1
|
He L, Lin F, Zhou Y, Dong M, Deng M, Li J, Jia N. Identification of hub genes related to DNA damage response in asthma via combinative bioinformatics strategy. J Int Med Res 2025; 53:3000605251332204. [PMID: 40288048 PMCID: PMC12035324 DOI: 10.1177/03000605251332204] [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: 11/19/2024] [Accepted: 03/14/2025] [Indexed: 04/29/2025] Open
Abstract
ObjectiveEmerging evidence has indicated the potential role of DNA damage response in asthma pathogenesis, but the underlying mechanisms remain elusive. Therefore, this study aimed to identify key diagnostic DNA damage response-related genes in asthma and explore their regulatory networks.MethodsDifferentially expressed genes between healthy individuals and patients with asthma were identified using the Gene Expression Omnibus database. Hub DNA damage response-related differentially expressed genes were determined via protein-protein interaction network and verified through gene expression analysis. Receiver operating characteristic curve was employed to identify diagnostic genes. Transcription factor-microRNA-target gene interactions were analyzed to uncover the regulatory networks in asthma pathogenesis. In this observational study, reverse transcription quantitative polymerase chain reaction was used to validate gene expression levels in healthy individuals and patients with asthma.ResultsSix of the nine hub genes (ATM, PCNA, CUL4A, PARP2, HLTF, and NBN) were identified as key diagnostic genes. These genes may contribute to asthma progression by regulating inflammatory pathways, such as cyclic GMP-AMP synthase-stimulator of interferon genes, senescence-associated secretory phenotype, autophagy, and apoptosis. Three microRNAs and eleven transcription factors were recognized as potential regulators. Reverse transcription quantitative polymerase chain reaction confirmed the downregulation of DNA damage response genes in asthma and revealed distinct expression patterns across different asthma endotypes.ConclusionSix DNA damage response-related genes may serve as diagnostic biomarkers for asthma, and the transcription factor-microRNA-DNA damage response gene network highlights the role of DNA damage response in asthmatic inflammation.
Collapse
Affiliation(s)
- Li He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, P.R. China
- Guangzhou National Laboratory, Guangzhou International Bio Island, China
| | - Fangmei Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, P.R. China
| | - Yawen Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, P.R. China
| | - Meihua Dong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, P.R. China
| | - Mingfang Deng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, P.R. China
| | - Jing Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, P.R. China
- Guangzhou National Laboratory, Guangzhou International Bio Island, China
| | - Nan Jia
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, P.R. China
| |
Collapse
|
2
|
Li F, Zhang Y, Li J, Jiang R, Ci S. NUP98-p65 complex regulates DNA repair to maintain glioblastoma stem cells. FASEB J 2025; 39:e70401. [PMID: 39960447 DOI: 10.1096/fj.202403256r] [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: 12/17/2024] [Revised: 01/26/2025] [Accepted: 02/07/2025] [Indexed: 05/09/2025]
Abstract
The nuclear pore complex (NPC) is an evolutionarily conserved structure that maintains the traffic between the nucleus and cytoplasm. Here, we profiled the expression of nucleoporins (NUPs) in glioblastoma stem cells (GSCs) and found that NUP98 promoted GSC maintenance and therapeutic resistance. GSCs preferentially expressed NUP98, which is essential for GSC tumorigenesis in vitro and in vivo. RNA sequencing demonstrated that NUP98 regulated the expression of key DNA damage and repair pathways. NUP98 formed a complex with transcription factor p65 to directly activate genes involved in homologous repair. Attenuation of NUP98 or p65 expression induced unrepaired intrinsic DNA damage and sensitized GSC to ionizing radiation. Clinically, overexpression of NUP98 informs poor clinical outcome among glioblastoma (GBM) patients. Collectively, our results demonstrate that NUP98-p65 represents a novel node in the regulation of DNA repair, suggesting a therapeutic strategy with potential clinical benefits for GBM patients.
Collapse
Affiliation(s)
- Feifei Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Ying Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Jiahui Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Ranran Jiang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Shusheng Ci
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
3
|
Liu D, Liu L, Che X, Wu G. Discovery of paradoxical genes: reevaluating the prognostic impact of overexpressed genes in cancer. Front Cell Dev Biol 2025; 13:1525345. [PMID: 39911323 PMCID: PMC11794808 DOI: 10.3389/fcell.2025.1525345] [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: 11/09/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Abstract
Oncogenes are typically overexpressed in tumor tissues and often linked to poor prognosis. However, recent advancements in bioinformatics have revealed that many highly expressed genes in tumors are associated with better patient outcomes. These genes, which act as tumor suppressors, are referred to as "paradoxical genes." Analyzing The Cancer Genome Atlas (TCGA) confirmed the widespread presence of paradoxical genes, and KEGG analysis revealed their role in regulating tumor metabolism. Mechanistically, discrepancies between gene and protein expression-affected by pre- and post-transcriptional modifications-may drive this phenomenon. Mechanisms like upstream open reading frames and alternative splicing contribute to these inconsistencies. Many paradoxical genes modulate the tumor immune microenvironment, exerting tumor-suppressive effects. Further analysis shows that the stage- and tumor-specific expression of these genes, along with their environmental sensitivity, influence their dual roles in various signaling pathways. These findings highlight the importance of paradoxical genes in resisting tumor progression and maintaining cellular homeostasis, offering new avenues for targeted cancer therapy.
Collapse
Affiliation(s)
| | | | - Xiangyu Che
- *Correspondence: Guangzhen Wu, ; Xiangyu Che,
| | | |
Collapse
|
4
|
Cen B, Zhang J, Pan X, Xu Z, Li R, Chen C, Wang B, Li Z, Zhang G, Ji A, Yuan Y. Stimuli-Responsive Peptide/siRNA Nanoparticles as a Radiation Sensitizer for Glioblastoma Treatment by Co-Inhibiting RELA/P65 and EGFR. Int J Nanomedicine 2024; 19:11517-11537. [PMID: 39539970 PMCID: PMC11559232 DOI: 10.2147/ijn.s483252] [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: 06/18/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Purpose To develop a novel approach for increasing radiosensitivity in glioblastoma (GBM) by using targeted nanoparticles to deliver siRNA aimed at silencing the EGFR and RELA/P65 genes, which are implicated in radioresistance. Patients and Methods We engineered biodegradable, tumor-targeted, self-assembled, and stimuli-responsive peptide nanoparticles for efficient siRNA delivery. We evaluated the nanoparticles' ability to induce gene silencing and enhance DNA damage under radiation in vitro and in vivo. The nanoparticles were designed to exhibit pH-responsive endosomal escape and αvβ3 integrin targeting, allowing for preferential accumulation at tumor sites and traversal of the blood-brain tumor barrier. Results The application of these nanoparticles resulted in significant gene silencing, increased apoptosis, and decreased cell viability. The treatment impaired DNA repair mechanisms, thereby enhancing radiosensitivity in GBM cells. In a GBM mouse model, the combination of nanoparticle treatment with radiotherapy notably prolonged survival without apparent toxicity. Conclusion Our findings suggest that nanoparticle-mediated dual gene silencing can effectively overcome GBM radioresistance. This strategy has the potential to improve clinical outcomes in GBM treatment, proposing a promising therapeutic avenue for this challenging malignancy.
Collapse
Affiliation(s)
- Bohong Cen
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510095, People’s Republic of China
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
| | - Jian Zhang
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510095, People’s Republic of China
| | - Xinghua Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, Guangdong, 510515, People’s Republic of China
| | - Zhongyuan Xu
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
| | - Rong Li
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510095, People’s Republic of China
| | - Chengcong Chen
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510095, People’s Republic of China
| | - Baiyao Wang
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510095, People’s Republic of China
| | - Zhiyong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, People’s Republic of China
| | - Guoqian Zhang
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510095, People’s Republic of China
| | - Aimin Ji
- Department of Pharmacy, The Seventh Affiliated Hospital of Southern Medical University, Foshan, Guangdong, 528244, People’s Republic of China
| | - Yawei Yuan
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510095, People’s Republic of China
| |
Collapse
|
5
|
Deng Q, Qiang J, Liu C, Ding J, Tu J, He X, Xia J, Peng X, Li S, Chen X, Ma W, Zhang L, Jiang Y, Shao Z, Chen C, Liu S, Xu J, Zhang L. SOSTDC1 Nuclear Translocation Facilitates BTIC Maintenance and CHD1-Mediated HR Repair to Promote Tumor Progression and Olaparib Resistance in TNBC. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306860. [PMID: 38864559 PMCID: PMC11304230 DOI: 10.1002/advs.202306860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 05/01/2024] [Indexed: 06/13/2024]
Abstract
Breast tumor-initiating cells (BTICs) of triple-negative breast cancer (TNBC) tissues actively repair DNA and are resistant to treatments including chemotherapy, radiotherapy, and targeted therapy. Herein, it is found that a previously reported secreted protein, sclerostin domain containing 1 (SOSTDC1), is abundantly expressed in BTICs of TNBC cells and positively correlated with a poor patient prognosis. SOSTDC1 knockdown impairs homologous recombination (HR) repair, BTIC maintenance, and sensitized bulk cells and BTICs to Olaparib. Mechanistically, following Olaparib treatment, SOSTDC1 translocates to the nucleus in an importin-α dependent manner. Nuclear SOSTDC1 interacts with the N-terminus of the nucleoprotein, chromatin helicase DNA-binding factor (CHD1), to promote HR repair and BTIC maintenance. Furthermore, nuclear SOSTDC1 bound to β-transducin repeat-containing protein (β-TrCP) binding motifs of CHD1 is found, thereby blocking the β-TrCP-CHD1 interaction and inhibiting β-TrCP-mediated CHD1 ubiquitination and degradation. Collectively, these findings identify a novel nuclear SOSTDC1 pathway in regulating HR repair and BTIC maintenance, providing insight into the TNBC therapeutic strategies.
