1
|
Feugere L, Silva De Freitas C, Bates A, Storey KB, Beltran-Alvarez P, Wollenberg Valero KC. Social context prevents heat hormetic effects against mutagens during fish development. FEBS Lett 2025. [PMID: 40265659 DOI: 10.1002/1873-3468.70047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 03/28/2025] [Indexed: 04/24/2025]
Abstract
Since stress can be transmitted to congeners via social metabolites, it is paramount to understand how the social context of abiotic stress influences aquatic organisms' responses to global changes. Here, we integrated the transcriptomic and phenotypic responses of zebrafish embryos to a UV damage/repair assay following scenarios of heat stress, its social context and their combination. Heat stress preceding UV exposure had a hormetic effect through the cellular stress response and DNA repair, rescuing and/or protecting embryos from UV damage. However, experiencing heat stress within a social context negated this molecular hormetic effect and lowered larval fitness. We discuss the molecular basis of interindividual chemical transmission within animal groups as another layer of complexity to organisms' responses to environmental stressors.
Collapse
Affiliation(s)
- Lauric Feugere
- Department of Biological and Marine Sciences, University of Hull, Kingston upon Hull, UK
| | | | - Adam Bates
- Department of Biological and Marine Sciences, University of Hull, Kingston upon Hull, UK
| | | | - Pedro Beltran-Alvarez
- Biomedical Institute for Multimorbidity, Centre for Biomedicine, Hull York Medical School, University of Hull, Kingston upon Hull, UK
| | - Katharina C Wollenberg Valero
- Department of Biological and Marine Sciences, University of Hull, Kingston upon Hull, UK
- School of Biology and Environmental Science, University College Dublin, Ireland
- Conway Institute, University College Dublin, Ireland
| |
Collapse
|
2
|
Bonolo de Campos C, McCabe CE, Bruins LA, O'Brien DR, Brown S, Tschumper RC, Allmer C, Zhu YX, Rabe KG, Parikh SA, Kay NE, Yan H, Cerhan JR, Allan JN, Furman RR, Weinberg JB, Brander DM, Jelinek DF, Chesi M, Slager SL, Braggio E. Genomic characterization of chronic lymphocytic leukemia in patients of African ancestry. Blood Cancer J 2025; 15:14. [PMID: 39910036 PMCID: PMC11799526 DOI: 10.1038/s41408-024-01207-3] [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: 09/24/2024] [Revised: 11/26/2024] [Accepted: 12/16/2024] [Indexed: 02/07/2025] Open
Abstract
Despite the considerable effort to characterize the genomic landscape of chronic lymphocytic leukemia (CLL), published data have been almost exclusively derived from patients of European Ancestry (EA), with significant underrepresentation of minorities, including patients of African Ancestry (AA). To begin to address this gap, we evaluated whether differences exist in the genetic and transcriptomic features of 157 AA and 440 EA individuals diagnosed with CLL. We sequenced 59 putative driver genes and found an increased frequency of high-impact mutations in AA CLL, including genes of the DNA damage repair (DDR) pathway. Telomere erosion was also increased in AA CLL, amplifying the notion of increased genomic instability in AA CLL. Furthermore, we found transcription enrichment of the Tumor Necrosis Factor-alpha (TNFα) Signaling via NF-κB pathway in AA CLL compared to EA CLL, suggesting that tumor promoting inflammation plays an important role in AA CLL. In summary, these results suggest that genomic instability and NF-kB activation is more prevalent in AA CLL than EA CLL.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/ethnology
- Female
- Male
- Genomic Instability
- Middle Aged
- Aged
- Genomics/methods
- NF-kappa B/metabolism
- NF-kappa B/genetics
- Mutation
- Black People/genetics
- White People/genetics
- Aged, 80 and over
- Black or African American/genetics
Collapse
Affiliation(s)
| | - Chantal E McCabe
- Division of Computational Biology, Mayo Clinic, Rochester, MN, USA
| | - Laura A Bruins
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Daniel R O'Brien
- Division of Computational Biology, Mayo Clinic, Rochester, MN, USA
| | - Sochilt Brown
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ, USA
| | | | - Cristine Allmer
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | - Yuan Xiao Zhu
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Kari G Rabe
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | | | - Neil E Kay
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Huihuang Yan
- Division of Computational Biology, Mayo Clinic, Rochester, MN, USA
| | - James R Cerhan
- Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - John N Allan
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Richard R Furman
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, NY, USA
| | - J Brice Weinberg
- Divisions of Hematology and Hematologic Malignancies & Cellular Therapy & VA Medical Center, Durham, NC, USA
| | - Danielle M Brander
- Division of Hematologic Malignancy and Cellular Therapy, Duke Cancer Institute, Durham, NC, USA
| | | | - Marta Chesi
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Susan L Slager
- Division of Computational Biology, Mayo Clinic, Rochester, MN, USA.
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
| | - Esteban Braggio
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ, USA.
| |
Collapse
|
3
|
Xu Y, Li C, Yin H, Nowsheen S, Xu X, Kang W, Liu X, Chen L, Lou Z, Yi J, Deng M. STK39-mediated amplification of γ-H2A.X promotes homologous recombination and contributes to PARP inhibitor resistance. Nucleic Acids Res 2024; 52:13881-13895. [PMID: 39588777 DOI: 10.1093/nar/gkae1099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/17/2024] [Accepted: 10/28/2024] [Indexed: 11/27/2024] Open
Abstract
The phosphorylation of histone H2A.X into γH2A.X is a crucial early event in the DNA damage response, marking DNA damage sites and initiating repair processes. While ATM kinase is traditionally recognized as the primary mediator of H2A.X phosphorylation, our study identifies serine/threonine kinase 39 (STK39) as a novel enhancer of this critical signaling pathway. We demonstrate that after DNA damage, STK39 undergoes phosphorylation by the ATM kinase, facilitating its interaction with the Mre11-Rad50-Nbs1 complex and subsequent recruitment to chromatin. This recruitment enables STK39 to further phosphorylate H2A.X, thus amplifying γH2A.X production and promoting homologous recombination repair. Notably, we observe a significant upregulation of STK39 in pancreatic adenocarcinoma (PAAD) tissues, correlating with heightened resistance to PARPi therapy. Furthermore, we demonstrate the synergistic efficacy of combining STK39 inhibition with PARP inhibitors in suppressing and reversing PAAD growth. This study not only provides new insights into the molecular dynamics of H2A.X phosphorylation but also highlights the therapeutic potential of targeting STK39 to enhance PARPi sensitivity in PAAD (created with BioRender).
