockout cells were implanted into nude mice. In rescue experiments, a short isoform of BAZ1A that was associated with alternative splicing by the DBIRD complex failed to restore DNA repair activity in colon cancer cells and maintained chemosensitivity to phleomycin treatment, unlike the full-length BAZ1A. A working model proposes that a buried domain in the N-terminus of the BAZ1A short isoform lacks the ability to access linker DNA, thereby disrupting the activity of the associated chromatin remodeling complexes. Given the current interest in RNA splicing deregulation and cancer etiology, additional mechanistic studies are warranted with new lead compounds targeting BAZ1A, and other members of the BAZ family, with a view to improved therapeutic interventions.
Collapse
Affiliation(s)
- Nivedhitha Mohan
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX, USA
| | - Gavin S Johnson
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX, USA
- CRISPR Therapeutics, South Boston, MA, USA
| | | | | | - Praveen Rajendran
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX, USA.
- Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX, USA.
| | - Roderick H Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX, USA.
- Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX, USA.
| |
Collapse
|
3
|
Ai Y, Abdel-Wahab O. Splicing regulation of DNA repair via CCAR1. Mol Cell 2024; 84:2591-2592. [PMID: 39059366 DOI: 10.1016/j.molcel.2024.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 06/12/2024] [Accepted: 06/12/2024] [Indexed: 07/28/2024]
Abstract
In this issue of Molecular Cell, Harada et al.1 and Karasu et al.2 identify CCAR1 as a novel regulator of the Fanconi anemia/BRCA DNA repair pathway via modulating the splicing of the mRNA encoding FANCA.
Collapse
Affiliation(s)
- Yuxi Ai
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar Abdel-Wahab
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
4
|
Karasu ME, Jahnke L, Joseph BJ, Amerzhanova Y, Mironov A, Shu X, Schröder MS, Gvozdenovic A, Sala I, Zavolan M, Jonas S, Corn JE. CCAR1 promotes DNA repair via alternative splicing. Mol Cell 2024; 84:2634-2647.e9. [PMID: 38964321 DOI: 10.1016/j.molcel.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 04/29/2024] [Accepted: 06/12/2024] [Indexed: 07/06/2024]
Abstract
DNA repair is directly performed by hundreds of core factors and indirectly regulated by thousands of others. We massively expanded a CRISPR inhibition and Cas9-editing screening system to discover factors indirectly modulating homology-directed repair (HDR) in the context of ∼18,000 individual gene knockdowns. We focused on CCAR1, a poorly understood gene that we found the depletion of reduced both HDR and interstrand crosslink repair, phenocopying the loss of the Fanconi anemia pathway. CCAR1 loss abrogated FANCA protein without substantial reduction in the level of its mRNA or that of other FA genes. We instead found that CCAR1 prevents inclusion of a poison exon in FANCA. Transcriptomic analysis revealed that the CCAR1 splicing modulatory activity is not limited to FANCA, and it instead regulates widespread changes in alternative splicing that would damage coding sequences in mouse and human cells. CCAR1 therefore has an unanticipated function as a splicing fidelity factor.
Collapse
Affiliation(s)
- Mehmet E Karasu
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich 8093, Switzerland
| | - Leonard Jahnke
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich 8093, Switzerland
| | - Brian J Joseph
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA
| | - Yerkezhan Amerzhanova
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich 8093, Switzerland
| | - Aleksei Mironov
- Computational and Systems Biology, Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Xuan Shu
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich 8093, Switzerland
| | - Markus S Schröder
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich 8093, Switzerland
| | - Ana Gvozdenovic
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich 8093, Switzerland
| | - Irene Sala
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich 8093, Switzerland
| | - Mihaela Zavolan
- Computational and Systems Biology, Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Stefanie Jonas
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich 8093, Switzerland
| | - Jacob E Corn
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich 8093, Switzerland.
| |
Collapse
|
5
|
Harada N, Asada S, Jiang L, Nguyen H, Moreau L, Marina RJ, Adelman K, Iyer DR, D'Andrea AD. The splicing factor CCAR1 regulates the Fanconi anemia/BRCA pathway. Mol Cell 2024; 84:2618-2633.e10. [PMID: 39025073 PMCID: PMC11321822 DOI: 10.1016/j.molcel.2024.06.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 05/15/2024] [Accepted: 06/25/2024] [Indexed: 07/20/2024]
Abstract
The twenty-three Fanconi anemia (FA) proteins cooperate in the FA/BRCA pathway to repair DNA interstrand cross-links (ICLs). The cell division cycle and apoptosis regulator 1 (CCAR1) protein is also a regulator of ICL repair, though its possible function in the FA/BRCA pathway remains unknown. Here, we demonstrate that CCAR1 plays a unique upstream role in the FA/BRCA pathway and is required for FANCA protein expression in human cells. Interestingly, CCAR1 co-immunoprecipitates with FANCA pre-mRNA and is required for FANCA mRNA processing. Loss of CCAR1 results in retention of a poison exon in the FANCA transcript, thereby leading to reduced FANCA protein expression. A unique domain of CCAR1, the EF hand domain, is required for interaction with the U2AF heterodimer of the spliceosome and for excision of the poison exon. Taken together, CCAR1 is a splicing modulator required for normal splicing of the FANCA mRNA and other mRNAs involved in various cellular pathways.