Collapse
Affiliation(s)
- Qiaodan Deng
- Fudan University Shanghai Cancer Center & Institutes of Biomedical SciencesState Key Laboratory of Genetic EngineeringCancer InstitutesKey Laboratory of Breast Cancer in ShanghaiThe Shanghai Key Laboratory of Medical EpigeneticsShanghai Key Laboratory of Radiation OncologyThe International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Jiankun Qiang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical SciencesState Key Laboratory of Genetic EngineeringCancer InstitutesKey Laboratory of Breast Cancer in ShanghaiThe Shanghai Key Laboratory of Medical EpigeneticsShanghai Key Laboratory of Radiation OncologyThe International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Research Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghai200120China
| | - Cuicui Liu
- Department of Breast SurgeryShanghai Cancer Center and Cancer InstituteFudan UniversityShanghai200032P. R. China
| | - Jiajun Ding
- Department of ThyroidBreast and Vascular SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'an710032P. R. China
| | - Juchuanli Tu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical SciencesState Key Laboratory of Genetic EngineeringCancer InstitutesKey Laboratory of Breast Cancer in ShanghaiThe Shanghai Key Laboratory of Medical EpigeneticsShanghai Key Laboratory of Radiation OncologyThe International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Xueyan He
- Fudan University Shanghai Cancer Center & Institutes of Biomedical SciencesState Key Laboratory of Genetic EngineeringCancer InstitutesKey Laboratory of Breast Cancer in ShanghaiThe Shanghai Key Laboratory of Medical EpigeneticsShanghai Key Laboratory of Radiation OncologyThe International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Jie Xia
- Fudan University Shanghai Cancer Center & Institutes of Biomedical SciencesState Key Laboratory of Genetic EngineeringCancer InstitutesKey Laboratory of Breast Cancer in ShanghaiThe Shanghai Key Laboratory of Medical EpigeneticsShanghai Key Laboratory of Radiation OncologyThe International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Xilei Peng
- Fudan University Shanghai Cancer Center & Institutes of Biomedical SciencesState Key Laboratory of Genetic EngineeringCancer InstitutesKey Laboratory of Breast Cancer in ShanghaiThe Shanghai Key Laboratory of Medical EpigeneticsShanghai Key Laboratory of Radiation OncologyThe International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Siqin Li
- Fudan University Shanghai Cancer Center & Institutes of Biomedical SciencesState Key Laboratory of Genetic EngineeringCancer InstitutesKey Laboratory of Breast Cancer in ShanghaiThe Shanghai Key Laboratory of Medical EpigeneticsShanghai Key Laboratory of Radiation OncologyThe International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Xian Chen
- Fudan University Shanghai Cancer Center & Institutes of Biomedical SciencesState Key Laboratory of Genetic EngineeringCancer InstitutesKey Laboratory of Breast Cancer in ShanghaiThe Shanghai Key Laboratory of Medical EpigeneticsShanghai Key Laboratory of Radiation OncologyThe International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Wei Ma
- Fudan University Shanghai Cancer Center & Institutes of Biomedical SciencesState Key Laboratory of Genetic EngineeringCancer InstitutesKey Laboratory of Breast Cancer in ShanghaiThe Shanghai Key Laboratory of Medical EpigeneticsShanghai Key Laboratory of Radiation OncologyThe International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Lu Zhang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical SciencesState Key Laboratory of Genetic EngineeringCancer InstitutesKey Laboratory of Breast Cancer in ShanghaiThe Shanghai Key Laboratory of Medical EpigeneticsShanghai Key Laboratory of Radiation OncologyThe International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Yi‐Zhou Jiang
- Department of Breast SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Key Laboratory of Breast Cancer in ShanghaiDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Zhi‐Ming Shao
- Department of Breast SurgeryFudan University Shanghai Cancer CenterShanghai200032China
- Key Laboratory of Breast Cancer in ShanghaiDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyKunming650201China
- Academy of Biomedical Engineering & The Third Affiliated HospitalKunming Medical UniversityKunming650118China
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical SciencesState Key Laboratory of Genetic EngineeringCancer InstitutesKey Laboratory of Breast Cancer in ShanghaiThe Shanghai Key Laboratory of Medical EpigeneticsShanghai Key Laboratory of Radiation OncologyThe International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Jiangsu Key Lab of Cancer BiomarkersPrevention and TreatmentCollaborative Innovation Center for Cancer MedicineNanjing Medical UniversityNanjing211166China
| | - Jiahui Xu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical SciencesState Key Laboratory of Genetic EngineeringCancer InstitutesKey Laboratory of Breast Cancer in ShanghaiThe Shanghai Key Laboratory of Medical EpigeneticsShanghai Key Laboratory of Radiation OncologyThe International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Lixing Zhang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical SciencesState Key Laboratory of Genetic EngineeringCancer InstitutesKey Laboratory of Breast Cancer in ShanghaiThe Shanghai Key Laboratory of Medical EpigeneticsShanghai Key Laboratory of Radiation OncologyThe International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyShanghai Medical CollegeFudan UniversityShanghai200032China
| |
Collapse
|
6
|
Sellars E, Savguira M, Wu J, Cancelliere S, Jen M, Krishnan R, Hakem A, Barsyte-Lovejoy D, Hakem R, Narod SA, Kotsopoulos J, Salmena L. A high-throughput approach to identify BRCA1-downregulating compounds to enhance PARP inhibitor sensitivity. iScience 2024; 27:110180. [PMID: 38993666 PMCID: PMC11238136 DOI: 10.1016/j.isci.2024.110180] [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: 11/09/2023] [Revised: 04/29/2024] [Accepted: 06/01/2024] [Indexed: 07/13/2024] Open
Abstract
PARP inhibitors (PARPi) are efficacious in BRCA1-null tumors; however, their utility is limited in tumors with functional BRCA1. We hypothesized that pharmacologically reducing BRCA1 protein levels could enhance PARPi effectiveness in BRCA1 wild-type tumors. To identify BRCA1 downregulating agents, we generated reporter cell lines using CRISPR-mediated editing to tag endogenous BRCA1 protein with HiBiT. These reporter lines enable the sensitive measurement of BRCA1 protein levels by luminescence. Validated reporter cells were used in a pilot screen of epigenetic-modifying probes and a larger screen of more than 6,000 compounds. We identified 7 compounds that could downregulate BRCA1-HiBiT expression and synergize with olaparib. Three compounds, N-acetyl-N-acetoxy chlorobenzenesulfonamide (NANAC), A-443654, and CHIR-124, were validated to reduce BRCA1 protein levels and sensitize breast cancer cells to the toxic effects of olaparib. These results suggest that BRCA1-HiBiT reporter cells hold promise in developing agents to improve the clinical utility of PARPi.
Collapse
Affiliation(s)
- Erin Sellars
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Women's College Research Institute, Women's College Hospital, Toronto, ON M5S 1B2, Canada
| | - Margarita Savguira
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jie Wu
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sabrina Cancelliere
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Mark Jen
- Lunenfeld-Tanenbaum Research Institute, Network Biology Collaborative Centre, High-Throughput Screening, Mt. Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Rehna Krishnan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Anne Hakem
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Dalia Barsyte-Lovejoy
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Structural Genomics Consortium, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Razqallah Hakem
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Steven A Narod
- Women's College Research Institute, Women's College Hospital, Toronto, ON M5S 1B2, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Joanne Kotsopoulos
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Women's College Research Institute, Women's College Hospital, Toronto, ON M5S 1B2, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Leonardo Salmena
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Women's College Research Institute, Women's College Hospital, Toronto, ON M5S 1B2, Canada
| |
Collapse
|
7
|
Soni D, Anjum Z, Raza K, Verma S. A Review on Picrosides Targeting NFκB and its Proteins for Treatment of Breast Cancer. Cell Biochem Biophys 2024; 82:575-591. [PMID: 38724755 DOI: 10.1007/s12013-024-01281-1] [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] [Accepted: 04/17/2024] [Indexed: 08/25/2024]
Abstract
Breast cancer is the most frequently diagnosed disease causing most deaths in women worldwide. Chemotherapy and neo-adjuvant therapy are the standard method of treatment in early stages of breast cancer. However drug resistance in breast cancer limit the use of these methods for treatment. Research focus is now shifted towards identifying natural phytochemicals with lower toxicity. This review illustrates the NF κB interaction with different signaling pathways in normal condition, breast cancer and other cancer and thus represent a potential target for treatment. No reports are available on the action of picrosides on NFκB and its associated proteins for anticancer activity. In the present review, potential interaction of picrosides with NF-κB and its associated proteins is reviewed for anticancer action. Further, an important facet of this review entails the ADMET analysis of Picroside, elucidating key ADMET properties which serves to underscore the crucial characteristics of Picroside as a potential drug for treating breast cancer. Furthermore, in silico analysis of Picrosides was executed in order to get potential binding modes between ligand (Picrosides II) and NFκB.
Collapse
Affiliation(s)
- Deepika Soni
- National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Zubina Anjum
- Department of Bioinformatics, Central University of South Bihar, Gaya, Bihar, India
| | - Khalid Raza
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Saurabh Verma
- Indian Council of Medical Research, HRD Division, V. Ramalingaswami Bhawan, Ansari Nagar, New Delhi, India.
| |
Collapse
|
8
|
Li C, Huang Y, Yi X, Tang Y, Okita R, He J. Pan-cancer prognostic model and immune microenvironment analysis of natural killer cell-related genes. Transl Cancer Res 2024; 13:1936-1953. [PMID: 38737690 PMCID: PMC11082681 DOI: 10.21037/tcr-24-434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 04/15/2024] [Indexed: 05/14/2024]
Abstract
Background Natural killer (NK) cells play a significant role in antitumor immunity and are closely related to tumor prognosis and recurrence. NK cell-based tumor immunotherapy, including immune checkpoint inhibition and CAR-engineered NK cells, is a promising area of research. However, there is a need for better NK cell-related models and associated biomarkers. Methods The sequences of NK cell-related genes were obtained from the published NK cell CRISPR/Cas9 library data, and the common genes were selected as NK cell-related genes. The RNA sequencing (RNA-seq) and clinical data of 32 solid tumors from The Cancer Genome Atlas (TCGA) were downloaded from the UCSC Xena database, and the RNA-seq data of normal samples were downloaded from the Genotype-Tissue Expression (GTEx) database. The differentially expressed NK cell-related genes (DENKGs) between the tumor and normal samples were analyzed. The DENKGs related to the prognosis of solid tumors were selected via univariate Cox analysis, and 32 kinds of solid tumor prognostic models were constructed using least absolute shrinkage and selection operator (LASSO) and multivariate Cox analysis. Survival, receiver operating characteristic (ROC), and independent prognostic analyses were employed to test the effectiveness of the model, along with a nomogram model and prediction curve. Differences in the immune pathways and microenvironment cells were analyzed between the high- and low-risk groups identified by the model. Results We constructed a pan-cancer prognostic model with 63 NK cell-related genes and further identified DEPDC1 and ASPM as potentially offering new directions in tumor research by literature screening. Conclusions In this study, 63 prognostic solid tumor markers were investigated using NK cell-related genes, and for the first time, a pan-cancer prognostic model was constructed to analyze their role in the immune microenvironment, which may contribute new insights into tumor research.