Collapse
Affiliation(s)
- Yi Xu
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Changying Li
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Huan Yin
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Somaira Nowsheen
- Department of Dermatology, University of California San Diego, 9500 Gilman Drive, La Jolla, San Diego, CA 92122, USA
| | - Xin Xu
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Wenjuan Kang
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Xin Liu
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Lifeng Chen
- Department of Gynecology, the First Affiliated Hospital, School of Medicine, Zhejiang University, No.79 Qingchun Road, Shangcheng District, Hangzhou 310003, China
| | - Zhenkun Lou
- Department of Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Junlin Yi
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| | - Min Deng
- State Key Laboratory of Molecular Oncology and Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No17, Chaoyang District, Beijing 100021, China
| |
Collapse
|
4
|
Corti G, Buzo K, Berrino E, Miotto M, Aquilano MC, Lentini M, Bellomo SE, Lorenzato A, Bartolini A, Mauri G, Lazzari L, Russo M, Di Nicolantonio F, Siena S, Marsoni S, Marchiò C, Bardelli A, Arena S. Prediction of homologous recombination deficiency identifies colorectal tumors sensitive to PARP inhibition. NPJ Precis Oncol 2024; 8:231. [PMID: 39402170 PMCID: PMC11473949 DOI: 10.1038/s41698-024-00706-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 09/09/2024] [Indexed: 10/17/2024] Open
Abstract
The synthetic lethal effect observed with the use of PARP inhibitors (PARPi) with tumors characterized by the loss of key players in the homologous recombination (HR) pathway, commonly referred to as "BRCAness", is maintaining high interest in oncology. While BRCAness is a well-established feature in breast, ovarian, prostate, and pancreatic carcinomas, our recent findings indicate that up to 15% of colorectal cancers (CRC) also harbor defects in the HR pathway, presenting promising opportunities for innovative therapeutic strategies in CRC patients. We developed a new tool called HRDirect, which builds upon the HRDetect algorithm and is able to predict HR deficiency (HRD) from reference-free tumor samples. We validated HRDirect using matched breast cancer and CRC patient samples. Subsequently, we assessed its efficacy in predicting response to the PARP inhibitor olaparib by comparing it with two other commercial assays: AmoyDx HRD by Amoy Diagnostics and the TruSight Oncology 500 HRD (TSO500-HRD) panel by Illumina NGS technology. While all three approaches successfully identified the most PARPi-sensitive CRC models, HRDirect demonstrated superior precision in distinguishing resistant models compared to AmoyDX and TSO500-HRD, which exhibited overlapping scores between sensitive and resistant cells. Furthermore, we propose integrating HRDirect scoring with ATM and RAD51C immunohistochemical analysis as part of our "composite biomarker approach" to enhance the identification of HRD tumors, with an immediate translational and clinical impact for CRC personalized treatment.
Collapse
Affiliation(s)
- Giorgio Corti
- Department of Oncology, University of Torino, Torino, Italy
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Kristi Buzo
- Department of Oncology, University of Torino, Torino, Italy
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Enrico Berrino
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Martina Miotto
- Department of Oncology, University of Torino, Torino, Italy
| | - Maria Costanza Aquilano
- Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Marilena Lentini
- Department of Oncology, University of Torino, Torino, Italy
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | | | | | | | - Gianluca Mauri
- Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Luca Lazzari
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | | | - Federica Di Nicolantonio
- Department of Oncology, University of Torino, Torino, Italy
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Salvatore Siena
- Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano, Italy
| | - Silvia Marsoni
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Caterina Marchiò
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Alberto Bardelli
- Department of Oncology, University of Torino, Torino, Italy.
- IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy.
| | - Sabrina Arena
- Department of Oncology, University of Torino, Torino, Italy.
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy.
| |
Collapse
|
5
|
Song Y, Wang W, Wang B, Shi Q. The Protective Mechanism of TFAM on Mitochondrial DNA and its Role in Neurodegenerative Diseases. Mol Neurobiol 2024; 61:4381-4390. [PMID: 38087167 DOI: 10.1007/s12035-023-03841-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/28/2023] [Indexed: 07/11/2024]
Abstract
Mitochondrial transcription factor A (TFAM) is a mitochondrial protein encoded by nuclear genes and transported from the cytoplasm to the mitochondria. TFAM is essential for the maintenance, expression, and delivery of mitochondrial DNA (mtDNA) and can regulate the replication and transcription of mtDNA. TFAM is associated with the formation of mtDNA nucleomimetic structures, mtDNA repair, and mtDNA stability. However, the mechanism by which TFAM protects mtDNA is still being studied. This review provides a summary of the protective mechanism of TFAM on mtDNA including the discrete regulatory effects of TFAM acetylation and phosphorylation on mtDNA, the regulation of Ca2+ levels by TFAM to activate transcription in mitochondria, and the increased binding of TFAM to mtDNA damage hot spots. This review also discusses the association between TFAM and some neurodegenerative diseases.
Collapse
Affiliation(s)
- Ying Song
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, People's Republic of China.
- Hangzhou King's Bio-Pharmaceutical Technology Co., Ltd., Hangzhou, 310007, Zhejiang, China.
| | - Wenjun Wang
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, People's Republic of China
| | - Beibei Wang
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, People's Republic of China
| | - Qiwen Shi
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, People's Republic of China
| |
Collapse
|
6
|
Zhang Z, Samsa WE, Gong Z. NUDT16 regulates CtIP PARylation to dictate homologous recombination repair. Nucleic Acids Res 2024; 52:3761-3777. [PMID: 38324469 PMCID: PMC11039996 DOI: 10.1093/nar/gkae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/04/2024] [Accepted: 01/22/2024] [Indexed: 02/09/2024] Open
Abstract
CtIP initiates DNA end resection and mediates homologous recombination (HR) repair. However, the underlying mechanisms of CtIP regulation and how the control of its regulation affects DNA repair remain incompletely characterized. In this study, NUDT16 loss decreases CtIP protein levels and impairs CtIP recruitment to double-strand breaks (DSBs). Furthermore, overexpression of a catalytically inactive NUDT16 mutant is unable to rescue decreased CtIP protein and impaired CtIP recruitment to DSBs. In addition, we identified a novel posttranslational modification of CtIP by ADP-ribosylation that is targeted by a PAR-binding E3 ubiquitin ligase, RNF146, leading to CtIP ubiquitination and degradation. These data suggest that the hydrolase activity of NUDT16 plays a major role in controlling CtIP protein levels. Notably, ADP-ribosylation of CtIP is required for its interaction with NUDT16, its localization at DSBs, and for HR repair. Interestingly, NUDT16 can also be ADP-ribosylated. The ADP-ribosylated NUDT16 is critical for CtIP protein stability, CtIP recruitment to DSBs, and HR repair in response to DNA damage. In summary, we demonstrate that NUDT16 and its PARylation regulate CtIP stability and CtIP recruitment to DSBs, providing new insights into our understanding of the regulation of CtIP-mediated DNA end resection in the HR repair pathway.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Cancer Biology, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - William E Samsa
- Department of Cancer Biology, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Zihua Gong
- Department of Cancer Biology, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| |
Collapse
|
7
|
Alomair A, Alamri A, Shaik J, Aljafari S, Ba Abdullah M, Alanazi M. Association between polymorphisms of the DNA repair genes RAD51 and OGG1 and risk of cardiovascular disease. Mol Med Rep 2024; 29:53. [PMID: 38334141 PMCID: PMC10865073 DOI: 10.3892/mmr.2024.13177] [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: 06/21/2023] [Accepted: 01/10/2024] [Indexed: 02/10/2024] Open
Abstract
Cardiovascular disease (CVD) is one of the leading causes of mortality worldwide, and multiple single‑nucleotide polymorphisms of DNA repair genes have been found to be associated with CVD. The aim of the present study was to assess the effects of the genetic variants of RAD51 recombinase (RAD51) and 8‑oxoguanine DNA glycosylase (OGG1) on CVD through genotyping and statistical analysis. Regardless of whether there is a significant association or not, the genotyping data on these two polymorphisms are valuable, because there is limited availability of it in certain populations. A total of 240 blood samples were analyzed and genotyped using TaqMan genotyping; 120 were obtained from cases with a history of CVD, and 120 from cases with no history of CVD. A questionnaire was administered to gather information on age, demographics, sex and clinical features, and confirmation was carried out using medical records. The results of the present study confirmed that the polymorphism rs1052133 in OGG1 had no significant association with CVD. On the other hand, the polymorphism rs1801321 in RAD51 exhibited a significant association with CVD. Collectively, the results of the present study revealed that the polymorphism rs1801321 in RAD51 exhibited a significant association with CVD, however a larger sample size to confirm the present findings, may allow for the early identification of CVD and may aid in the decision‑making process concerning treatments for CVD.