Collapse
Affiliation(s)
- Naoya Harada
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Shuhei Asada
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Lige Jiang
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Huy Nguyen
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Lisa Moreau
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Ryan J Marina
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Karen Adelman
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Divya R Iyer
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Alan D D'Andrea
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
6
|
Venkatesh J, Muthu M, Singaravelu I, Cheriyan VT, Sekhar SC, Acharige NCPN, Levi E, Assad H, Pflum MKH, Rishi AK. Phosphorylation of cell cycle and apoptosis regulatory protein-1 by stress activated protein kinase P38γ is a novel mechanism of apoptosis signaling by genotoxic chemotherapy. Front Oncol 2024; 14:1376666. [PMID: 38756656 PMCID: PMC11096501 DOI: 10.3389/fonc.2024.1376666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/08/2024] [Indexed: 05/18/2024] Open
Abstract
CARP-1, a perinuclear phospho-protein, regulates cell survival and apoptosis signaling induced by genotoxic drugs. However, kinase(s) phosphorylating CARP-1 and down-stream signal transduction events remain unclear. Here we find that CARP-1 Serine (S)626 and Threonine (T)627 substitution to Alanines (AA) inhibits genotoxic drug-induced apoptosis. CARP-1 T627 is followed by a Proline (P), and this TP motif is conserved in vertebrates. Based on these findings, we generated affinity-purified, anti-phospho-CARP-1 T627 rabbit polyclonal antibodies, and utilized them to elucidate chemotherapy-activated, CARP-1-dependent cell growth signaling mechanisms. Our kinase profiling studies revealed that MAPKs/SAPKs phosphorylated CARP-1 T627. We then UV cross-linked protein extracts from Adriamycin-treated HeLa cervical cancer cells with a CARP-1 (614-638) peptide, and conducted liquid chromatography-tandem mass spectrometry (LC-MS/MS) analyses of the peptide-bound protein complexes. This experiment revealed SAPK p38γ interaction with CARP-1 (614-638) peptide. Our studies further established that SAPK p38γ, but not other MAPKs, phosphorylates CARP-1 T627 in cancer cells treated with genotoxic drugs. Loss of p38γ abrogates CARP-1 T627 phosphorylation, and results in enhanced survival of breast cancer cells by genotoxic drugs. CARP-1 T627 phosphorylation was also noted in breast tumors from patients treated with radiation or endocrine therapies. We conclude that genotoxic drugs activate p38γ-dependent CARP-1 T627 phosphorylation to inhibit cell growth.
Collapse
Affiliation(s)
- Jaganathan Venkatesh
- John D. Dingell V.A. Medical Center, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
- Department of Oncology, Wayne State University, Detroit, MI, United States
| | - Magesh Muthu
- John D. Dingell V.A. Medical Center, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
- Department of Oncology, Wayne State University, Detroit, MI, United States
| | - Indulekha Singaravelu
- John D. Dingell V.A. Medical Center, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
- Department of Oncology, Wayne State University, Detroit, MI, United States
| | - Vino T. Cheriyan
- John D. Dingell V.A. Medical Center, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
- Department of Oncology, Wayne State University, Detroit, MI, United States
| | - Sreeja C. Sekhar
- John D. Dingell V.A. Medical Center, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
- Department of Oncology, Wayne State University, Detroit, MI, United States
| | | | - Edi Levi
- John D. Dingell V.A. Medical Center, Wayne State University, Detroit, MI, United States
- Department of Pathology, Wayne State University, Detroit, MI, United States
| | - Hadeel Assad
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
- Department of Oncology, Wayne State University, Detroit, MI, United States
| | - Mary Kay H. Pflum
- Department of Chemistry, Wayne State University, Detroit, MI, United States
| | - Arun K. Rishi
- John D. Dingell V.A. Medical Center, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
- Department of Oncology, Wayne State University, Detroit, MI, United States
| |
Collapse
|
7
|
Yang N, Zhang N, Lu G, Zeng S, Xing Y, Du L. RNA-binding proteins potentially regulate the alternative splicing of cell cycle-associated genes in proliferative diabetic retinopathy. Sci Rep 2024; 14:6731. [PMID: 38509306 PMCID: PMC10954754 DOI: 10.1038/s41598-024-57516-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 03/19/2024] [Indexed: 03/22/2024] Open
Abstract
RNA-binding proteins (RBPs) contribute to the pathogenesis of proliferative diabetic retinopathy (PDR) by regulating gene expression through alternative splicing events (ASEs). However, the RBPs differentially expressed in PDR and the underlying mechanisms remain unclear. Thus, this study aimed to identify the differentially expressed genes in the neovascular membranes (NVM) and retinas of patients with PDR. The public transcriptome dataset GSE102485 was downloaded from the Gene Expression Omnibus database, and samples of PDR and normal retinas were analyzed. A mouse model of oxygen-induced retinopathy was used to confirm the results. The top 20 RBPs were screened for co-expression with alternative splicing genes (ASGs). A total of 403 RBPs were abnormally expressed in the NVM and retina samples. Functional analysis demonstrated that the ASGs were enriched in cell cycle pathways. Cell cycle-associated ASEs and an RBP-AS regulatory network, including 15 RBPs and their regulated ASGs, were extracted. Splicing factor proline/glutamine rich (SFPQ), microtubule-associated protein 1 B (MAP1B), heat-shock protein 90-alpha (HSP90AA1), microtubule-actin crosslinking factor 1 (MACF1), and CyclinH (CCNH) expression remarkably differed in the mouse model. This study provides novel insights into the RBP-AS interaction network in PDR and for developing screening and treatment options to prevent diabetic retinopathy-related blindness.