Collapse
Affiliation(s)
- Caihong Li
- Department of Radiotherapy, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yuxin Huang
- Department of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Xiaojuan Yi
- Department of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Youpan Tang
- Department of Gastroenterology, Zhongjiang People’s Hospital, Deyang, China
| | - Riki Okita
- Department of Thoracic Surgery, National Hospital Organization Yamaguchi Ube Medical Center, Ube, Japan
| | - Jun He
- Department of Oncology, The Third Hospital of Mian Yang (Sichuan Mental Health Center), Mianyang, China
| |
Collapse
|
9
|
Zhang B, Li N, Gao J, Zhao Y, Jiang J, Xie S, Zhang C, Zhang Q, Liu L, Wang Z, Ji D, Wu L, Ren R. Targeting of focal adhesion kinase enhances the immunogenic cell death of PEGylated liposome doxorubicin to optimize therapeutic responses of immune checkpoint blockade. J Exp Clin Cancer Res 2024; 43:51. [PMID: 38373953 PMCID: PMC10875809 DOI: 10.1186/s13046-024-02974-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 02/03/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUNDS Immune checkpoint blockade (ICB) is widely considered to exert long-term treatment benefits by activating antitumor immunity. However, many cancer patients show poor clinical responses to ICB due in part to the lack of an immunogenic niche. Focal adhesion kinase (FAK) is frequently amplified and acts as an immune modulator across cancer types. However, evidence illustrates that targeting FAK is most effective in combination therapy rather than in monotherapy. METHODS Here, we used drug screening, in vitro and in vivo assays to filter out that doxorubicin and its liposomal form pegylated liposome doxorubicin (PLD) showed synergistic anti-tumor effects in combination with FAK inhibitor IN10018. We hypothesized that anti-tumor immunity and immunogenic cell death (ICD) may be involved in the treatment outcomes through the data analysis of our clinical trial testing the combination of IN10018 and PLD. We then performed cell-based assays and animal studies to detect whether FAK inhibition by IN10018 can boost the ICD of PLD/doxorubicin and further established syngeneic models to test the antitumor effect of triplet combination of PLD, IN10018, and ICB. RESULTS We demonstrated that the combination of FAK inhibitor IN10018, and PLD/doxorubicin exerted effective antitumor activity. Notably, the doublet combination regimen exhibited response latency and long-lasting treatment effects clinically, outcomes frequently observed in immunotherapy. Our preclinical study confirmed that the 2-drug combination can maximize the ICD of cancer cells. This approach primed the tumor microenvironment, supplementing it with sufficient tumor-infiltrating lymphocytes (TILs) to activate antitumor immunity. Finally, different animal studies confirmed that the antitumor effects of ICB can be significantly enhanced by this doublet regimen. CONCLUSIONS We confirmed that targeting FAK by IN10018 can enhance the ICD of PLD/doxorubicin, further benefiting the anti-tumor effect of ICB. The animal tests of the triplet regimen warrant further discovery in the real world.
Collapse
Affiliation(s)
- Baoyuan Zhang
- State Key Laboratory for Medical Genomics, Collaborative Innovation Center of Hematology, Shanghai Institute of HematologyNational Research Center for Translational MedicineRuijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning Li
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinses Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaming Gao
- State Key Laboratory for Medical Genomics, Collaborative Innovation Center of Hematology, Shanghai Institute of HematologyNational Research Center for Translational MedicineRuijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuxi Zhao
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinses Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Jiang
- InxMed (Shanghai) Co., Ltd, Beijing, China
| | - Shuang Xie
- InxMed (Shanghai) Co., Ltd, Beijing, China
| | - Cuiping Zhang
- Department of Pathology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Qingyu Zhang
- Laboratory of Obstetrics and Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Leo Liu
- InxMed (Shanghai) Co., Ltd, Beijing, China
| | - Zaiqi Wang
- InxMed (Shanghai) Co., Ltd, Beijing, China
| | - Dongmei Ji
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Lingying Wu
- Department of Gynecologic Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinses Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Ruibao Ren
- State Key Laboratory for Medical Genomics, Collaborative Innovation Center of Hematology, Shanghai Institute of HematologyNational Research Center for Translational MedicineRuijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- International Center for Aging and Cancer, Hainan Medical University, Hainan Province, Haikou, China.
| |
Collapse
|
10
|
Lee DH, Imran M, Choi JH, Park YJ, Kim YH, Min S, Park TJ, Choi YW. CDK4/6 inhibitors induce breast cancer senescence with enhanced anti-tumor immunogenic properties compared with DNA-damaging agents. Mol Oncol 2024; 18:216-232. [PMID: 37854019 PMCID: PMC10766199 DOI: 10.1002/1878-0261.13541] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/05/2023] [Accepted: 10/17/2023] [Indexed: 10/20/2023] Open
Abstract
Since therapy-induced senescence (TIS) can either support or inhibit cancer progression, identifying which types of chemotherapeutic agents can produce the strongest anti-tumor TIS is an important issue. Here, cyclin-dependent kinase4/6 inhibitors (CDK4/6i)-induced senescence was compared to the TIS induced by conventional DNA-damaging agents. Despite both types of agents eliciting a similar degree of senescence, we observed increased expression of the senescence-associated secretory phenotype (SASP) and ligands related to pro-tumor immunity (IL6, CXCL8, TGFβ, CD274, and CEACAM1) and angiogenesis (VEGFA) mainly in TIS induced by DNA-damaging agents rather than by CDK4/6i. Additionally, although all agents increased the expression of anti-tumor immunomodulatory proteins related to antigen presentation (MHC-I, B2M) and T cell chemokines (CXCL9, 10, 11), CDK4/6i-induced senescent cells still maintained this expression at a similar or even higher intensity than cells treated with DNA-damaging agents, despite the absence of nuclear factor-kappa-B (NF-κB) and p53 activation. These data suggest that in contrast with DNA-damaging agents, which augment the pro-tumorigenic microenvironment via pro-inflammatory SASP, CDK4/6i can generate TIS only with antitumor immunomodulatory proteins.
Collapse
Affiliation(s)
- Dong Hyun Lee
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwonKorea
- Department of Biomedical SciencesAjou University Graduate School of MedicineSuwonKorea
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
| | - Muhammad Imran
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwonKorea
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
| | - Jae Ho Choi
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
- Department of Hematology‐OncologyAjou University School of MedicineSuwonKorea
| | - Yoo Jung Park
- Department of Hematology‐OncologyAjou University School of MedicineSuwonKorea
| | - Young Hwa Kim
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
| | - Sunwoo Min
- Department of Biological SciencesKorea Advanced Institute of Science and Technology (KAIST)DaejeonKorea
| | - Tae Jun Park
- Department of Biochemistry and Molecular BiologyAjou University School of MedicineSuwonKorea
- Department of Biomedical SciencesAjou University Graduate School of MedicineSuwonKorea
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
| | - Yong Won Choi
- Inflamm‐Aging Translational Research CenterAjou University Medical CenterSuwonKorea
- Department of Hematology‐OncologyAjou University School of MedicineSuwonKorea
| |
Collapse
|
11
|
Handwerk L, Schreier HK, Kraft D, Shreder K, Hemmersbach R, Hauslage J, Bonig H, Wiesmüller L, Fournier C, Rall-Scharpf M. Simulating Space Conditions Evokes Different DNA Damage Responses in Immature and Mature Cells of the Human Hematopoietic System. Int J Mol Sci 2023; 24:13761. [PMID: 37762064 PMCID: PMC10531023 DOI: 10.3390/ijms241813761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
The impact of space radiation and microgravity on DNA damage responses has been discussed controversially, largely due to the variety of model systems engaged. Here, we performed side-by-side analyses of human hematopoietic stem/progenitor cells (HSPC) and peripheral blood lymphocytes (PBL) cultivated in a 2D clinostat to simulate microgravity before, during and after photon and particle irradiation. We demonstrate that simulated microgravity (SMG) accelerates the early phase of non-homologous end joining (NHEJ)-mediated repair of simple, X-ray-induced DNA double-strand breaks (DSBs) in PBL, while repair kinetics in HSPC remained unaltered. Repair acceleration was lost with increasing LET of ion exposures, which increases the complexity of DSBs, precluding NHEJ and requiring end resection for successful repair. Such cell-type specific effect of SMG on DSB repair was dependent on the NF-кB pathway pre-activated in PBL but not HSPC. Already under unperturbed growth conditions HSPC and PBL suffered from SMG-induced replication stress associated with accumulation of single-stranded DNA and DSBs, respectively. We conclude that in PBL, SMG-induced DSBs promote repair of radiation-induced damage in an adaptive-like response. HSPC feature SMG-induced single-stranded DNA and FANCD2 foci, i.e., markers of persistent replication stress and senescence that may contribute to a premature decline of the immune system in space.
Collapse
Affiliation(s)
- Leonie Handwerk
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, 64291 Darmstadt, Germany
| | | | - Daniela Kraft
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, 64291 Darmstadt, Germany
| | - Kateryna Shreder
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, 64291 Darmstadt, Germany
| | - Ruth Hemmersbach
- Department of Gravitational Biology, German Aerospace Center, Institute of Aerospace Medicine, 51147 Cologne, Germany; (R.H.)
| | - Jens Hauslage
- Department of Gravitational Biology, German Aerospace Center, Institute of Aerospace Medicine, 51147 Cologne, Germany; (R.H.)