Collapse
Affiliation(s)
- Amar Alomair
- Genome Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Abdullah Alamri
- Genome Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Jilani Shaik
- Genome Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Salman Aljafari
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 11481, Kingdom of Saudi Arabia
| | - Mohammed Ba Abdullah
- Department of Biological Sciences, College of Science, King Faisal University, Al‑Ahsa 31982, Kingdom of Saudi Arabia
| | - Mohammad Alanazi
- Genome Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| |
Collapse
|
8
|
Kirk A, Graham SV. The human papillomavirus late life cycle and links to keratinocyte differentiation. J Med Virol 2024; 96:e29461. [PMID: 38345171 DOI: 10.1002/jmv.29461] [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: 08/30/2023] [Revised: 12/21/2023] [Accepted: 01/25/2024] [Indexed: 02/15/2024]
Abstract
Regulation of human papillomavirus (HPV) gene expression is tightly linked to differentiation of the keratinocytes the virus infects. HPV late gene expression is confined to the cells in the upper layers of the epithelium where the virus capsid proteins are synthesized. As these proteins are highly immunogenic, and the upper epithelium is an immune-privileged site, this spatial restriction aids immune evasion. Many decades of work have contributed to the current understanding of how this restriction occurs at a molecular level. This review will examine what is known about late gene expression in HPV-infected lesions and will dissect the intricacies of late gene regulation. Future directions for novel antiviral approaches will be highlighted.
Collapse
Affiliation(s)
- Anna Kirk
- Centre for Virus Research, University of Glasgow, Glasgow, UK
| | - Sheila V Graham
- Centre for Virus Research, University of Glasgow, Glasgow, UK
| |
Collapse
|
9
|
Zhao D, Long X, Wang J. Dose Adjustment of Poly (ADP‑Ribose) Polymerase Inhibitors in Patients with Hepatic or Renal Impairment. Drug Des Devel Ther 2022; 16:3947-3955. [PMID: 36405648 PMCID: PMC9673935 DOI: 10.2147/dddt.s387920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/08/2022] [Indexed: 08/30/2023] Open
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors are small-molecule inhibitors of PARP enzymes (including PARP1, PARP2, and PARP3) that exhibit activity against tumor cells with defects in DNA repair. In recent years, five PARP inhibitors, olaparib, niraparib, rucaparib, talazoparib and veliparib, have been developed for the treatment of solid tumors, particularly in patients with breast-related cancer antigen (BRCA) 1/2 mutations, or those without a functional homologous recombination repair pathway. These novel treatments exhibit improved efficacy and toxicity when compared to conventional chemotherapy agents. The five PARP inhibitors are eliminated primarily via the liver and kidneys, hepatic or renal impairment may significantly affect their pharmacokinetics (PK). Therefore, it is important to know the effects of hepatic or renal impairment on the PK and safety of PARP inhibitors. In this review, we characterize and summarize the effects of hepatic and renal function on the PK of PARP inhibitors and provide specific recommendations for clinicians when prescribing PARP inhibitors in patients with hepatic or renal impairment.
Collapse
Affiliation(s)
- Dehua Zhao
- Department of Clinical Pharmacy, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan, People’s Republic of China
| | - Xiaoqing Long
- Department of Clinical Pharmacy, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan, People’s Republic of China
| | - Jisheng Wang
- Department of Clinical Pharmacy, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan, People’s Republic of China
| |
Collapse
|
10
|
Lohberger B, Barna S, Glänzer D, Eck N, Kerschbaum-Gruber S, Stasny K, Leithner A, Georg D. Cellular and Molecular Biological Alterations after Photon, Proton, and Carbon Ions Irradiation in Human Chondrosarcoma Cells Linked with High-Quality Physics Data. Int J Mol Sci 2022; 23:11464. [PMID: 36232764 PMCID: PMC9569755 DOI: 10.3390/ijms231911464] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/08/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Chondrosarcomas are particularly difficult to treat due to their resistance to chemotherapy and radiotherapy. However, particle therapy can enhance local control and patient survival rates. To improve our understanding of the basic cellular radiation response, as a function of dose and linear energy transfer (LET), we developed a novel water phantom-based setup for cell culture experiments and characterized it dosimetrically. In a direct comparison, human chondrosarcoma cell lines were analyzed with regard to their viability, cell proliferation, cell cycle, and DNA repair behavior after irradiation with X-ray, proton, and carbon ions. Our results clearly showed that cell viability and proliferation were inhibited according to the increasing ionization density, i.e., LET, of the irradiation modes. Furthermore, a prominent G2/M arrest was shown. Gene expression profiling proved the upregulation of the senescence genes CDKN1A (p21), CDKN2A (p16NK4a), BMI1, and FOXO4 after particle irradiation. Both proton or C-ion irradiation caused a positive regulation of the repair genes ATM, NBN, ATXR, and XPC, and a highly significant increase in XRCC1/2/3, ERCC1, XPC, and PCNA expression, with C-ions appearing to activate DNA repair mechanisms more effectively. The link between the physical data and the cellular responses is an important contribution to the improvement of the treatment system.
Collapse
Affiliation(s)
- Birgit Lohberger
- Department of Orthopaedics and Trauma, Medical University of Graz, 8036 Graz, Austria
| | - Sandra Barna
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Dietmar Glänzer
- Department of Orthopaedics and Trauma, Medical University of Graz, 8036 Graz, Austria
| | - Nicole Eck
- Department of Orthopaedics and Trauma, Medical University of Graz, 8036 Graz, Austria
| | | | | | - Andreas Leithner
- Department of Orthopaedics and Trauma, Medical University of Graz, 8036 Graz, Austria
| | - Dietmar Georg
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria
- MedAustron Ion Therapy Center, 2700 Wiener Neustadt, Austria
| |
Collapse
|
11
|
Yin C, Kulasekaran M, Roy T, Decker B, Alexander S, Margolis M, Jha RC, Kupfer GM, He AR. Homologous Recombination Repair in Biliary Tract Cancers: A Prime Target for PARP Inhibition? Cancers (Basel) 2022; 14:2561. [PMID: 35626165 PMCID: PMC9140037 DOI: 10.3390/cancers14102561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/10/2022] [Accepted: 05/14/2022] [Indexed: 01/27/2023] Open
Abstract
Biliary tract cancers (BTCs) are a heterogeneous group of malignancies that make up ~7% of all gastrointestinal tumors. It is notably aggressive and difficult to treat; in fact, >70% of patients with BTC are diagnosed at an advanced, unresectable stage and are not amenable to curative therapy. For these patients, chemotherapy has been the mainstay treatment, providing an inadequate overall survival of less than one year. Despite the boom in targeted therapies over the past decade, only a few targeted agents have been approved in BTCs (i.e., IDH1 and FGFR inhibitors), perhaps in part due to its relatively low incidence. This review will explore current data on PARP inhibitors (PARPi) used in homologous recombination deficiency (HRD), particularly with respect to BTCs. Greater than 28% of BTC cases harbor mutations in genes involved in homologous recombination repair (HRR). We will summarize the mechanisms for PARPi and its role in synthetic lethality and describe select genes in the HRR pathway contributing to HRD. We will provide our rationale for expanding patient eligibility for PARPi use based on literature and anecdotal evidence pertaining to mutations in HRR genes, such as RAD51C, and the potential use of reliable surrogate markers of HRD.