Collapse
Affiliation(s)
- Ning Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ningzhi Zhang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guojing Lu
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Siyu Zeng
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yiqiao Xing
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Du
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
8
|
Lugano D, Barrett L, Westerheide SD, Kee Y. Multifaceted roles of CCAR family proteins in the DNA damage response and cancer. Exp Mol Med 2024; 56:59-65. [PMID: 38172598 PMCID: PMC10834508 DOI: 10.1038/s12276-023-01139-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/18/2023] [Accepted: 10/09/2023] [Indexed: 01/05/2024] Open
Abstract
The cell cycle apoptosis regulator (CCAR) family of proteins consists of two proteins, CCAR1 and CCAR2, that play a variety of roles in cellular physiology and pathology. These multidomain proteins are able to perform multiple interactions and functions, playing roles in processes such as stress responses, metabolism, and the DNA damage response. The evolutionary conservation of CCAR family proteins allows their study in model organisms such as Caenorhabditis elegans, where a role for CCAR in aging was revealed. This review particularly highlights the multifaceted roles of CCAR family proteins and their implications in the DNA damage response and in cancer biology.
Collapse
Affiliation(s)
- D Lugano
- Department of Molecular Biosciences, College of Arts and Sciences, University of South Florida, Tampa, FL, 33647, USA
| | - L Barrett
- Department of Molecular Biosciences, College of Arts and Sciences, University of South Florida, Tampa, FL, 33647, USA
| | - S D Westerheide
- Department of Molecular Biosciences, College of Arts and Sciences, University of South Florida, Tampa, FL, 33647, USA
| | - Y Kee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno-Joongang-daero, Dalseong-gun, Daegu, 42988, Republic of Korea.
| |
Collapse
|
9
|
Norouzi Kamareh M, Samadi M, Arabzadeh E, Abdollahi M, Sheidaei S, Riyahi Malayeri S, Schlicht J, Shirvani H, Rostamkhani F. The effect of 24-hour sleep deprivation and anaerobic exercise on the expression of BAX, BCL2, BMAL1 and CCAR2 genes in peripheral blood mononuclear cells after L-arginine supplementation. Gene 2023; 887:147732. [PMID: 37625565 DOI: 10.1016/j.gene.2023.147732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 06/26/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
Sleep deprivation disrupt the circadian clock and exercise performance. Defective oxidative stress caused by sleep deprivation may affect the expression of genes involved in cell apoptosis. Since a number of studies have shown the anti-apoptotic effect of L-arginine, so the aim of this study was to evaluate the effect of eight weeks of L-arginine supplementation on the expression of brain and muscle ARNT-like protein 1 (BMAL1), cell cycle and apoptosis regulator 2 (CCAR2), and BAX and BCL2 genes during sleep deprivation and acute anaerobic exercise. Participants included 20 healthy men age 26-35 years, randomized into the L-arginine intervention group (n = 10) and a placebo control (n = 10). The running-based anaerobic sprint test (RAST) was used for anaerobic exercise. Intervention subjects took one 1000 mg L-arginine tablet daily for 8 weeks. The Real-Time PCR method was used to determine apoptosis gene expression in peripheral blood mononuclear cells (PBMCs). Acute anaerobic exercise and sleep deprivation both increased the expression of BAX and CCAR2 genes, and decreased the expression of BCL2 and BMAL1 genes (p < 0.05 for all). L-arginine supplementation increased the expression of BMAL1 and BCL2 genes and decreased the expression of BAX and CCAR2 genes relative to control (p < 0.05). L-Arginine controlled the increase in expression of BAX and CCAR2 genes and the decrease in expression of BCL2 and BMAL1 genes in response to sleep deprivation and acute anaerobic exercise (p < 0.05). Our results showed that 24-hour sleep deprivation and acute anaerobic exercise increased the expression of pro-apoptotic genes (BAX and CCAR2) and decreased the expression of anti-apoptotic genes (BCL2 and BMAL1), although the effect of sleep deprivation is greater. In this situation, L-arginine supplementation may balance the apoptotic state of peripheral blood mononuclear cells. However, any recommendation about this needs further research.
Collapse
Affiliation(s)
- Mirzahossein Norouzi Kamareh
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Samadi
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ehsan Arabzadeh
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahkameh Abdollahi
- Department of Physical Education and Sport Sciences, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Sadra Sheidaei
- Department of Physical Education and Sport Sciences, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Shahin Riyahi Malayeri
- Department of Physical Education and Sport Sciences, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Jeffrey Schlicht
- Department of Health Promotion and Exercise Sciences, Western Connecticut State University, Danbury, CT 06810, USA
| | - Hossein Shirvani
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Rostamkhani
- Department of Biology, Yadegar-e-Imam Khomeini (RAH) Shahre Rey Branch, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
10
|
Kim HJ, Moon SJ, Kim JH. Mechanistic insights into the dual role of CCAR2/DBC1 in cancer. Exp Mol Med 2023; 55:1691-1701. [PMID: 37524873 PMCID: PMC10474295 DOI: 10.1038/s12276-023-01058-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/21/2023] [Accepted: 05/17/2023] [Indexed: 08/02/2023] Open
Abstract
Cell cycle and apoptosis regulator 2 (CCAR2), also known as deleted in breast cancer 1 (DBC1), has been recently identified as a master regulator of transcriptional processes and plays diverse roles in physiology and pathophysiology, including as a regulator of apoptosis, DNA repair, metabolism, and tumorigenesis. CCAR2 functions as a coregulator of various transcription factors and a critical regulator of numerous epigenetic modifiers. Based on its ability to stimulate apoptosis by activating and stabilizing p53, CCAR2 was initially considered to be a tumor suppressor. However, an increasing number of studies have shown that CCAR2 also functions as a tumor-promoting coregulator by activating oncogenic transcription factors and regulating the enzymatic activity of epigenetic modifiers, indicating that CCAR2 may play a dual role in cancer progression by acting as a tumor suppressor and tumor promoter. Here, we review recent progress in understanding the dual tumor-suppressing and oncogenic roles of CCAR2 in cancer. We discuss CCAR2 domain structures, its interaction partners, and the molecular mechanisms by which it regulates the activities of transcription factors and epigenetic modifiers.