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology, Johann Wolfgang Goethe-University Hospital, and German Red Cross Blood Service, Baden-Wuerttemberg–Hessen, 60528 Frankfurt, Germany
| | - Lisa Wiesmüller
- Department of Obstetrics and Gynecology, Ulm University, 89075 Ulm, Germany
| | - Claudia Fournier
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, 64291 Darmstadt, Germany
| | | |
Collapse
|
12
|
Marion W, Koppe T, Chen CC, Wang D, Frenis K, Fierstein S, Sensharma P, Aumais O, Peters M, Ruiz-Torres S, Chihanga T, Boettcher S, Shimamura A, Bauer DE, Schlaeger T, Wells SI, Ebert BL, Starczynowski D, da Rocha EL, Rowe RG. RUNX1 mutations mitigate quiescence to promote transformation of hematopoietic progenitors in Fanconi anemia. Leukemia 2023; 37:1698-1708. [PMID: 37391485 PMCID: PMC11009868 DOI: 10.1038/s41375-023-01945-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 05/25/2023] [Accepted: 06/13/2023] [Indexed: 07/02/2023]
Abstract
Many inherited bone marrow failure syndromes (IBMFSs) present a high risk of transformation to myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). During transformation of IBMFSs, hematopoietic stem and progenitor cells (HSPCs) with poor fitness gain ectopic, dysregulated self-renewal secondary to somatic mutations via undefined mechanisms. Here, in the context of the prototypical IBMFS Fanconi anemia (FA), we performed multiplexed gene editing of mutational hotspots in MDS-associated genes in human induced pluripotent stem cells (iPSCs) followed by hematopoietic differentiation. We observed aberrant self-renewal and impaired differentiation of HSPCs with enrichment of RUNX1 insertions and deletions (indels), generating a model of IBMFS-associated MDS. We observed that compared to the failure state, FA MDS cells show mutant RUNX1-mediated blunting of the G1/S cell cycle checkpoint that is normally activated in FA in response to DNA damage. RUNX1 indels also lead to activation of innate immune signaling, which stabilizes the homologous recombination (HR) effector BRCA1, and this pathway can be targeted to abrogate viability and restore sensitivity to genotoxins in FA MDS. Together, these studies develop a paradigm for modeling clonal evolution in IBMFSs, provide basic understanding of the pathogenesis of MDS, and uncover a therapeutic target in FA-associated MDS.
Collapse
Affiliation(s)
- William Marion
- Department of Hematology-Oncology, Boston Children's Hospital, Boston, MA, USA
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - Tiago Koppe
- Department of Hematology-Oncology, Boston Children's Hospital, Boston, MA, USA
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - Chun-Chin Chen
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dahai Wang
- Department of Hematology-Oncology, Boston Children's Hospital, Boston, MA, USA
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - Katie Frenis
- Department of Hematology-Oncology, Boston Children's Hospital, Boston, MA, USA
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - Sara Fierstein
- Department of Hematology-Oncology, Boston Children's Hospital, Boston, MA, USA
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - Prerana Sensharma
- Department of Hematology-Oncology, Boston Children's Hospital, Boston, MA, USA
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - Olivia Aumais
- Department of Hematology-Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Michael Peters
- Department of Hematology-Oncology, Boston Children's Hospital, Boston, MA, USA
| | | | | | - Steffen Boettcher
- Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital of Zurich, Zurich, Switzerland
| | - Akiko Shimamura
- Department of Hematology-Oncology, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Daniel E Bauer
- Department of Hematology-Oncology, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Susanne I Wells
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Benjamin L Ebert
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Daniel Starczynowski
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati Cancer Center, Cincinnati, OH, USA
| | | | - R Grant Rowe
- Department of Hematology-Oncology, Boston Children's Hospital, Boston, MA, USA.
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Pavitra E, Kancharla J, Gupta VK, Prasad K, Sung JY, Kim J, Tej MB, Choi R, Lee JH, Han YK, Raju GSR, Bhaskar L, Huh YS. The role of NF-κB in breast cancer initiation, growth, metastasis, and resistance to chemotherapy. Biomed Pharmacother 2023; 163:114822. [PMID: 37146418 DOI: 10.1016/j.biopha.2023.114822] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/27/2023] [Accepted: 04/30/2023] [Indexed: 05/07/2023] Open
Abstract
Breast cancer (BC) is the second most fatal disease and is the prime cause of cancer allied female deaths. BC is caused by aberrant tumor suppressor genes and oncogenes regulated by transcription factors (TFs) like NF-κB. NF-κB is a pro-inflammatory TF that crucially alters the expressions of various genes associated with inflammation, cell progression, metastasis, and apoptosis and modulates a network of genes that underlie tumorigenesis. Herein, we focus on NF-κB signaling pathways, its regulators, and the rationale for targeting NF-κB. This review also includes TFs that maintain NF-κB crosstalk and their roles in promoting angiogenesis and metastasis. In addition, we discuss the importance of combination therapies, resistance to treatment, and potential novel therapeutic strategies including nanomedicine that targets NF-κB.
Collapse
Affiliation(s)
- Eluri Pavitra
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea; 3D Convergence Center, Inha University, Incheon 22212, Republic of Korea
| | - Jyothsna Kancharla
- Department of Bioscience and Biotechnology, Banasthali University, Vanasthali, Rajasthan 304022, India
| | - Vivek Kumar Gupta
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Kiran Prasad
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur- 495009, Chhattisgarh, India
| | - Ju Yong Sung
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Jigyeong Kim
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Mandava Bhuvan Tej
- Department of Health care informatics, Sacred Heart University, 5151Park Avenue, Fair fields, CT06825, USA
| | - Rino Choi
- 3D Convergence Center, Inha University, Incheon 22212, Republic of Korea; Department of Materials Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Jeong-Hwan Lee
- 3D Convergence Center, Inha University, Incheon 22212, Republic of Korea; Department of Materials Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Young-Kyu Han
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul 04620, Republic of Korea
| | - Ganji Seeta Rama Raju
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul 04620, Republic of Korea.
| | - Lvks Bhaskar
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur- 495009, Chhattisgarh, India.
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea.
| |
Collapse
|
14
|
Pang HH, Huang CY, Chen PY, Li NS, Hsu YP, Wu JK, Fan HF, Wei KC, Yang HW. Bioengineered Bacteriophage-Like Nanoparticles as RNAi Therapeutics to Enhance Radiotherapy against Glioblastomas. ACS NANO 2023; 17:10407-10422. [PMID: 37120837 DOI: 10.1021/acsnano.3c01102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Since glioblastomas (GBMs) are radioresistant malignancies and most GBM recurrences occur in radiotherapy, increasing the effectiveness of radiotherapy by gene-silencing has recently attracted attention. However, the difficulty in precisely tuning the composition and RNA loading in nanoparticles leads to batch-to-batch variations of the RNA therapeutics, thus significantly restricting their clinical translation. Here, we bioengineer bacteriophage Qβ particles with a designed broccoli light-up three-way junction (b-3WJ) RNA scaffold (contains two siRNA/miRNA sequences and one light-up aptamer) packaging for the silencing of genes in radioresistant GBM cells. The in vitro results demonstrate that the cleavage of de novo designed b-3WJ RNA by Dicer enzyme can be easily monitored in real-time using fluorescence microscopy, and the TrQβ@b-3WJLet-7gsiEGFR successfully knocks down EGFR and IKKα simultaneously and thereby inactivates NF-κB signaling to inhibit DNA repair. Delivery of TrQβ@b-3WJLet-7gsiEGFR through convection-enhanced delivery (CED) infusion followed by 2Gy X-ray irradiation demonstrated that the median survival was prolonged to over 60 days compared with the 2Gy X-ray irradiated group (median survival: 31 days). Altogether, the results of this study could be critical for the design of RNAi-based genetic therapeutics, and CED infusion serves as a powerful delivery system for promoting radiotherapy against GBMs without evidence of systemic toxicity.
Collapse
Affiliation(s)
- Hao-Han Pang
- Department of Biomedical Engineering, National Cheng Kung University, No. 1, University Rd., Tainan 70101, Taiwan
| | - Chiung-Yin Huang
- Department of Neurosurgery, Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou, 5 Fuxing St., Guishan Dist., Taoyuan 33305, Taiwan
| | - Pin-Yuan Chen
- Department of Neurosurgery, Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou, 5 Fuxing St., Guishan Dist., Taoyuan 33305, Taiwan
- School of Medicine, Chang Gung University, 259 Wenhua 1st Rd., Guishan Dist., Taoyuan 33302, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital, Keelung, 222 Maijin Rd., Keelung 20401, Taiwan
| | - Nan-Si Li
- Department of Biomedical Engineering, National Cheng Kung University, No. 1, University Rd., Tainan 70101, Taiwan
| | - Ying-Pei Hsu
- Department of Biomedical Engineering, National Cheng Kung University, No. 1, University Rd., Tainan 70101, Taiwan
- Department of Materials and Optoelectronic Science, National Sun Yat-sen University, 70 Lienhai Rd., Kaohsiung 80424, Taiwan
| | - Jan-Kai Wu
- Department of Chemistry, National Sun Yat-sen University, 70 Lienhai Rd., Kaohsiung 80424, Taiwan
| | - Hsiu-Fang Fan
- Department of Chemistry, National Sun Yat-sen University, 70 Lienhai Rd., Kaohsiung 80424, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, 70 Lienhai Rd., Kaohsiung 80424, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou, 5 Fuxing St., Guishan Dist., Taoyuan 33305, Taiwan
- School of Medicine, Chang Gung University, 259 Wenhua 1st Rd., Guishan Dist., Taoyuan 33302, Taiwan
- Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, 6, Sec 2, JunCheng Rd., New Taipei City 23652, Taiwan
| | - Hung-Wei Yang
- Department of Biomedical Engineering, National Cheng Kung University, No. 1, University Rd., Tainan 70101, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, No. 1, University Rd., Tainan 70101, Taiwan
| |
Collapse
|
15
|
cGAS-STING signalling in cancer: striking a balance with chromosomal instability. Biochem Soc Trans 2023; 51:539-555. [PMID: 36876871 DOI: 10.1042/bst20220838] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 03/07/2023]
Abstract
Chromosomal instability (CIN) is a hallmark of cancer that drives tumour evolution. It is now recognised that CIN in cancer leads to the constitutive production of misplaced DNA in the form of micronuclei and chromatin bridges. These structures are detected by the nucleic acid sensor cGAS, leading to the production of the second messenger 2'3'-cGAMP and activation of the critical hub of innate immune signalling STING. Activation of this immune pathway should instigate the influx and activation of immune cells, resulting in the eradication of cancer cells. That this does not universally occur in the context of CIN remains an unanswered paradox in cancer. Instead, CIN-high cancers are notably adept at immune evasion and are highly metastatic with typically poor outcomes. In this review, we discuss the diverse facets of the cGAS-STING signalling pathway, including emerging roles in homeostatic processes and their intersection with genome stability regulation, its role as a driver of chronic pro-tumour inflammation, and crosstalk with the tumour microenvironment, which may collectively underlie its apparent maintenance in cancers. A better understanding of the mechanisms whereby this immune surveillance pathway is commandeered by chromosomally unstable cancers is critical to the identification of new vulnerabilities for therapeutic exploitation.