Collapse
Affiliation(s)
- Chao Yin
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA; (C.Y.); (M.K.); (T.R.)
| | - Monika Kulasekaran
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA; (C.Y.); (M.K.); (T.R.)
| | - Tina Roy
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA; (C.Y.); (M.K.); (T.R.)
| | - Brennan Decker
- Foundation Medicine, Cambridge, MA 20007, USA; (B.D.); (S.A.); (M.M.)
| | - Sonja Alexander
- Foundation Medicine, Cambridge, MA 20007, USA; (B.D.); (S.A.); (M.M.)
| | - Mathew Margolis
- Foundation Medicine, Cambridge, MA 20007, USA; (B.D.); (S.A.); (M.M.)
| | - Reena C. Jha
- Department of Radiology, Georgetown University Medical Center, Washington, DC 20007, USA;
| | - Gary M. Kupfer
- Departments of Oncology and Pediatrics, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA;
| | - Aiwu R. He
- Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA; (C.Y.); (M.K.); (T.R.)
| |
Collapse
|
12
|
Wang S, Xiao X, Qiu M, Wang W, Xiao Y, Yang H, Dang Y, Wang W. Transcriptomic Responses of Salmonella enterica Serovars Enteritidis in Sodium Hypochlorite. Front Cell Infect Microbiol 2022; 12:853064. [PMID: 35521218 PMCID: PMC9065344 DOI: 10.3389/fcimb.2022.853064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/24/2022] [Indexed: 11/24/2022] Open
Abstract
Salmonella enterica serovars Enteritidis (S. Enteritidis) can survive extreme food processing environments including bactericidal sodium hypochlorite (NaClO) treatments generally recognized as safe. In order to reveal the molecular regulatory mechanisms underlying the phenotypes, the overall regulation of genes at the transcription level in S. Enteritidis after NaClO stimulation were investigated by RNA-sequencing. We identified 1399 differentially expressed genes (DEG) of S. Enteritidis strain CVCC 1806 following treatment in liquid culture with 100 mg/L NaClO for 20 min (915 upregulated and 484 downregulated). NaClO stress affects the transcription of genes related to a range of important biomolecular processes such as membrane damage, membrane transport function, energy metabolism, oxidative stress, DNA repair, and other important processes in Salmonella enterica. First, NaClO affects the structural stability of cell membranes, which induces the expression of a range of outer and inner membrane proteins. This may lead to changes in cell membrane permeability, accelerating the frequency of DNA conversion and contributing to the production of drug-resistant bacteria. In addition, the expression of exocytosis pump genes (emrB, yceE, ydhE, and ydhC) was able to expel NaClO from the cell, thereby increasing bacterial tolerance to NaClO. Secondly, downregulation of genes related to the Kdp-ATPase transporter system (kdpABC) and the amino acid transporter system (aroP, brnQ and livF) may to some extent reduce active transport by bacterial cells, thereby reducing their own metabolism and the entry of disinfectants. Downregulation of genes related to the tricarboxylic acid (TCA) cycle may drive bacterial cells into a viable but non-culturable (VBNC) state, resisting NaClO attack by reducing energy metabolism. In addition, significant upregulation of genes related to oxidative stress could mitigate damage caused by disinfectants by eliminating alkyl hydroperoxides, while upregulation of genes related to DNA repair could repair damage to bacterial cells caused by oxidative stress. Therefore, this study indicated that S. Enteritidis has genomic mechanisms to adapt to NaClO stress.
Collapse
Affiliation(s)
- Sheng Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, China
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, MOA Laboratory of Quality & Safety Risk Assessment for Agro-Products (Hangzhou), Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Xingning Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, MOA Laboratory of Quality & Safety Risk Assessment for Agro-Products (Hangzhou), Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Mengjia Qiu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, MOA Laboratory of Quality & Safety Risk Assessment for Agro-Products (Hangzhou), Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Wensi Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, MOA Laboratory of Quality & Safety Risk Assessment for Agro-Products (Hangzhou), Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Yingping Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, MOA Laboratory of Quality & Safety Risk Assessment for Agro-Products (Hangzhou), Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Hua Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, MOA Laboratory of Quality & Safety Risk Assessment for Agro-Products (Hangzhou), Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Yali Dang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, China
- *Correspondence: Wen Wang, ; Yali Dang,
| | - Wen Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, MOA Laboratory of Quality & Safety Risk Assessment for Agro-Products (Hangzhou), Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
- *Correspondence: Wen Wang, ; Yali Dang,
| |
Collapse
|
13
|
Quantitative proteomic analysis of the lysine acetylome reveals diverse SIRT2 substrates. Sci Rep 2022; 12:3822. [PMID: 35264593 PMCID: PMC8907344 DOI: 10.1038/s41598-022-06793-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 02/02/2022] [Indexed: 12/24/2022] Open
Abstract
Sirtuin 2 (SIRT2) is a NAD+-dependent deacetylase, which regulates multiple biological processes, including genome maintenance, aging, tumor suppression, and metabolism. While a number of substrates involved in these processes have been identified, the global landscape of the SIRT2 acetylome remains unclear. Using a label-free quantitative proteomic approach following enrichment for acetylated peptides from SIRT2-depleted and SIRT2-overexpressing HCT116 human colorectal cancer cells, we identified a total of 2,846 unique acetylation sites from 1414 proteins. 896 sites from 610 proteins showed a > 1.5-fold increase in acetylation with SIRT2 knockdown, and 509 sites from 361 proteins showed a > 1.5-fold decrease in acetylation with SIRT2 overexpression, with 184 proteins meeting both criteria. Sequence motif analyses identified several site-specific consensus sequence motifs preferentially recognized by SIRT2, most commonly KxxxxK(ac). Gene Ontology, KEGG, and MetaCore pathway analyses identified SIRT2 substrates involved in diverse pathways, including carbon metabolism, glycolysis, spliceosome, RNA transport, RNA binding, transcription, DNA damage response, the cell cycle, and colorectal cancer. Collectively, our findings expand on the number of known acetylation sites, substrates, and cellular pathways targeted by SIRT2, providing support for SIRT2 in regulating networks of proteins in diverse pathways and opening new avenues of investigation into SIRT2 function.