Collapse
Affiliation(s)
- Hwa Jin Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea
| | - Sue Jin Moon
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea
| | - Jeong Hoon Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea.
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea.
| |
Collapse
|
11
|
He S, Zhu H, Zhang J, Yang X, Zhao L. Genome-wide screening for circRNAs in epicardial adipose tissue of heart failure patients with preserved ejection fraction. Am J Transl Res 2023; 15:4610-4619. [PMID: 37560213 PMCID: PMC10408519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/01/2023] [Indexed: 08/11/2023]
Abstract
OBJECTIVE Heart failure with preserved ejection fraction (HFpEF) is a complex cardiovascular syndrome. Along with pro-inflammatory and metabolic factors, epicardial adipose tissue (EAT) is believed to play a key role in the pathogenesis of HFpEF. Studies have increasingly shown a critical role of circRNAs in the development of cardiovascular diseases; however, their role in the pathogenetic mechanism of HFpEF is not well characterized. The objective of this study was to investigate the expression profiles of circRNAs in EAT of HFpEF patients. METHODS Samples of epicardial adipose tissue were obtained from patients with HFpEF (n=5) and patients without heart failure (non-HF; n=5). CircRNA expression profiles were screened using RNA sequencing method. RNA-sequencing results were confirmed by qRT-PCR analysis. Gene Ontology enrichment and Kyoto Encyclopedia of Genes and Genomes pathway analysis were performed on the differentially expressed circRNAs. RESULTS A total of 131 circRNAs were differentially expressed between HFpEF and non-HF groups (77 upregulated and 54 downregulated). Among these, hsa_circ_0118464 corresponding to HECW2 gene which showed the highest fold-change was assessed by qRT-PCR, and the outcome was consistent with RNA-sequencing results. The differentially expressed circRNAs corresponded to genes mainly involved in regulation of cellular and metabolic processes. CONCLUSION This study provides the expression profile of circRNAs in EAT of HFpEF patients and the associated molecular mechanism. Our findings may provide insight into diagnostic markers and therapeutic targets in the context of HFpEF.
Collapse
Affiliation(s)
- Shan He
- Heart Center, Beijing Chaoyang Hospital Jingxi Branch, Capital Medical UniversityBeijing 100043, China
| | - Huagang Zhu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical UniversityBeijing 100029, China
| | - Jianjun Zhang
- Heart Center, Beijing Chaoyang Hospital Jingxi Branch, Capital Medical UniversityBeijing 100043, China
| | - Xinchun Yang
- Heart Center, Beijing Chaoyang Hospital, Capital Medical UniversityBeijing 100020, China
| | - Lei Zhao
- Heart Center, Beijing Chaoyang Hospital, Capital Medical UniversityBeijing 100020, China
| |
Collapse
|
12
|
Targeting ACE2-BRD4 crosstalk in colorectal cancer and the deregulation of DNA repair and apoptosis. NPJ Precis Oncol 2023; 7:20. [PMID: 36801948 PMCID: PMC9938505 DOI: 10.1038/s41698-023-00361-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 02/10/2023] [Indexed: 02/20/2023] Open
Abstract
ACE2 overexpression in colorectal cancer patients might increase susceptibility to SARS-CoV-2 infection. We report that knockdown, forced overexpression, and pharmacologic inhibition in human colon cancer cells targeted ACE2-BRD4 crosstalk to mediate marked changes in DNA damage/repair and apoptosis. In colorectal cancer patients for whom high ACE2 plus high BRD4 expression is predictive of poor survival, pan-BET inhibition would need to consider proviral/antiviral actions of different BET proteins during SARS-CoV-2 infection.
Collapse
|
13
|
Brane A, Arora I, Tollefsbol TO. Peripubertal Nutritional Prevention of Cancer-Associated Gene Expression and Phenotypes. Cancers (Basel) 2023; 15:674. [PMID: 36765634 PMCID: PMC9913820 DOI: 10.3390/cancers15030674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/25/2023] Open
Abstract
Breast cancer (BC) is a nearly ubiquitous malignancy that effects the lives of millions worldwide. Recently, nutritional prevention of BC has received increased attention due to its efficacy and ease of application. Chief among chemopreventive compounds are plant-based substances known as dietary phytochemicals. Sulforaphane (SFN), an epigenetically active phytochemical found in cruciferous vegetables, has shown promise in BC prevention. In addition, observational studies suggest that the life stage of phytochemical consumption may influence its anticancer properties. These life stages, called critical periods (CPs), are associated with rapid development and increased susceptibility to cellular damage. Puberty, a CP in which female breast tissue undergoes proliferation and differentiation, is of particular interest for later-life BC development. However, little is known about the importance of nutritional chemoprevention to CPs. We sought to address this by utilizing two estrogen receptor-negative [ER(-)] transgenic mouse models fed SFN-containing broccoli sprout extract during the critical period of puberty. We found that this treatment resulted in a significant decrease in tumor incidence and weight, as well as an increase in tumor latency. Further, we found significant alterations in the long-term expression of cancer-associated genes, including p21, p53, and BRCA2. Additionally, our transcriptomic analyses identified expressional changes in many cancer-associated genes, and bisulfite sequencing revealed that the antiproliferation-associated gene Erich4 was both hypomethylated and overexpressed in our experimental group. Our study indicates that dietary interventions during the CP of puberty may be important for later-life ER(-) BC prevention and highlights potential important genetic and epigenetic targets for treatment and study of the more deadly variants of BC.