Collapse
|
16
|
Salazar-Valdivia FE, Valdez-Cornejo VA, Ulloque-Badaracco JR, Hernandez-Bustamante EA, Alarcón-Braga EA, Mosquera-Rojas MD, Garrido-Matta DP, Herrera-Añazco P, Benites-Zapata VA, Hernandez AV. Systemic Immune-Inflammation Index and Mortality in Testicular Cancer: A Systematic Review and Meta-Analysis. Diagnostics (Basel) 2023; 13:843. [PMID: 36899987 PMCID: PMC10000460 DOI: 10.3390/diagnostics13050843] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/25/2023] Open
Abstract
The systemic immune-inflammation index (SIII) is a marker studied in multiple types of urologic cancer. This systematic review evaluates the association between SIII values with overall survival (OS) and progression-free survival (PFS) in testicular cancer. We searched observational studies in five databases. The quantitative synthesis was performed using a random-effects model. The risk of bias was assessed using the Newcastle-Ottawa Scale (NOS). The only measure of the effect was the hazard ratio (HR). A sensitivity analysis was performed according to the risk of bias in the studies. There were 833 participants in a total of 6 cohorts. We found that high SIII values were associated with worse OS (HR = 3.28; 95% CI 1.3-8.9; p < 0.001; I2 = 78) and PFS (HR = 3.9; 95% CI 2.53-6.02; p < 0.001; I2 = 0). No indication of small study effects was found in the association between SIII values and OS (p = 0.5301). High SIII values were associated with worse OS and PFS. However, further primary studies are suggested to enhance the effect of this marker in different outcomes of testicular cancer patients.
Collapse
Affiliation(s)
- Farley E. Salazar-Valdivia
- Escuela de Medicina, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
- Sociedad Científica de Estudiantes de Medicina de la Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
| | - Valeria A. Valdez-Cornejo
- Escuela de Medicina, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
- Sociedad Científica de Estudiantes de Medicina de la Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
| | | | - Enrique A. Hernandez-Bustamante
- Sociedad Científica de Estudiantes de Medicina de la Universidad Nacional de Trujillo, Trujillo 13011, Peru
- Grupo Peruano de Investigación Epidemiológica, Unidad para la Generación y Síntesis de Evidencias en Salud, Universidad San Ignacio de Loyola, Lima 15012, Peru
| | - Esteban A. Alarcón-Braga
- Escuela de Medicina, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
- Sociedad Científica de Estudiantes de Medicina de la Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
| | - Melany D. Mosquera-Rojas
- Escuela de Medicina, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
- Sociedad Científica de Estudiantes de Medicina de la Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
| | | | - Percy Herrera-Añazco
- Escuela de Medicina, Universidad Privada San Juan Bautista, Lima 15067, Peru
- Universidad Privada del Norte, Trujillo 13011, Peru
| | - Vicente A. Benites-Zapata
- Unidad de Investigación para la Generación y Síntesis de Evidencias en Salud, Vicerrectorado de Investigación, Universidad San Ignacio de Loyola, Lima 14072, Peru
| | - Adrian V. Hernandez
- Unidad de Revisiones Sistemáticas y Meta-análisis, Guías de Práctica Clínica y Evaluaciones de Tecnología Sanitaria, Vicerrectorado de Investigación, Universidad San Ignacio de Loyola, Lima 15012, Peru
- Health Outcomes, Policy, and Evidence Synthesis Group, University of Connecticut School of Pharmacy, Mansfield, CT 06269, USA
| |
Collapse
|
17
|
Liao C, Talluri S, Zhao J, Mu S, Kumar S, Shi J, Buon L, Munshi NC, Shammas MA. RAD51 Is Implicated in DNA Damage, Chemoresistance and Immune Dysregulation in Solid Tumors. Cancers (Basel) 2022; 14:cancers14225697. [PMID: 36428789 PMCID: PMC9688595 DOI: 10.3390/cancers14225697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND In normal cells, homologous recombination (HR) is tightly regulated and plays an important role in the maintenance of genomic integrity and stability through precise repair of DNA damage. RAD51 is a recombinase that mediates homologous base pairing and strand exchange during DNA repair by HR. Our previous data in multiple myeloma and esophageal adenocarcinoma (EAC) show that dysregulated HR mediates genomic instability. Purpose of this study was to investigate role of HR in genomic instability, chemoresistance and immune dysregulation in solid tumors including colon and breast cancers. METHODS The GEO dataset were used to investigate correlation of RAD51 expression with patient survival and expression of various immune markers in EAC, breast and colorectal cancers. RAD51 was inhibited in cancer cell lines using shRNAs and a small molecule inhibitor. HR activity was evaluated using a plasmid-based assay, DNA breaks assessed by evaluating expression of γ-H2AX (a marker of DNA breaks) and p-RPA32 (a marker of DNA end resection) using Western blotting. Genomic instability was monitored by investigating micronuclei (a marker of genomic instability). Impact of RAD51 inhibitor and/or a DNA-damaging agent was assessed on viability and apoptosis in EAC, breast and colon cancer cell lines in vitro and in a subcutaneous tumor model of EAC. Impact of RAD51 inhibitor on expression profile was monitored by RNA sequencing. RESULTS Elevated RAD51 expression correlated with poor survival of EAC, breast and colon cancer patients. RAD51 knockdown in cancer cell lines inhibited DNA end resection and strand exchange activity (key steps in the initiation of HR) as well as spontaneous DNA breaks, whereas its overexpression increased DNA breaks and genomic instability. Treatment of EAC, colon and breast cancer cell lines with a small molecule inhibitor of RAD51 inhibited DNA breaking agent-induced DNA breaks and genomic instability. RAD51 inhibitor potentiated cytotoxicity of DNA breaking agent in all cancer cell types tested in vitro as well as in a subcutaneous model of EAC. Evaluation by RNA sequencing demonstrated that DNA repair and cell cycle related pathways were induced by DNA breaking agent whereas their induction either prevented or reversed by RAD51 inhibitor. In addition, immune-related pathways such as PD-1 and Interferon Signaling were also induced by DNA breaking agent whereas their induction prevented by RAD51 inhibitor. Consistent with these observations, elevated RAD51 expression also correlated with that of genes involved in inflammation and other immune surveillance. CONCLUSIONS Elevated expression of RAD51 and associated HR activity is involved in spontaneous and DNA damaging agent-induced DNA breaks and genomic instability thus contributing to chemoresistance, immune dysregulation and poor prognosis in cancer. Therefore, inhibitors of RAD51 have great potential as therapeutic agents for EAC, colon, breast and probably other solid tumors.
Collapse
Affiliation(s)
- Chengcheng Liao
- Department of Adult Oncology, Harvard (Dana Farber) Cancer Institute, 450 Brookline Ave, Boston, MA 02215, USA
- VA Health Care System, Boston, MA 02215, USA
- Department of Hematology/Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Srikanth Talluri
- Department of Adult Oncology, Harvard (Dana Farber) Cancer Institute, 450 Brookline Ave, Boston, MA 02215, USA
- VA Health Care System, Boston, MA 02215, USA
| | - Jiangning Zhao
- Department of Adult Oncology, Harvard (Dana Farber) Cancer Institute, 450 Brookline Ave, Boston, MA 02215, USA
- VA Health Care System, Boston, MA 02215, USA
| | - Shidai Mu
- Department of Adult Oncology, Harvard (Dana Farber) Cancer Institute, 450 Brookline Ave, Boston, MA 02215, USA
- VA Health Care System, Boston, MA 02215, USA
| | - Subodh Kumar
- Department of Adult Oncology, Harvard (Dana Farber) Cancer Institute, 450 Brookline Ave, Boston, MA 02215, USA
- VA Health Care System, Boston, MA 02215, USA
| | - Jialan Shi
- Department of Adult Oncology, Harvard (Dana Farber) Cancer Institute, 450 Brookline Ave, Boston, MA 02215, USA
- VA Health Care System, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Leutz Buon
- Department of Adult Oncology, Harvard (Dana Farber) Cancer Institute, 450 Brookline Ave, Boston, MA 02215, USA
| | - Nikhil C. Munshi
- Department of Adult Oncology, Harvard (Dana Farber) Cancer Institute, 450 Brookline Ave, Boston, MA 02215, USA
- VA Health Care System, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Masood A. Shammas
- Department of Adult Oncology, Harvard (Dana Farber) Cancer Institute, 450 Brookline Ave, Boston, MA 02215, USA
- VA Health Care System, Boston, MA 02215, USA
- Correspondence:
| |
Collapse
|
18
|
The Molecular and Cellular Strategies of Glioblastoma and Non-Small-Cell Lung Cancer Cells Conferring Radioresistance. Int J Mol Sci 2022; 23:ijms232113577. [PMID: 36362359 PMCID: PMC9656305 DOI: 10.3390/ijms232113577] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Ionizing radiation (IR) has been shown to play a crucial role in the treatment of glioblastoma (GBM; grade IV) and non-small-cell lung cancer (NSCLC). Nevertheless, recent studies have indicated that radiotherapy can offer only palliation owing to the radioresistance of GBM and NSCLC. Therefore, delineating the major radioresistance mechanisms may provide novel therapeutic approaches to sensitize these diseases to IR and improve patient outcomes. This review provides insights into the molecular and cellular mechanisms underlying GBM and NSCLC radioresistance, where it sheds light on the role played by cancer stem cells (CSCs), as well as discusses comprehensively how the cellular dormancy/non-proliferating state and polyploidy impact on their survival and relapse post-IR exposure.