Collapse
|
14
|
Lohberger B, Glänzer D, Eck N, Kerschbaum-Gruber S, Mara E, Deycmar S, Madl T, Kashofer K, Georg P, Leithner A, Georg D. Activation of efficient DNA repair mechanisms after photon and proton irradiation of human chondrosarcoma cells. Sci Rep 2021; 11:24116. [PMID: 34916568 PMCID: PMC8677811 DOI: 10.1038/s41598-021-03529-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/24/2021] [Indexed: 01/02/2023] Open
Abstract
Although particle therapy with protons has proven to be beneficial in the treatment of chondrosarcoma compared to photon-based (X-ray) radiation therapy, the cellular and molecular mechanisms have not yet been sufficiently investigated. Cell viability and colony forming ability were analyzed after X-ray and proton irradiation (IR). Cell cycle was analyzed using flow cytometry and corresponding regulator genes and key players of the DNA repair mechanisms were measured using next generation sequencing, protein expression and immunofluorescence staining. Changes in metabolic phenotypes were determined with nuclear magnetic resonance spectroscopy. Both X-ray and proton IR resulted in reduced cell survival and a G2/M phase arrest of the cell cycle. Especially 1 h after IR, a significant dose-dependent increase of phosphorylated γH2AX foci was observed. This was accompanied with a reprogramming in cellular metabolism. Interestingly, within 24 h the majority of clearly visible DNA damages were repaired and the metabolic phenotype restored. Involved DNA repair mechanisms are, besides the homology directed repair (HDR) and the non-homologous end-joining (NHEJ), especially the mismatch mediated repair (MMR) pathway with the key players EXO1, MSH3, and PCNA. Chondrosarcoma cells regenerates the majority of DNA damages within 24 h. These molecular mechanisms represent an important basis for an improved therapy.
Collapse
Affiliation(s)
- Birgit Lohberger
- Department of Orthopedics and Trauma, Medical University of Graz, Auenbruggerplatz 5-7, 8036, Graz, Austria.
| | - Dietmar Glänzer
- Department of Orthopedics and Trauma, Medical University of Graz, Auenbruggerplatz 5-7, 8036, Graz, Austria
| | - Nicole Eck
- Department of Orthopedics and Trauma, Medical University of Graz, Auenbruggerplatz 5-7, 8036, Graz, Austria
| | - Sylvia Kerschbaum-Gruber
- Department of Radiation Oncology, Medical University of Vienna, 1090, Vienna, Austria
- MedAustron Ion Therapy Center, 2700, Wiener Neustadt, Austria
| | - Elisabeth Mara
- MedAustron Ion Therapy Center, 2700, Wiener Neustadt, Austria
- University of Applied Science, 2700, Wiener Neustadt, Austria
| | - Simon Deycmar
- Laboratory for Applied Radiobiology, University Zurich, 8006, Zurich, Switzerland
| | - Tobias Madl
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Molecular Biology and Biochemistry, Medical University of Graz, 8010, Graz, Austria
- BioTechMed-Graz, 8010, Graz, Austria
| | - Karl Kashofer
- Institute of Pathology, Medical University of Graz, 8010, Graz, Austria
| | - Petra Georg
- MedAustron Ion Therapy Center, 2700, Wiener Neustadt, Austria
| | - Andreas Leithner
- Department of Orthopedics and Trauma, Medical University of Graz, Auenbruggerplatz 5-7, 8036, Graz, Austria
| | - Dietmar Georg
- Department of Radiation Oncology, Medical University of Vienna, 1090, Vienna, Austria
- MedAustron Ion Therapy Center, 2700, Wiener Neustadt, Austria
| |
Collapse
|
15
|
Fan XZ, Chen YF, Zhang SB, He DH, Wei SF, Wang Q, Pan HF, Liu YQ. Centipeda minima extract sensitizes lung cancer cells to DNA-crosslinking agents via targeting Fanconi anemia pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153689. [PMID: 34446320 DOI: 10.1016/j.phymed.2021.153689] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 06/19/2021] [Accepted: 07/28/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Intrinsic and acquired chemoresistance remains a critical challenge in lung cancer chemotherapy. Fanconi anemia (FA) pathway plays an important role in antagonizing the cytotoxic effects of chemotherapeutics by repairing DNA damage. We recently demonstrated that the traditional Chinese medicinal herb, Centipeda minima (C. minima), possessed anti-inflammatory and antioxidant properties. However, the potential anticancer application of C. minima and the underlying mechanisms remain unclear. PURPOSE We aimed to investigate the combined anticancer effects of the ethanol extract of C. minima (ECM) and DNA-crosslinking agents on non-small cell lung cancer (NSCLC) and elucidate the underlying mechanisms. METHODS Cell viability and flow cytometry assay were performed to determine the synergistic cytotoxicity of ECM and DNA-crosslinking agents, cisplatin (CDDP) or mitomycin C (MMC), in NSCLC cells. Western blotting and immunofluorescence were conducted to examine the effects of ECM on protein expression in DNA damage repair pathway. Comet assay was applied to evaluate DNA damage levels. Subcutaneous xenografts of NSCLC were established to evaluate the combined anticancer effects of ECM and CDDP. RESULTS Combined treatments with ECM and DNA-crosslinking agents exhibited synergistic cytotoxic effects against A549 and H1299 cells. FANCD2 was highly expressed in NSCLC that correlates with poor prognosis of NSCLC patients, based on the online database analysis. ECM significantly inhibited DNA damage-induced monoubiquitination and nuclear foci formation of FANCD2, thereby sensitizing NSCLC to CDDP- or MMC-induced DNA damage and apoptosis, as evidenced by increased expression of γ-H2AX, increased cleavage of caspases-3 and PARP, and enhanced Annexin V-FITC/PI staining. Further, ECM can also decrease the protein level of FANCD2 that contributes to the chemosensitizing effects. Moreover, ECM significantly attenuated CDDP-mediated S-phase arrest by antagonizing the activation of ATR/Chk1 pathway in NSCLC cells. Animal experiments further demonstrated that ECM and CDDP combination treatment synergistically inhibited tumor growth by decreasing FANCD2 protein level in tumor tissues. CONCLUSION Our results demonstrated that ECM can inhibit DNA-crosslinking agents-induced activation of FA pathway by attenuating both the expression and monoubiquitination of FANCD2. ECM and CDDP combination therapy exhibited synergistic anticancer effects both in vitro and in vivo, indicating that ECM and its active components might serve as novel anticancer drugs in the combination chemotherapy.