Collapse
Affiliation(s)
- Andrew Brane
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Itika Arora
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Trygve O. Tollefsbol
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- University Wide Microbiome Center, University of Alabama Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
14
|
Bader JM, Deigendesch N, Misch M, Mann M, Koch A, Meissner F. Proteomics separates adult-type diffuse high-grade gliomas in metabolic subgroups independent of 1p/19q codeletion and across IDH mutational status. Cell Rep Med 2023; 4:100877. [PMID: 36584682 PMCID: PMC9873829 DOI: 10.1016/j.xcrm.2022.100877] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 07/15/2022] [Accepted: 12/07/2022] [Indexed: 12/30/2022]
Abstract
High-grade adult-type diffuse gliomas are malignant neuroepithelial tumors with poor survival rates in combined chemoradiotherapy. The current WHO classification is based on IDH1/2 mutational and 1p/19q codeletion status. Glioma proteome alterations remain undercharacterized despite their promise for a better molecular patient stratification and therapeutic target identification. Here, we use mass spectrometry to characterize 42 formalin-fixed, paraffin-embedded (FFPE) samples from IDH-wild-type (IDHwt) gliomas, IDH-mutant (IDHmut) gliomas with and without 1p/19q codeletion, and non-neoplastic controls. Based on more than 5,500 quantified proteins and 5,000 phosphosites, gliomas separate by IDH1/2 mutational status but not by 1p/19q status. Instead, IDHmut gliomas split into two proteomic subtypes with widespread perturbations, including aerobic/anaerobic energy metabolism. Validations with three independent glioma proteome datasets confirm these subgroups and link the IDHmut subtypes to the established proneural and classic/mesenchymal subtypes in IDHwt glioma. This demonstrates common phenotypic subtypes across the IDH status with potential therapeutic implications for patients with IDHmut gliomas.
Collapse
Affiliation(s)
- Jakob Maximilian Bader
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Nikolaus Deigendesch
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Martin Misch
- Department of Neurosurgery, Charité, Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Arend Koch
- Department of Neuropathology, Charité, Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany.
| | - Felix Meissner
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; Department of Systems Immunology and Proteomics, Institute of Innate Immunity, University Hospital Bonn, 53127 Bonn, Germany.
| |
Collapse
|
15
|
CCAR2 functions downstream of the Shieldin complex to promote double-strand break end-joining. Proc Natl Acad Sci U S A 2022; 119:e2214935119. [PMID: 36442094 PMCID: PMC9894118 DOI: 10.1073/pnas.2214935119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The 53BP1-RIF1 pathway restricts the resection of DNA double-strand breaks (DSBs) and promotes blunt end-ligation by non-homologous end joining (NHEJ) repair. The Shieldin complex is a downstream effector of the 53BP1-RIF1 pathway. Here, we identify a component of this pathway, CCAR2/DBC1, which is also required for restriction of DNA end-resection. CCAR2 co-immunoprecipitates with the Shieldin complex, and knockout of CCAR2 in a BRCA1-deficient cell line results in elevated DSB end-resection, RAD51 loading, and PARP inhibitor (PARPi) resistance. Knockout of CCAR2 is epistatic with knockout of other Shieldin proteins. The S1-like RNA-binding domain of CCAR2 is required for its interaction with the Shieldin complex and for suppression of DSB end-resection. CCAR2 functions downstream of the Shieldin complex, and CCAR2 knockout cells have delayed resolution of Shieldin complex foci. Forkhead-associated (FHA)-dependent targeting of CCAR2 to DSB sites re-sensitized BRCA1-/-SHLD2-/- cells to PARPi. Taken together, CCAR2 is a functional component of the 53BP1-RIF1 pathway, promotes the refill of resected DSBs, and suppresses homologous recombination.
Collapse
|
16
|
Okpara MO, Hermann C, van der Watt PJ, Garnett S, Blackburn JM, Leaner VD. A mass spectrometry-based approach for the identification of Kpnβ1 binding partners in cancer cells. Sci Rep 2022; 12:20171. [PMID: 36418423 PMCID: PMC9684564 DOI: 10.1038/s41598-022-24194-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 11/11/2022] [Indexed: 11/25/2022] Open
Abstract
Karyopherin beta 1 (Kpnβ1) is the principal nuclear importer of cargo proteins and plays a role in many cellular processes. Its expression is upregulated in cancer and essential for cancer cell viability, thus the identification of its binding partners might help in the discovery of anti-cancer therapeutic targets and cancer biomarkers. Herein, we applied immunoprecipitation coupled to mass spectrometry (IP-MS) to identify Kpnβ1 binding partners in normal and cancer cells. IP-MS identified 100 potential Kpnβ1 binding partners in non-cancer hTERT-RPE1, 179 in HeLa cervical cancer, 147 in WHCO5 oesophageal cancer and 176 in KYSE30 oesophageal cancer cells, including expected and novel interaction partners. 38 binding proteins were identified in all cell lines, with the majority involved in RNA metabolism. 18 binding proteins were unique to the cancer cells, with many involved in protein translation. Western blot analysis validated the interaction of known and novel binding partners with Kpnβ1 and revealed enriched interactions between Kpnβ1 and select proteins in cancer cells, including proteins involved in cancer development, such as Kpnα2, Ran, CRM1, CCAR1 and FUBP1. Together, this study shows that Kpnβ1 interacts with numerous proteins, and its enhanced interaction with certain proteins in cancer cells likely contributes to the cancer state.