Collapse
|
19
|
Neuroprotective effects of linear ubiquitin E3 ligase against aging-induced DNA damage and amyloid β neurotoxicity in the brain of Drosophila melanogaster. Biochem Biophys Res Commun 2022; 637:196-202. [DOI: 10.1016/j.bbrc.2022.11.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
|
20
|
Schavinski CR, Santos MBD, Londero JEL, Rocha MCD, Amaral AMBD, Ruiz NQ, Leandro GDS, Loro VL, Schuch AP. Effects of isolated and combined exposures of Boana curupi (Anura: Hylidae) tadpoles to environmental doses of trichlorfon and ultraviolet radiation. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2022; 883-884:503549. [PMID: 36462791 DOI: 10.1016/j.mrgentox.2022.503549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 07/22/2022] [Accepted: 09/16/2022] [Indexed: 06/17/2023]
Abstract
The biodiversity collapse strongly affects the amphibian group and many factors have been pointed out as catalytic agents. It is estimated that several events in the amphibian population decline worldwide may have been caused by the interaction of multiple drivers. Thus, this study aimed to evaluate the stressful effects of the exposure to environmental doses of trichlorfon (TCF) pesticide (0.5 μg/L; and an additional 100-fold concentration of 50 µg/L) and ultraviolet radiation (UV) (184.0 kJ/m² of UVA and 3.4 kJ/m² of UVB, which correspond to 5% of the daily dose) in tadpoles of the Boana curupi species (Anura: Hylidae). The isolated and combined exposures to TCF happened within 24 h of acute treatments under laboratory-controlled conditions. In the combined treatments, we adopted three different moments (M) of tadpole irradiation from the beginning of the exposures to TCF (0 h - M1; 12 h - M2; and 24 h - M3). Then, we evaluated tadpole survival, change in morphological characters, induction of apoptotic cells, lipid peroxidation (LPO), protein carbonyl content (PCC), glutathione S-transferase (GST), non-protein thiols (NPSH), and acetylcholinesterase (AChE), as well as the induction of genomic DNA (gDNA) damage. UVB treatment alone resulted in high mortality, along with a high level of apoptosis induction. Both UVA, UVB, and TCF increased LPO, PC, and AChE, while decreased GST activity. Regarding co-exposures, the most striking effect was observed in the interaction between UVB and TCF, which surprisingly decreased UVB-induced tadpole mortality, apoptosis, and gDNA damage. These results reinforce the B. curupi sensitivity to solar UVB radiation and indicate a complex response in face of UVB interaction with TCF, which may be related to activation of DNA repair pathways and/or inhibition of apoptosis, decreasing UVB-induced tadpole mortality.
Collapse
Affiliation(s)
- Cassiano Ricardo Schavinski
- Post-Graduation Program in Biological Sciences: Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Maurício Beux Dos Santos
- Post-Graduation Program in Biological Sciences: Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - James Eduardo Lago Londero
- Post-Graduation Program in Biological Sciences: Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Marcelo Carvalho da Rocha
- Post-Graduation Program in Animal Biodiversity, Department of Ecology and Evolution, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Aline Monique Blank do Amaral
- Post-Graduation Program in Animal Biodiversity, Department of Ecology and Evolution, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Nathalia Quintero Ruiz
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Giovana da Silva Leandro
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Vania Lucia Loro
- Post-Graduation Program in Biological Sciences: Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - André Passaglia Schuch
- Post-Graduation Program in Biological Sciences: Toxicological Biochemistry, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
21
|
Dotto-Vasquez G, Villacorta-Ampuero AK, Ulloque-Badaracco JR, Hernandez-Bustamante EA, Alarcón-Braga EA, Herrera-Añazco P, Benites-Zapata VA, Hernandez AV. Lymphocyte-to-Monocyte Ratio and Clinical Outcomes in Cholangiocarcinoma: A Systematic Review and Meta-Analysis. Diagnostics (Basel) 2022; 12:2655. [PMID: 36359498 PMCID: PMC9689307 DOI: 10.3390/diagnostics12112655] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 11/06/2022] Open
Abstract
Lymphocyte-to-Monocyte ratio (LMR) has shown an association with survival outcomes in several oncological diseases. This study aimed to evaluate the association between LMR and clinical outcomes for cholangiocarcinoma patients. A systematic review and meta-analysis were performed to assess the association between LMR values and overall survival (OS), disease-free survival (DFS), recurrence-free survival (RFS) and time to recurrence (TTR) in cholangiocarcinoma patients. We used Hazard ratio (HR) and their 95% confidence interval (CI) as a measure of effect for the random effect model meta-analysis. The Newcastle-Ottawa Scale was used for quality assessment. The Egger test and funnel plot were developed for approaching publication bias. A total of 19 studies were included in this study (n = 3860). The meta-analysis showed that cholangiocarcinoma patients with low values of LMR were associated with worse OS (HR: 0.82; 95% CI: 0.71-0.96; I2 = 86%) and worse TTR (HR: 0.71; 95% CI: 0.58-0.86; I2 = 0%). DFS and RFS also were evaluated; however, they did not show statistically significant associations. Low LMR values were associated with a worse OS and TTR.
Collapse
Affiliation(s)
| | | | | | - Enrique A. Hernandez-Bustamante
- Sociedad Científica de Estudiantes de Medicina de la Universidad Nacional de Trujillo, Trujillo 13011, Peru
- Grupo Peruano de Investigación Epidemiológica, Unidad para la Generación y Síntesis de Evidencias en Salud, Universidad San Ignacio de Loyola, Lima 15012, Peru
| | - Esteban A. Alarcón-Braga
- Escuela de Medicina, Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
- Sociedad Científica de Estudiantes de Medicina de la Universidad Peruana de Ciencias Aplicadas, Lima 15023, Peru
| | - Percy Herrera-Añazco
- Escuela de Enfermería, Universidad Privada San Juan Bautista, Lima 15067, Peru
- Instituto de Evaluación de Tecnologías en Salud e Investigación—IETSI, EsSalud, Lima 14072, Peru
| | - Vicente A. Benites-Zapata
- Unidad de Investigación para la Generación y Síntesis de Evidencias en Salud, Vicerrectorado de Investigación, Universidad San Ignacio de Loyola, Lima 14072, Peru
| | - Adrian V. Hernandez
- Unidad de Revisiones Sistemáticas y Meta-análisis, Guías de Práctica Clínica y Evaluaciones de Tecnología Sanitaria, Vicerrectorado de Investigación, Universidad San Ignacio de Loyola, Lima 15012, Peru
- Health Outcomes, Policy, and Evidence Synthesis Group, University of Connecticut School of Pharmacy, Mansfield, CT 06269, USA
| |
Collapse
|
22
|
Blankenstein LJ, Cordes N, Kunz-Schughart LA, Vehlow A. Targeting of p21-Activated Kinase 4 Radiosensitizes Glioblastoma Cells via Impaired DNA Repair. Cells 2022; 11:cells11142133. [PMID: 35883575 PMCID: PMC9316146 DOI: 10.3390/cells11142133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 01/27/2023] Open
Abstract
Glioblastoma is a devastating malignant disease with poor patient overall survival. Strong invasiveness and resistance to radiochemotherapy have challenged the identification of molecular targets that can finally improve treatment outcomes. This study evaluates the influence of all six known p21-activated kinase (PAK) protein family members on the invasion capacity and radio-response of glioblastoma cells by employing a siRNA-based screen. In a panel of human glioblastoma cell models, we identified PAK4 as the main PAK isoform regulating invasion and clonogenic survival upon irradiation and demonstrated the radiosensitizing potential of PAK4 inhibition. Mechanistically, we show that PAK4 depletion and pharmacological inhibition enhanced the number of irradiation-induced DNA double-strand breaks and reduced the expression levels of various DNA repair proteins. In conclusion, our data suggest PAK4 as a putative target for radiosensitization and impairing DNA repair in glioblastoma, deserving further scrutiny in extended combinatorial treatment testing.
Collapse
Affiliation(s)
- Leon J. Blankenstein
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, PF 41, 01307 Dresden, Germany; (L.J.B.); (N.C.); (L.A.K.-S.)
- National Center for Tumor Diseases, Partner Site Dresden: German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Nils Cordes
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, PF 41, 01307 Dresden, Germany; (L.J.B.); (N.C.); (L.A.K.-S.)
- National Center for Tumor Diseases, Partner Site Dresden: German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, PF 50, 01307 Dresden, Germany
- Helmholtz-Zentrum Dresden—Rossendorf, Institute of Radiooncology—OncoRay, Bautzner Landstr. 400, 01328 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69192 Heidelberg, Germany
| | - Leoni A. Kunz-Schughart
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, PF 41, 01307 Dresden, Germany; (L.J.B.); (N.C.); (L.A.K.-S.)
- National Center for Tumor Diseases, Partner Site Dresden: German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Anne Vehlow
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, PF 41, 01307 Dresden, Germany; (L.J.B.); (N.C.); (L.A.K.-S.)