Collapse
Affiliation(s)
- Xiang-Zhen Fan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Yu-Fei Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Research Center of Chinese Herbal Resources Science and Engineering, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Shi-Bing Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Dan-Hua He
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Research Center of Chinese Herbal Resources Science and Engineering, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Su-Fen Wei
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Hua-Feng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Yong-Qiang Liu
- Research Center of Chinese Herbal Resources Science and Engineering, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Key Laboratory of Chinese Medicinal Resource from Lingnan, Ministry of Education, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|
16
|
de Barros TT, Venâncio VDP, Hernandes LC, Greggi Antunes LM, Hillesheim E, Salomão RG, Mathias MG, Coelho-Landell CA, Toffano RBD, Almada MORDV, Camelo- JS, Moco S, Ued FDV, Kaput J, Monteiro JP. DNA damage is inversely associated to blood levels of DHA and EPA fatty acids in Brazilian children and adolescents. Food Funct 2021; 11:5115-5121. [PMID: 32432238 DOI: 10.1039/c9fo02551k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This study aimed to investigate the association between DNA damage and blood levels of docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), retinol, beta-carotene and riboflavin in Brazilian children and adolescents. Subjects (n = 140) were healthy boys and girls aged 9 to 13 years in Ribeirão Preto (SP, Brazil). Data collection included anthropometry, assessment of energy intake and blood sampling. DNA damage was evaluated by single-cell gel electrophoresis (comet assay). Principal component analysis (PCA) was used to verify associations between blood concentrations of vitamins, polyunsaturated fatty acids and DNA damage. Multiple regression analyses, k-means cluster, and analysis of covariance (ANCOVA), adjusted for confounding variables such as age, sex, energy intake, body mass index and total cholesterol (when needed), were applied to confirm the associations. PCA explained 69.4% of the inverse relationships between DNA damage and blood levels of DHA, EPA, retinol, and beta-carotene. Results were confirmed by ANCOVA and multiple regression analyses for DHA and EPA. In conclusion, omega-3-fatty acids were inversely associated with DNA damage in Brazilian children and adolescents and may be a protective factor against the development of future diseases.
Collapse
Affiliation(s)
- Tamiris Trevisan de Barros
- Department of Pediatrics and Department of Health Sciences, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Vinicius de Paula Venâncio
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Lívia Cristina Hernandes
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Elaine Hillesheim
- Department of Pediatrics and Department of Health Sciences, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Roberta Garcia Salomão
- Department of Pediatrics and Department of Health Sciences, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Mariana Giaretta Mathias
- Department of Pediatrics and Department of Health Sciences, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Carolina Almeida Coelho-Landell
- Department of Pediatrics and Department of Health Sciences, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Roseli Borges Donegá Toffano
- Department of Pediatrics and Department of Health Sciences, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Maria Olimpia Ribeiro do Vale Almada
- Department of Pediatrics and Department of Health Sciences, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - José Simon Camelo-
- Department of Pediatrics and Department of Health Sciences, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Sofia Moco
- Nestlé Institute of Health Sciences, Lausanne, Switzerland
| | - Fábio da Veiga Ued
- Department of Pediatrics and Department of Health Sciences, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Jim Kaput
- Nestlé Institute of Health Sciences, Lausanne, Switzerland
| | - Jacqueline Pontes Monteiro
- Department of Pediatrics and Department of Health Sciences, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
17
|
Katoueezadeh M, Pilehvari N, Fatemi A, Hassanshahi G, Torabizadeh SA. Inhibition of DNA damage response pathway using combination of DDR pathway inhibitors and radiation in treatment of acute lymphoblastic leukemia cells. Future Oncol 2021; 17:2803-2816. [PMID: 33960207 DOI: 10.2217/fon-2020-1072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
An alarming increase in acute lymphoblastic leukemia cases among children and adults has attracted the attention of researchers to discover new therapeutic strategies with a better prognosis. In cancer cells, the DNA damage response (DDR) pathway elements have been recognized to protect tumor cells from various stresses and cause tumor progression; targeting these DDR members is an attractive strategy for treatment of cancers. The inhibition of the DDR pathway in cancer cells for the treatment of cancers has recently been introduced. Hence, effective treatment strategies are needed for this purpose. Chemotherapy in combination with radiotherapy is considered a potential therapeutic strategy for acute leukemia. This review aims to assess the synergistic effects of these inhibitors with irradiation for the treatment of leukemia.
Collapse
Affiliation(s)
- Maryam Katoueezadeh
- Department of Hematology & Medical Laboratory Science, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, 7616911333, Iran
| | - Niloofar Pilehvari
- Department of Hematology & Medical Laboratory Science, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, 7616911333, Iran
| | - Ahmad Fatemi
- Department of Hematology & Medical Laboratory Science, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, 7616911333, Iran
| | - Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, 7718796755, Iran
| | - Seyedeh Atekeh Torabizadeh
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, 7616911319, Iran
| |
Collapse
|
18
|
Comparative Therapeutic Exploitability of Acute Adaptation Mechanisms to Photon and Proton Irradiation in 3D Head and Neck Squamous Cell Carcinoma Cell Cultures. Cancers (Basel) 2021; 13:cancers13061190. [PMID: 33801853 PMCID: PMC8000891 DOI: 10.3390/cancers13061190] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/23/2021] [Accepted: 03/06/2021] [Indexed: 12/16/2022] Open
Abstract
For better tumor control, high-precision proton beam radiation therapy is currently being intensively discussed relative to conventional photon therapy. Here, we assumed that radiation type-specific molecular response profiles in more physiological 3D, matrix-based head and neck squamous cell carcinoma (HNSCC) cell cultures can be identified and therapeutically exploited. While proton irradiation revealed superimposable clonogenic survival and residual DNA double strand breaks (DSB) relative to photon irradiation, kinome profiles showed quantitative differences between both irradiation types. Pharmacological inhibition of a subset of radiation-induced kinases, predominantly belonging to the mitogen-activated protein kinase (MAPK) family, failed to sensitize HNSCC cells to either proton or photon irradiation. Likewise, inhibitors for ATM, DNA-PK and PARP did not discriminate between proton and photon irradiation but generally elicited a radiosensitization. Conclusively, our results suggest marginal cell line-specific differences in the radiosensitivity and DSB repair without a superiority of one radiation type over the other in 3D grown HNSCC cell cultures. Importantly, radiation-induced activity changes of cytoplasmic kinases induced during the first, acute phase of the cellular radiation response could neither be exploited for sensitization of HNSCC cells to photon nor proton irradiation.
Collapse
|
19
|
Pursuwani BH, Bhatt BS, Vaidya FU, Pathak C, Patel MN. Synthesis, Characterization, and Biological Evaluation of Osmium(IV) Pyrazole Carbothioamide Complexes. Polycycl Aromat Compd 2020. [DOI: 10.1080/10406638.2020.1852581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Bharat H. Pursuwani
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| | - Bhupesh S. Bhatt
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| | - Foram U. Vaidya
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| | - Chandramani Pathak
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| | - Mohan N. Patel
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| |
Collapse
|
20
|
Deville SS, Luft S, Kaufmann M, Cordes N. Keap1 inhibition sensitizes head and neck squamous cell carcinoma cells to ionizing radiation via impaired non-homologous end joining and induced autophagy. Cell Death Dis 2020; 11:887. [PMID: 33087706 PMCID: PMC7578798 DOI: 10.1038/s41419-020-03100-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022]
Abstract
The function of Keap1 (Kelch-like ECH-associated protein 1), a sensor of oxidative and electrophilic stress, in the radiosensitivity of cancer cells remains elusive. Here, we investigated the effects of pharmacological inhibition of Keap1 with ML344 on radiosensitivity, DNA double-strand break (DSB) repair and autophagy in head and neck squamous cell carcinoma (HNSCC) cell lines. Our data demonstrate that Keap1 inhibition enhances HNSCC cell radiosensitivity. Despite elevated, Nrf2-dependent activity of non-homologous end joining (NHEJ)-related DNA repair, Keap1 inhibition seems to impair DSB repair through delayed phosphorylation of DNA-PKcs. Moreover, Keap1 inhibition elicited autophagy and increased p62 levels when combined with X-ray irradiation. Our findings suggest HNSCC cell radiosensitivity, NHEJ-mediated DSB repair, and autophagy to be co-regulated by Keap1.