Collapse
Affiliation(s)
- Michael O. Okpara
- grid.7836.a0000 0004 1937 1151Division of Medical Biochemistry and Structural Biology, University of Cape Town, Cape Town, South Africa
| | - Clemens Hermann
- grid.7836.a0000 0004 1937 1151Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Pauline J. van der Watt
- grid.7836.a0000 0004 1937 1151Division of Medical Biochemistry and Structural Biology, University of Cape Town, Cape Town, South Africa ,grid.7836.a0000 0004 1937 1151Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Shaun Garnett
- grid.7836.a0000 0004 1937 1151Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jonathan M. Blackburn
- grid.7836.a0000 0004 1937 1151Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa ,grid.7836.a0000 0004 1937 1151Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Virna D. Leaner
- grid.7836.a0000 0004 1937 1151Division of Medical Biochemistry and Structural Biology, University of Cape Town, Cape Town, South Africa ,grid.7836.a0000 0004 1937 1151SAMRC Gynaecology Cancer Research Centre, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
17
|
Xiao R, Yuan Y, Xia H, Ge Q, Chen L, Zhu F, Xu J, Wang X, Fan Y, Wang Q, Yang Y, Chen K. Comparative transcriptome and proteome reveal synergistic functions of differentially expressed genes and proteins implicated in an over-dominant silkworm heterosis of increased silk yield. INSECT MOLECULAR BIOLOGY 2022; 31:551-567. [PMID: 35445454 DOI: 10.1111/imb.12779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/09/2022] [Accepted: 04/14/2022] [Indexed: 06/14/2023]
Abstract
We previously observed an over-dominant silkworm heterosis of increased yield in a cross of Bombyx mori nuclear polyhydrosis virus-resistant strain NB with a susceptible strain 306. In the present study, we found that heterosis also exists in crosses of NB with other susceptible strains, indicating it is a more general phenomenon. We performed comparative transcriptome and proteome and identified 1624 differentially expressed genes (DEGs) and 298 differentially expressed proteins (DEPs) in silk glands between parents and F1 hybrids, of which 24 DEGs/DEPs showed consistent expression at mRNA and protein levels revealed by Venn joint analysis. Their expressions are completely non-additive, mainly transgressive and under low-parent, suggesting recombination of parental genomes may be the major genetic mechanism for the heterosis. GO and KEGG analyses revealed that they may function in generally similar but distinctive aspects of metabolisms and processes with signal transduction and translation being most affected. Notably, they may not only up-regulate biosynthesis and transport of silk proteins but also down-regulate other unrelated processes, synergistically and globally remodelling the silk gland to increase yield and cause the heterosis. Our findings contribute insights into the understanding of silkworm heterosis and silk gland development and provide targets for transgenic manipulation to further increase the silk yield.
Collapse
Affiliation(s)
- Rui Xiao
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yi Yuan
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Hengchuan Xia
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Qi Ge
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Liang Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Feifei Zhu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jia Xu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xueqi Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yixuan Fan
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Qiang Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yanhua Yang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Keping Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
18
|
Proteomic profiling reveals neuronal ion channel dysregulation and cellular responses to DNA damage-induced cell cycle arrest and senescence in human neuroblastoma SH-SY5Y cells exposed to cypermethrin. Neurotoxicology 2022; 93:71-83. [PMID: 36063984 DOI: 10.1016/j.neuro.2022.08.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/27/2022] [Accepted: 08/28/2022] [Indexed: 11/20/2022]
Abstract
Cypermethrin (CYP), a synthetic pyrethroid of class II, is widely used as a pesticide worldwide. The primary target of cypermethrin is a voltage-gated sodium channel. The neurotoxicity of CYP has been extensively studied in terms of affecting neuronal development, increasing cellular oxidative stress, and apoptosis. However, little is known about how it affects the expression of channel proteins involved in synaptic transmission, as well as the effects of cypermethrin on DNA damage and cell cycle processes. We found that the ligand and voltage-gated calcium channels and proteins involved in synaptic transmission including NMDA 1 receptor subunit, alpha 1A-voltage-dependent calcium channel, synaptotagmin-17, and synaptojanin-2 were downregulated in CYP-treated cells. After 48h of CYP exposure, cell viability was reduced with flattened and enlarged morphology. The levels of 23 proteins regulating cell cycle processes were altered in CYP-treated cells, according to a proteomic study. The cell cycle analysis showed elevated G0/G1 cell cycle arrest and DNA fragmentation at the sub-G0 stage after CYP exposure. CYP treatment also increased senescence-associated β-galactosidase positive cells, DNA damage, and apoptotic markers. Taken together, the current study showed that cypermethrin exposure caused DNA damage and hastened cellular senescence and apoptosis via disrupting cell cycle regulation. In addition, despite its primary target sodium channel, CYP might cause synaptic dysfunction via the downregulation of synaptic proteins and dysregulation of synapse-associated ion channels.