- National Center for Tumor Diseases, Partner Site Dresden: German Cancer Research Center, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69192 Heidelberg, Germany
- Correspondence:
| |
Collapse
|
23
|
Budke B, Zhong A, Sullivan K, Park C, Gittin DI, Kountz TS, Connell PP. Noncanonical NF-κB factor p100/p52 regulates homologous recombination and modulates sensitivity to DNA-damaging therapy. Nucleic Acids Res 2022; 50:6251-6263. [PMID: 35689636 PMCID: PMC9226503 DOI: 10.1093/nar/gkac491] [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: 04/13/2020] [Revised: 05/17/2022] [Accepted: 05/25/2022] [Indexed: 11/14/2022] Open
Abstract
Homologous recombination (HR) serves multiple roles in DNA repair that are essential for maintaining genomic stability, including double-strand DNA break (DSB) repair. The central HR protein, RAD51, is frequently overexpressed in human malignancies, thereby elevating HR proficiency and promoting resistance to DNA-damaging therapies. Here, we find that the non-canonical NF-κB factors p100/52, but not RelB, control the expression of RAD51 in various human cancer subtypes. While p100/p52 depletion inhibits HR function in human tumor cells, it does not significantly influence the proficiency of non-homologous end joining, the other key mechanism of DSB repair. Clonogenic survival assays were performed using a pair DLD-1 cell lines that differ only in their expression of the key HR protein BRCA2. Targeted silencing of p100/p52 sensitizes the HR-competent cells to camptothecin, while sensitization is absent in HR-deficient control cells. These results suggest that p100/p52-dependent signaling specifically controls HR activity in cancer cells. Since non-canonical NF-κB signaling is known to be activated after various forms of genomic crisis, compensatory HR upregulation may represent a natural consequence of DNA damage. We propose that p100/p52-dependent signaling represents a promising oncologic target in combination with DNA-damaging treatments.
Collapse
Affiliation(s)
- Brian Budke
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Alison Zhong
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Katherine Sullivan
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Chanyoung Park
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - David I Gittin
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Timothy S Kountz
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Philip P Connell
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
24
|
Gupta P, Majumdar AG, Patro BS. Non-enzymatic function of WRN RECQL helicase regulates removal of topoisomerase-I-DNA covalent complexes and triggers NF-κB signaling in cancer. Aging Cell 2022; 21:e13625. [PMID: 35582959 PMCID: PMC9197415 DOI: 10.1111/acel.13625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/25/2022] [Accepted: 04/20/2022] [Indexed: 11/26/2022] Open
Abstract
Mutation in Werner (WRN) RECQL helicase is associated with premature aging syndrome (Werner syndrome, WS) and predisposition to multiple cancers. In patients with solid cancers, deficiency of the WRN RECQL helicase is paradoxically associated with enhanced overall survival in response to treatment with TOP1 inhibitors, which stabilize pathological TOP1-DNA-covalent-complexes (TOP1cc) on the genome. However, the underlying mechanism of WRN in development of chemoresistance to TOP1 inhibitors is not yet explored. Our whole-genome transcriptomic analysis for ~25,000 genes showed robust activation of NF-κB-dependent prosurvival genes in response to TOP1cc. CRISPR-Cas9 knockout, shRNA silencing, and under-expression of WRN confer high-sensitivity of multiple cancers to TOP1 inhibitor. We demonstrated that WRN orchestrates TOP1cc repair through proteasome-dependent and proteasome-independent process, unleashing robust ssDNA generation. This in turn ensues signal transduction for CHK1 mediated NF-κB-activation through IκBα-degradation and nuclear localization of p65 protein. Intriguingly, our site-directed mutagenesis and rescue experiments revealed that neither RECQL-helicase nor DNA-exonuclease enzyme activity of WRN (WRNE84A , WRNK577M , and WRNE84A-K577M ) were required for TOP1cc removal, ssDNA generation and signaling for NF-κB activation. In correlation with patient data and above results, the TOP1 inhibitor-based targeted therapy showed that WRN-deficient melanoma tumors were highly sensitive to TOP1 inhibition in preclinical in vivo mouse model. Collectively, our findings identify hitherto unknown non-enzymatic role of WRN RECQL helicase in pathological mechanisms underlying TOP1cc processing and subsequent NF-κB-activation, offering a potential targeted therapy for WRN-deficient cancer patients.
Collapse
Affiliation(s)
- Pooja Gupta
- Bio‐Organic DivisionBhabha Atomic Research CentreTrombayMumbaiIndia
- Homi Bhabha National InstituteAnushaktinagarMumbaiIndia
| | - Ananda Guha Majumdar
- Bio‐Organic DivisionBhabha Atomic Research CentreTrombayMumbaiIndia
- Homi Bhabha National InstituteAnushaktinagarMumbaiIndia
| | - Birija Sankar Patro
- Bio‐Organic DivisionBhabha Atomic Research CentreTrombayMumbaiIndia
- Homi Bhabha National InstituteAnushaktinagarMumbaiIndia
| |
Collapse
|
25
|
Moslehi M, Moazamiyanfar R, Dakkali MS, Rezaei S, Rastegar-Pouyani N, Jafarzadeh E, Mouludi K, Khodamoradi E, Taeb S, Najafi M. Modulation of the immune system by melatonin; implications for cancer therapy. Int Immunopharmacol 2022; 108:108890. [PMID: 35623297 DOI: 10.1016/j.intimp.2022.108890] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/07/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022]
Abstract
Immune system interactions within the tumour have a key role in the resistance or sensitization of cancer cells to anti-cancer agents. On the other hand, activation of the immune system in normal tissues following chemotherapy or radiotherapy is associated with acute and late effects such as inflammation and fibrosis. Some immune responses can reduce the efficiency of anti-cancer therapy and also promote normal tissue toxicity. Modulation of immune responses can boost the efficiency of anti-tumour therapy and alleviate normal tissue toxicity. Melatonin is a natural body agent that has shown promising results for modulating tumour response to therapy and also alleviating normal tissue toxicity. This review tries to focus on the immunomodulatory actions of melatonin in both tumour and normal tissues. We will explain how anti-cancer drugs may cause toxicity for normal tissues and how tumours can adapt themselves to ionizing radiation and anti-cancer drugs. Then, cellular and molecular mechanisms of immunoregulatory effects of melatonin alone or combined with other anti-cancer agents will be discussed.
Collapse
Affiliation(s)
- Masoud Moslehi
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Moazamiyanfar
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Sepideh Rezaei
- Department of Chemistry, University of Houston, 3585 Cullen Blvd., Fleming Bldg. Rm 112, Houston, TX 77204-5003, USA
| | - Nima Rastegar-Pouyani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Emad Jafarzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Kave Mouludi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ehsan Khodamoradi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Shahram Taeb
- Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran; Medical Biotechnology Research Center, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran; Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
26
|
Cáceres-Gutiérrez RE, Alfaro-Mora Y, Andonegui MA, Díaz-Chávez J, Herrera LA. The Influence of Oncogenic RAS on Chemotherapy and Radiotherapy Resistance Through DNA Repair Pathways. Front Cell Dev Biol 2022; 10:751367. [PMID: 35359456 PMCID: PMC8962660 DOI: 10.3389/fcell.2022.751367] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 02/15/2022] [Indexed: 11/27/2022] Open
Abstract
RAS oncogenes are chief tumorigenic drivers, and their mutation constitutes a universal predictor of poor outcome and treatment resistance. Despite more than 30 years of intensive research since the identification of the first RAS mutation, most attempts to therapeutically target RAS mutants have failed to reach the clinic. In fact, the first mutant RAS inhibitor, Sotorasib, was only approved by the FDA until 2021. However, since Sotorasib targets the KRAS G12C mutant with high specificity, relatively few patients will benefit from this therapy. On the other hand, indirect approaches to inhibit the RAS pathway have revealed very intricate cascades involving feedback loops impossible to overcome with currently available therapies. Some of these mechanisms play different roles along the multistep carcinogenic process. For instance, although mutant RAS increases replicative, metabolic and oxidative stress, adaptive responses alleviate these conditions to preserve cellular survival and avoid the onset of oncogene-induced senescence during tumorigenesis. The resulting rewiring of cellular mechanisms involves the DNA damage response and pathways associated with oxidative stress, which are co-opted by cancer cells to promote survival, proliferation, and chemo- and radioresistance. Nonetheless, these systems become so crucial to cancer cells that they can be exploited as specific tumor vulnerabilities. Here, we discuss key aspects of RAS biology and detail some of the mechanisms that mediate chemo- and radiotherapy resistance of mutant RAS cancers through the DNA repair pathways. We also discuss recent progress in therapeutic RAS targeting and propose future directions for the field.
Collapse
Affiliation(s)
- Rodrigo E. Cáceres-Gutiérrez
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
| | - Yair Alfaro-Mora
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
- Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Marco A. Andonegui
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
| | - José Díaz-Chávez
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
- *Correspondence: Luis A. Herrera, ; José Díaz-Chávez,
| | - Luis A. Herrera
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
- Instituto Nacional de Medicina Genómica, Mexico City, Mexico
- *Correspondence: Luis A. Herrera, ; José Díaz-Chávez,
| |
Collapse
|
27
|
Matsuya Y, Hamada N, Yachi Y, Satou Y, Ishikawa M, Date H, Sato T. Inflammatory Signaling and DNA Damage Responses after Local Exposure to an Insoluble Radioactive Microparticle. Cancers (Basel) 2022; 14:cancers14041045. [PMID: 35205797 PMCID: PMC8869995 DOI: 10.3390/cancers14041045] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/09/2022] [Accepted: 02/15/2022] [Indexed: 12/30/2022] Open
Abstract
Simple Summary A cesium-bearing microparticle (Cs-BMP) is an insoluble radioactive microparticle possessing high specific radioactivity, which was discovered after the incident at the Fukushima nuclear power plant. Due to their insoluble nature, such Cs-BMPs are assumed to adhere in the long term to normal tissue, leading to chronic local exposure. However, radiation risk due to the intake of internal exposure to radioactive cesium is conventionally estimated from the organ dose given by uniform exposure to soluble cesium. As such, it is critical to clarify the normal tissue effects posed by heterogeneous exposure to Cs-BMPs. This in vitro study reports on the relationship between the inflammatory responses and DNA damage induction during local exposure to a Cs-BMP. Abstract Cesium-bearing microparticles (Cs-BMPs) can reach the human respiratory system after inhalation, resulting in chronic local internal exposure. We previously investigated the spatial distribution of DNA damage induced in areas around a Cs-BMP; however, the biological impacts have not been fully clarified due to the limited amount of data. Here, we investigated the inflammatory signaling and DNA damage responses after local exposure to a Cs-BMP in vitro. We used two normal human lung cell lines, i.e., lung fibroblast cells (WI-38) and bronchial epithelial cells (HBEC3-KT). After 24 h exposure to a Cs-BMP, inflammation was evaluated by immunofluorescent staining for nuclear factor κB (NF-κB) p65 and cyclooxygenase 2 (COX-2). The number of DNA double-strand breaks (DSBs) was also detected by means of phospholylated histone H2AX (γ-H2AX) focus formation assay. Cs-BMP exposure significantly increased NF-κB p65 and COX-2 expressions, which were related to the number of γ-H2AX foci in the cell nuclei. Compared to the uniform (external) exposure to 137Cs γ-rays, NF-κB tended to be more activated in the cells proximal to the Cs-BMP, while both NF-κB p65 and COX-2 were significantly activated in the distal cells. Experiments with chemical inhibitors for NF-κB p65 and COX-2 suggested the involvement of such inflammatory responses both in the reduced radiosensitivity of the cells proximal to Cs-BMP and the enhanced radiosensitivity of the cells distal from Cs-BMP. The data show that local exposure to Cs-BMP leads to biological effects modified by the NF-κB pathway, suggesting that the radiation risk for Cs-BMP exposure can differ from that estimated based on conventional uniform exposure to normal tissues.