Collapse
Affiliation(s)
- Sara Sofia Deville
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany
| | - Susanne Luft
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Maria Kaufmann
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nils Cordes
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany. .,Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany. .,Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany. .,German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany. .,German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
21
|
Gao A, Guo M. Epigenetic based synthetic lethal strategies in human cancers. Biomark Res 2020; 8:44. [PMID: 32974031 PMCID: PMC7493427 DOI: 10.1186/s40364-020-00224-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 09/04/2020] [Indexed: 02/08/2023] Open
Abstract
Over the past decades, it is recognized that loss of DNA damage repair (DDR) pathways is an early and frequent event in tumorigenesis, occurring in 40-50% of many cancer types. The basis of synthetic lethality in cancer therapy is DDR deficient cancers dependent on backup DNA repair pathways. In cancer, the concept of synthetic lethality has been extended to pairs of genes, in which inactivation of one by deletion or mutation and pharmacological inhibition of the other leads to death of cancer cells whereas normal cells are spared the effect of the drug. The paradigm study is to induce cell death by inhibiting PARP in BRCA1/2 defective cells. Since the successful application of PARP inhibitor, a growing number of developed DDR inhibitors are ongoing in preclinical and clinical testing, including ATM, ATR, CHK1/2 and WEE1 inhibitors. Combination of PARP inhibitors and other DDR inhibitors, or combination of multiple components of the same pathway may have great potential synthetic lethality efficiency. As epigenetics joins Knudson’s two hit theory, silencing of DDR genes by aberrant epigenetic changes provide new opportunities for synthetic lethal therapy in cancer. Understanding the causative epigenetic changes of loss-of-function has led to the development of novel therapeutic agents in cancer. DDR and related genes were found frequently methylated in human cancers, including BRCA1/2, MGMT, WRN, MLH1, CHFR, P16 and APC. Both genetic and epigenetic alterations may serve as synthetic lethal therapeutic markers.
Collapse
Affiliation(s)
- Aiai Gao
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing, 100853 China
| | - Mingzhou Guo
- Department of Gastroenterology and Hepatology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing, 100853 China.,Henan Key Laboratory for Esophageal Cancer Research, Zhengzhou University, 40 Daxue Road, Zhengzhou, 450052 Henan China.,State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, #28 Fuxing Road, Beijing, 100853 China
| |
Collapse
|
22
|
Zhang AL, Tang SF, Yang Y, Li CZ, Ding XJ, Zhao H, Wang JH, Yang GH, Li J. Histone demethylase JHDM2A regulates H3K9 dimethylation in response to arsenic-induced DNA damage and repair in normal human liver cells. J Appl Toxicol 2020; 40:1661-1672. [PMID: 32608101 DOI: 10.1002/jat.4026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/17/2022]
Abstract
Long-term arsenic exposure is a worldwide public health problem that causes serious harm to human health. The liver is the main target organ of arsenic toxicity; arsenic induces disruption of the DNA damage repair pathway, but its mechanisms remain unclear. In recent years, studies have found that epigenetic mechanisms play an important role in arsenic-induced lesions. In this study, we conducted experiments in vitro using normal human liver cells (L-02) to explore the mechanism by which the histone demethylase JHDM2A regulates H3K9 dimethylation (me2) in response to arsenic-induced DNA damage. Our results indicated that arsenic exposure upregulated the expression of JHDM2A, downregulated global H3K9me2 modification levels, increased the H3K9me2 levels at the promoters of base excision repair (BER) genes (N-methylpurine-DNA glycosylase [MPG], XRCC1 and poly(ADP-ribose)polymerase 1) and inhibited their expression levels, causing DNA damage in cells. In addition, we studied the effects of overexpression and inhibition of JHDM2A and found that JHDM2A can participate in the molecular mechanism of arsenic-induced DNA damage via the BER pathway, which may not be involved in the BER process because H3K9me2 levels at the promoter region of the BER genes were unchanged following JHDM2A interference. These results suggest a potential mechanism by which JHDM2A can regulate the MPG and XRCC1 genes in the process of responding to DNA damage induced by arsenic exposure and can participate in the process of DNA damage repair, which provides a scientific basis for understanding the epigenetic mechanisms and treatments for endemic arsenic poisoning.
Collapse
Affiliation(s)
- An-Liu Zhang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Shun-Fang Tang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Yue Yang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Chang-Zhe Li
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Xue-Jiao Ding
- First Affiliated Hospital of Jiangxi Medical College, Shangrao, Jiangxi, China
| | - Hua Zhao
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Jun-Hua Wang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Guang-Hong Yang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - Jun Li
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| |
Collapse
|
23
|
Deville SS, Vehlow A, Förster S, Dickreuter E, Borgmann K, Cordes N. The Intermediate Filament Synemin Regulates Non-Homologous End Joining in an ATM-Dependent Manner. Cancers (Basel) 2020; 12:cancers12071717. [PMID: 32605308 PMCID: PMC7407367 DOI: 10.3390/cancers12071717] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/20/2020] [Accepted: 06/24/2020] [Indexed: 01/26/2023] Open
Abstract
The treatment resistance of cancer cells is a multifaceted process in which DNA repair emerged as a potential therapeutic target. DNA repair is predominantly conducted by nuclear events; yet, how extra-nuclear cues impact the DNA damage response is largely unknown. Here, using a high-throughput RNAi-based screen in three-dimensionally-grown cell cultures of head and neck squamous cell carcinoma (HNSCC), we identified novel focal adhesion proteins controlling DNA repair, including the intermediate filament protein, synemin. We demonstrate that synemin critically regulates the DNA damage response by non-homologous end joining repair. Mechanistically, synemin forms a protein complex with DNA-PKcs through its C-terminal tail domain for determining DNA repair processes upstream of this enzyme in an ATM-dependent manner. Our study discovers a critical function of the intermediate filament protein, synemin in the DNA damage response, fundamentally supporting the concept of cytoarchitectural elements as co-regulators of nuclear events.
Collapse
Affiliation(s)
- Sara Sofia Deville
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (S.S.D.); (A.V.); (S.F.); (E.D.)
- Helmholtz-Zentrum Dresden—Rossendorf (HZDR), Institute of Radiooncology—OncoRay, 01328 Dresden, Germany
| | - Anne Vehlow
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (S.S.D.); (A.V.); (S.F.); (E.D.)
- National Center for Tumor Diseases, Partner Site Dresden: German Cancer Research Center, 69120 Heidelberg, Germany
- German Cancer Consortium, Partner Site Dresden: German Cancer Research Center, 69120 Heidelberg, Germany
| | - Sarah Förster
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (S.S.D.); (A.V.); (S.F.); (E.D.)
- Helmholtz-Zentrum Dresden—Rossendorf (HZDR), Institute of Radiooncology—OncoRay, 01328 Dresden, Germany
| | - Ellen Dickreuter
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (S.S.D.); (A.V.); (S.F.); (E.D.)
| | - Kerstin Borgmann
- Laboratory of Radiobiology and Experimental Radiation Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Nils Cordes
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (S.S.D.); (A.V.); (S.F.); (E.D.)