Collapse
|
19
|
Kim HJ, Moon SJ, Hong S, Won HH, Kim JH. DBC1 is a key positive regulator of enhancer epigenomic writers KMT2D and p300. Nucleic Acids Res 2022; 50:7873-7888. [PMID: 35801925 PMCID: PMC9371912 DOI: 10.1093/nar/gkac585] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/14/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Histone modification is a key epigenetic mechanism for regulation of chromatin dynamics and gene expression. Deleted in breast cancer 1 (DBC1) has been shown to act as a negative regulator of epigenetic modifiers and as a co-activator for nuclear receptors and other transcription factors. However, little is known about the role of DBC1 in the regulation of histone modifications and chromatin landscapes. Here, we analyzed genome-wide profiles of active enhancer and promoter marks in colorectal cancer cells and report DBC1 as a critical positive regulator of histone epigenetic writers KMT2D (H3K4 methyltransferase) and p300 (histone acetyltransferase). DBC1 is required for establishing the landscape of active enhancers, for genome-wide chromatin binding and enhancer recruitment of KMT2D and p300, and for gene activation involved in colorectal cancer progression. DBC1 interacts directly with KMT2D and p300, and enhances KMT2D-mediated histone H3K4 methylation (H3K4me1/2/3) and p300-mediated H3 acetylation. Importantly, DBC1 contributes to super-enhancer formation and function by facilitating the recruitment of KMT2D and p300 and by enhancing their functional interaction and cooperative cross-talk. Our results highlight the critical role of DBC1 as a key positive regulator of KMT2D and p300, and provide insights into regulatory mechanisms underlying the interplay between the enhancer epigenomic writers in enhancer activation.
Collapse
Affiliation(s)
- Hwa Jin Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea.,Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea
| | - Sue Jin Moon
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea.,Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea
| | - Sanghoon Hong
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea
| | - Hong-Hee Won
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea.,Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea
| | - Jeong Hoon Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06351, South Korea.,Research Institute for Future Medicine, Samsung Medical Center, Seoul, 06351, South Korea
| |
Collapse
|
20
|
Gao X, Tang Y, Ma Y. Bone Marrow Mesenchymal Stem Cells (BMSCs)-Triggered Up-Regulation of miR-198 Impedes the Aggressive Migration and Invasion of Cervical Cancer Cells. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
As a subset of RNAs without protein-coding function, short non-coding RNAs (microRNAs) are reported to contribute to the progress of multiple disorders. Nevertheless, the precise function of miR-198 in human cervical cancer is still an open question. RNA sequencing between cervical
cancer cell lines and normal cervical epithelial cells identified CCAR1 to be highly expressed in cervical cancer. Cells were transfected with si-CCAR1 followed by analysis of cell behaviors using clonogenic assay and transwell migrating assay. The binding of miR-198 and CCAR1 was verified
by a dual-luciferase reporter gene experiment. CCAR1 was highly expressed in cervical cancer tissues and cell lines and associated with tumor staging. Knockdown of CCAR1 restrained the malignant phenotypes of cervical cancer cells. CCAR1 was a target of miR198. Co-culture with BMSCs upregulated
miR-198 expression, resulting in impediment of the aggressive phenotypes of cervical cancer cells, which was mediated by suppression of CCAR1 and release of inflammatory factors. In conclusion, CCAR1 level is increased in cervical cancer tissues or cell lines. Co-culture of BMSCs can facilitate
the proliferating, migrating and invading activities of cervical cancer cells but reduce the release of inflammatory factors which is possibly through manipulating the axis of miR-198/CCAR1.
Collapse
Affiliation(s)
- Xueying Gao
- Department of Obstetrics, Changyi People’s Hospital, Shandong Province, Changyi, Shandong, 261300, China
| | - Ying Tang
- Department of Obstetrics, Changyi People’s Hospital, Shandong Province, Changyi, Shandong, 261300, China
| | - Yunping Ma
- Department of Obstetrics, Changyi People’s Hospital, Shandong Province, Changyi, Shandong, 261300, China
| |
Collapse
|
21
|
Turnier JL, Kahlenberg JM. Using autoantibody signatures to define cancer risk in dermatomyositis. J Clin Invest 2022; 132:e156025. [PMID: 35040442 PMCID: PMC8759773 DOI: 10.1172/jci156025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Dermatomyositis is an idiopathic inflammatory myopathy with a highly heterogeneous disease course. Although there is a known increase in cancer risk surrounding the time of dermatomyositis diagnosis, the mechanisms driving this increased risk are not well understood. Further, there are no current standardized cancer screening guidelines for dermatomyositis patients. In this issue of the JCI, Fiorentino, Mecoli, et al. discovered additional autoantibodies in patients with dermatomyositis and anti-TIF1-γ autoantibodies, a known risk factor for malignancy. They observed a decreased cancer risk with an increasing number of autoantibodies. Importantly, these findings indicate that more detailed autoantibody phenotyping at diagnosis might better predict cancer risk and also suggest that diversity and kinetics of the host immune response might influence cancer development.
Collapse
Affiliation(s)
| | - J. Michelle Kahlenberg
- Department of Internal Medicine, Division of Rheumatology
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
22
|
CCAR2 promotes a malignant phenotype of osteosarcoma through Wnt/β-catenin-dependent transcriptional activation of SPARC. Biochem Biophys Res Commun 2021; 580:67-73. [PMID: 34624572 DOI: 10.1016/j.bbrc.2021.09.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/21/2021] [Accepted: 09/26/2021] [Indexed: 11/22/2022]
Abstract
CCAR2 plays a pivotal role in the regulation of the DNA damage response and cancer progression. Although aberrant expression of CCAR2 has been reported in several types of cancer, its biological function and molecular mechanism in osteosarcoma (OS) have not yet been fully elucidated. Here, we show that silence of CCAR2 prevented the malignant phenotype of OS cell in vitro and decreased tumor growth in nude mice. By analyzing the transcriptomic profile of CCAR2 knockdown U2OS cells, we identified secreted protein acidic and rich in cysteine (SPARC) is tightly regulated by CCAR2. Mechanically, we found that SPARC is transcriptionally regulated by Wnt/β-catenin signaling, and CCAR2 acts as a co-activator of Wnt/β-catenin signaling to regulate the expression of SPARC in OS cells. Additionally, SPARC knockdown largely eliminated the malignant phenotype induced by CCAR2 overexpression and forced expression of SPARC promoted the malignant phenotype of CCAR2-depleted cells. In conclusion, our results suggest that CCAR2 exerted oncogenic roles in OS cells mainly via up-regulating SPARC expression and targeting the CCAR2-SPARC axis might have promising application prospect for the treatment of osteosarcoma.