- Helmholtz-Zentrum Dresden—Rossendorf (HZDR), Institute of Radiooncology—OncoRay, 01328 Dresden, Germany
- German Cancer Consortium, Partner Site Dresden: German Cancer Research Center, 69120 Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Correspondence: ; Tel.: +49-(0)351-458-7401; Fax: +49-(0)351-458-7311
| |
Collapse
|
24
|
Zhang AL, Chen L, Ma L, Ding XJ, Tang SF, Zhang AH, Li J. Role of H3K18ac-regulated nucleotide excision repair-related genes in arsenic-induced DNA damage and repair of HaCaT cells. Hum Exp Toxicol 2020; 39:1168-1177. [PMID: 32031413 DOI: 10.1177/0960327120903482] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Arsenic is an environmental poison and is a grade I human carcinogen that can cause many types of damage to the body. The skin is one of the main target organs of arsenic damage, but the molecular mechanisms underlying arsenic poisoning are not clear. Arsenic is an epigenetic agent. Histone acetylation is one of the earliest covalent modifications to be discovered and is closely related to the occurrence and development of tumors. To investigate the role of acetylated histone H3K18 (H3K18 ac) in arsenic-induced DNA damage, HaCaT cells were exposed to sodium arsenite (NaAsO2) for 24 h. It was found that arsenic induced the downregulation of xeroderma pigmentosum A, D, and F (XPA, XPD, and XPF-nucleotide excision repair (NER)-related genes) expression, as well as histone H3K18 ac expression, and aggravated DNA damage. Chromatin immunoprecipitation quantitative polymerase chain reaction (ChIP-qPCR) analysis showed that H3K18 acetylation in the promoter regions of XPA, XPD, and XPF was downregulated. In addition, the use of the histone deacetylase inhibitor trichostatin A (TSA) partially inhibited arsenic-induced DNA damage, inhibited deacetylation of H3K18 ac in the promoter regions of XPA, XPD, and XPF genes, increased acetylation of H3K18, and promoted the transcriptional expression of NER-related genes. Our study revealed that NaAsO2 induces DNA damage and inhibits the expression of NER-related genes, while TSA increases the H3K18 ac enrichment level and promotes the transcriptional expression of NER, thereby inhibiting DNA damage. These findings provide new ideas for understanding the molecular mechanisms underlying arsenic-induced skin damage.
Collapse
Affiliation(s)
- A L Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - L Chen
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - L Ma
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - X J Ding
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - S F Tang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - A H Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| | - J Li
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, China
| |
Collapse
|
25
|
Vodicka P, Vodenkova S, Buchler T, Vodickova L. DNA repair capacity and response to treatment of colon cancer. Pharmacogenomics 2019; 20:1225-1233. [PMID: 31691643 DOI: 10.2217/pgs-2019-0070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
DNA repair, a complex biological process, ensures genomic integrity. Alterations in DNA repair, occurring in many cancers, contribute to the accumulation of mutations in the genome, resulting in genomic instability and cancer progression. DNA repair also plays a substantial role in response to chemotherapeutics: rapidly dividing colon cancer cells, vulnerable to DNA-damaging agents and overcoming DNA repair, undergo cell death. DNA repair capacity represents a complex biomarker, integrating gene variants, gene expressions, the stability of gene products, the effect of inhibitors/stimulators, lifestyle and environmental factors. Here, we discuss DNA repair capacity in sporadic colon cancer, a frequent malignancy worldwide, in relation to tumor heterogeneity, prognosis and prediction, measurements in surrogate and target tissues and suggest important tasks to be addressed.
Collapse
Affiliation(s)
- Pavel Vodicka
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine, The Czech Academy of Sciences, Prague, Czech Republic.,Institute of Biology & Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic.,Laboratory of Cancer Treatment and Tissue Regeneration, Faculty of Medicine & Biomedical Center in Pilsen, Charles University, Pilsen, Czech Republic
| | - Sona Vodenkova
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine, The Czech Academy of Sciences, Prague, Czech Republic.,Institute of Biology & Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic.,Department of Medical Genetics, Third Faculty of Medicine, Charles University, Ruska 2411/87, 100 00 Prague, Czech Republic
| | - Tomas Buchler
- Department of Oncology, First Faculty of Medicine, Charles University & Thomayer Hospital, Prague, Czech Republic
| | - Ludmila Vodickova
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine, The Czech Academy of Sciences, Prague, Czech Republic.,Institute of Biology & Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic.,Laboratory of Cancer Treatment and Tissue Regeneration, Faculty of Medicine & Biomedical Center in Pilsen, Charles University, Pilsen, Czech Republic
| |
Collapse
|
26
|
DeGracia DJ, Taha D, Anggraini FT, Huang ZF. Nonautonomous dynamics of acute cell injury. Phys Rev E 2019; 100:052407. [PMID: 31870014 DOI: 10.1103/physreve.100.052407] [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: 05/13/2018] [Indexed: 06/10/2023]
Abstract
Medical conditions due to acute cell injury, such as stroke and heart attack, are of tremendous impact and have attracted huge amounts of research effort. The biomedical research that seeks cures for these conditions has been dominated by a qualitative, inductive mind-set. Although the inductive approach has not been effective in developing medical treatments, it has amassed enough information to allow construction of quantitative, deductive models of acute cell injury. In this work we develop a modeling approach by extending an autonomous nonlinear dynamic theory of acute cell injury that offered new ways to conceptualize cell injury but possessed limitations that decrease its effectiveness. Here we study the global dynamics of the cell injury theory using a nonautonomous formulation. Different from the standard scenario in nonlinear dynamics that is determined by the steady state and fixed points of the model equations, in this nonautonomous model with a trivial fixed point, the system property is dominated by the transient states and the corresponding dynamic processes. The model gives rise to four qualitative types of dynamical patterns that can be mapped to the behavior of cells after clinical acute injuries. The nonautonomous theory predicts the existence of a latent stress response capacity (LSRC) possessed by injured cells. The LSRC provides a theoretical explanation of how therapies, such as hypothermia, can prevent cell death after lethal injuries. The nonautonomous theory of acute cell injury provides an improved quantitative framework for understanding cell death and recovery and lays a foundation for developing effective therapeutics for acute injury.
Collapse
Affiliation(s)
- Donald J DeGracia
- Department of Physiology, Wayne State University, Detroit, Michigan 48201, USA
| | - Doaa Taha
- Department of Physics and Astronomy, Wayne State University, Detroit, Michigan 48201, USA
| | - Fika Tri Anggraini
- Department of Physiology, Wayne State University, Detroit, Michigan 48201, USA
| | - Zhi-Feng Huang
- Department of Physics and Astronomy, Wayne State University, Detroit, Michigan 48201, USA
| |
Collapse
|
27
|
Symonds P, Jones GDD. Hot Topics in Radiobiology. Clin Oncol (R Coll Radiol) 2019; 31:269-271. [PMID: 30885531 DOI: 10.1016/j.clon.2019.02.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 10/27/2022]
Affiliation(s)
- P Symonds
- Leicester Cancer Research Centre, University of Leicester, Leicester Royal Infirmary, Leicester, UK.
| | - G D D Jones
- Leicester Cancer Research Centre, University of Leicester, Leicester Royal Infirmary, Leicester, UK
| |
Collapse
|