Collapse
|
23
|
Vadivel Gnanasundram S, Bonczek O, Wang L, Chen S, Fahraeus R. p53 mRNA Metabolism Links with the DNA Damage Response. Genes (Basel) 2021; 12:1446. [PMID: 34573428 PMCID: PMC8465283 DOI: 10.3390/genes12091446] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 12/14/2022] Open
Abstract
Human cells are subjected to continuous challenges by different genotoxic stress attacks. DNA damage leads to erroneous mutations, which can alter the function of oncogenes or tumor suppressors, resulting in cancer development. To circumvent this, cells activate the DNA damage response (DDR), which mainly involves cell cycle regulation and DNA repair processes. The tumor suppressor p53 plays a pivotal role in the DDR by halting the cell cycle and facilitating the DNA repair processes. Various pathways and factors participating in the detection and repair of DNA have been described, including scores of RNA-binding proteins (RBPs) and RNAs. It has become increasingly clear that p53's role is multitasking, and p53 mRNA regulation plays a prominent part in the DDR. This review is aimed at covering the p53 RNA metabolism linked to the DDR and highlights the recent findings.
Collapse
Affiliation(s)
| | - Ondrej Bonczek
- Department of Medical Biosciences, Umeå University, 901-87 Umeå, Sweden; (O.B.); (L.W.); (S.C.)
- RECAMO, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656-53 Brno, Czech Republic
| | - Lixiao Wang
- Department of Medical Biosciences, Umeå University, 901-87 Umeå, Sweden; (O.B.); (L.W.); (S.C.)
| | - Sa Chen
- Department of Medical Biosciences, Umeå University, 901-87 Umeå, Sweden; (O.B.); (L.W.); (S.C.)
| | - Robin Fahraeus
- Department of Medical Biosciences, Umeå University, 901-87 Umeå, Sweden; (O.B.); (L.W.); (S.C.)
- RECAMO, Masaryk Memorial Cancer Institute, Zluty Kopec 7, 656-53 Brno, Czech Republic
- Inserm UMRS1131, Institut de Genetique Moleculaire, Universite Paris 7, Hopital St Louis, F-75010 Paris, France
- International Centre for Cancer Vaccine Science, University of Gdansk, 80-822 Gdansk, Poland
| |
Collapse
|
24
|
Kapoor S, Gustafson T, Zhang M, Chen YS, Li J, Nguyen N, Perez JET, Dashwood WM, Rajendran P, Dashwood RH. Deacetylase Plus Bromodomain Inhibition Downregulates ERCC2 and Suppresses the Growth of Metastatic Colon Cancer Cells. Cancers (Basel) 2021; 13:cancers13061438. [PMID: 33809839 PMCID: PMC8004213 DOI: 10.3390/cancers13061438] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/19/2021] [Indexed: 01/14/2023] Open
Abstract
There is growing evidence that DNA repair factors have clinical value for cancer treatment. Nucleotide excision repair (NER) proteins, including excision repair cross-complementation group 2 (ERCC2), play a critical role in maintaining genome integrity. Here, we examined ERCC2 expression following epigenetic combination drug treatment. Attention was drawn to ERCC2 for three reasons. First, from online databases, colorectal cancer (CRC) patients exhibited significantly reduced survival when ERCC2 was overexpressed in colon tumors. Second, ERCC2 was the most highly downregulated RNA transcript in human colon cancer cells, plus Ercc2 in rat tumors, after treatment with the histone deacetylase 3 (HDAC3) inhibitor sulforaphane (SFN) plus JQ1, which is an inhibitor of the bromodomain and extraterminal domain (BET) family. Third, as reported here, RNA-sequencing of polyposis in rat colon (Pirc) polyps following treatment of rats with JQ1 plus 6-methylsulfinylhexyl isothiocyanate (6-SFN) identified Ercc2 as the most highly downregulated gene. The current work also defined promising second-generation epigenetic drug combinations with enhanced synergy and efficacy, especially in metastasis-lineage colon cancer cells cultured as 3D spheroids and xenografts. This investigation adds to the growing interest in combination approaches that target epigenetic 'readers', 'writers', and 'erasers' that are deregulated in cancer and other pathologies, providing new avenues for precision oncology and cancer interception.
Collapse
Affiliation(s)
- Sabeeta Kapoor
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Trace Gustafson
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Mutian Zhang
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Ying-Shiuan Chen
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Jia Li
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Nhung Nguyen
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Jorge Enrique Tovar Perez
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Wan Mohaiza Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Praveen Rajendran
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
- Antibody & Biopharmaceuticals Core, Texas A&M Health, Houston, TX 77030, USA
- Correspondence: (P.R.); (R.H.D.)
| | - Roderick H. Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
- Correspondence: (P.R.); (R.H.D.)
| |
Collapse
|