Collapse
Affiliation(s)
- Yusuke Matsuya
- Nuclear Science and Engineering Center, Japan Atomic Energy Agency (JAEA), 2-4 Shirakata, Tokai 319-1195, Ibaraki, Japan;
- Correspondence:
| | - Nobuyuki Hamada
- Radiation Safety Unit, Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), 2-11-1 Iwado-kita, Komae 201-8511, Tokyo, Japan;
| | - Yoshie Yachi
- Graduate School of Health Sciences, Hokkaido University, Kita-12 Nishi-8, Kita-ku, Sapporo 060-0812, Hokkaido, Japan;
| | - Yukihiko Satou
- Collaborative Laboratories for Advanced Decommissioning Science (CLADS), Japan Atomic Energy Agency (JAEA), 790-1 Otsuka, Motooka Tomioka, Futaba 979-1151, Fukushima, Japan;
| | - Masayori Ishikawa
- Faculty of Health Sciences, Hokkaido University, Kita-12 Nishi-8, Kita-ku, Sapporo 060-0812, Hokkaido, Japan; (M.I.); (H.D.)
| | - Hiroyuki Date
- Faculty of Health Sciences, Hokkaido University, Kita-12 Nishi-8, Kita-ku, Sapporo 060-0812, Hokkaido, Japan; (M.I.); (H.D.)
| | - Tatsuhiko Sato
- Nuclear Science and Engineering Center, Japan Atomic Energy Agency (JAEA), 2-4 Shirakata, Tokai 319-1195, Ibaraki, Japan;
| |
Collapse
|
28
|
Nouri-Vaskeh M, Mirza-Aghazadeh-Attari M, Pashazadeh F, Azami-Aghdash S, Alizadeh H, Pouya P, Halimi M, Jadideslam G, Zarei M. Prognostic Impact of Monocyte to Lymphocyte Ratio in Clinical Outcome of Patients with Hepatocellular Carcinoma: A Systematic Review and Meta-analysis. Galen Med J 2021; 9:e1948. [PMID: 34466618 PMCID: PMC8344106 DOI: 10.31661/gmj.v9i0.1948] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/23/2020] [Accepted: 06/30/2020] [Indexed: 01/14/2023] Open
Abstract
Background: Lymphocyte to monocyte ratio (LMR) is a surrogate marker of systemic inflammation which is shown to be related to the patient’s survival in multiple malignancies. An important implication of this marker potentially is neoplasms in which there is no correlation between prognosis and histopathological staging and also has no reliable chemical markers associated with prognosis. Herein, this meta-analysis aimed to investigate the prognostic role of LMR in patients with hepatocellular carcinoma (HCC). Materials and Methods: In the current systemic review and meta-analysis, we conducted a systemic search of databases and indexing sources, including PubMed, EMBASE, Cochrane, Scopus, and ProQuest up to May 2019 toinclude studies on the prognostic significance of LMR on patients with HCC. Overall survival (OS), disease-free survival (DFS) and recurrence-free survival (RFS) values were extracted from the studies and analyzed. The pooled hazard ratio with a 95% confidence interval was explored to identify the prognostic value of the LMR in the survival of the patients with HCC. Results: A total of 12 studies with a total sample size of 3750 cases were included. There was significant heterogeneity among the studies; therefore, subgroup analysis was also performed. Overall analysis regarding OS showed an insignificant relationship between LMR and patient’s prognosis, dividing to subgroups based on LMR cut-offs did not yield any significant result, subgroup analysis for RFS founded statistically significant results and LMR was significantly related to DFS. Conclusion: High LMR was associated with increased DFS and RFS, in return this association was not observed for OS.
Collapse
Affiliation(s)
- Masoud Nouri-Vaskeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity, Universal Scientific Education and Research Network, Tehran, Iran
| | | | - Fariba Pashazadeh
- Iranian EBM Centre: A Joanna Briggs Institute Affiliated Group
- Research Center for Evidence-Based Medicine, Health Management and Safety Promotion Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saber Azami-Aghdash
- Tabriz Health Services Management Research Center, Health Management and Safety Promotion Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Alizadeh
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parnia Pouya
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Monireh Halimi
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Correspondence to: Monireh Halimi, Associate Professor of Pathology, Department of Pathology, Tabriz University of Medical Sciences, Tabriz, Iran Telephone Number: +984133376923 Email Address:
| | - Golamreza Jadideslam
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz university of Medical Sciences, Tabriz, Iran
| | - Mohammad Zarei
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Mitochondrial and Epigenomic Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
29
|
Transcription/Replication Conflicts in Tumorigenesis and Their Potential Role as Novel Therapeutic Targets in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13153755. [PMID: 34359660 PMCID: PMC8345052 DOI: 10.3390/cancers13153755] [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: 05/12/2021] [Revised: 07/13/2021] [Accepted: 07/22/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Multiple myeloma is a hematologic cancer characterized by the accumulation of malignant plasma cells in the bone marrow. It remains a mostly incurable disease due to the inability to overcome refractory disease and drug-resistant relapse. Oncogenic transformation of PC in multiple myeloma is thought to occur within the secondary lymphoid organs. However, the precise molecular events leading to myelomagenesis remain obscure. Here, we identified genes involved in the prevention and the resolution of conflicts between the replication and transcription significantly overexpressed during the plasma cell differentiation process and in multiple myeloma cells. We discussed the potential role of these factors in myelomagenesis and myeloma biology. The specific targeting of these factors might constitute a new therapeutic strategy in multiple myeloma. Abstract Plasma cells (PCs) have an essential role in humoral immune response by secretion of antibodies, and represent the final stage of B lymphocytes differentiation. During this differentiation, the pre-plasmablastic stage is characterized by highly proliferative cells that start to secrete immunoglobulins (Igs). Thus, replication and transcription must be tightly regulated in these cells to avoid transcription/replication conflicts (TRCs), which could increase replication stress and lead to genomic instability. In this review, we analyzed expression of genes involved in TRCs resolution during B to PC differentiation and identified 41 genes significantly overexpressed in the pre-plasmablastic stage. This illustrates the importance of mechanisms required for adequate processing of TRCs during PCs differentiation. Furthermore, we identified that several of these factors were also found overexpressed in purified PCs from patients with multiple myeloma (MM) compared to normal PCs. Malignant PCs produce high levels of Igs concomitantly with cell cycle deregulation. Therefore, increasing the TRCs occurring in MM cells could represent a potent therapeutic strategy for MM patients. Here, we describe the potential roles of TRCs resolution factors in myelomagenesis and discuss the therapeutic interest of targeting the TRCs resolution machinery in MM.
Collapse
|
30
|
Abadi B, Shahsavani Y, Faramarzpour M, Rezaei N, Rahimi HR. Antidepressants with anti-tumor potential in treating glioblastoma: A narrative review. Fundam Clin Pharmacol 2021; 36:35-48. [PMID: 34212424 DOI: 10.1111/fcp.12712] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 06/13/2021] [Accepted: 06/25/2021] [Indexed: 11/30/2022]
Abstract
Glioblastoma multiforme (GBM) is known as the deadliest form of brain tumor. In addition, its high treatment resistance, heterogeneity, and invasiveness make it one of the most challenging tumors. Depression is a common psychological disorder among patients with cancer, especially GBM. Due to the high occurrence rates of depression in GBM patients and the overlap of molecular and cellular mechanisms involved in the pathogenesis of these diseases, finding antidepressants with antitumor effects could be considered as an affordable strategy for the treatment of GBM. Antidepressants exert their antitumor properties through different mechanisms. According to available evidence in this regard, some of them can eliminate the adverse effects resulting from chemo-radiotherapy in several cancers along with their synergistic effects caused by chemotherapy. Therefore, providing comprehensive insight into this issue would guide scientists and physicians in developing further preclinical studies and clinical trials, in order to evaluate antidepressants' antitumor potential. Considering that no narrative review has been recently published on this issue, specifically on these classes of drugs, we present this article with the purpose of describing the antitumor cellular mechanisms of three classes of antidepressants as follows: tricyclic antidepressants (TCAs), selective serotonin reuptake inhibitors (SSRIs), and monoamine oxidase inhibitors (MAOIs) in GBM.
Collapse
Affiliation(s)
- Banafshe Abadi
- Brain Cancer Research Core (BCRC), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Yasamin Shahsavani
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran.,Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahsa Faramarzpour
- Brain Cancer Research Core (BCRC), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman University of Medical Sciences, Kerman, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hamid-Reza Rahimi
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
31
|
Eberle J, Wiehe RS, Gole B, Mattis LJ, Palmer A, Ständker L, Forssmann WG, Münch J, Gebhardt JCM, Wiesmüller L. A Fibrinogen Alpha Fragment Mitigates Chemotherapy-Induced MLL Rearrangements. Front Oncol 2021; 11:689063. [PMID: 34222016 PMCID: PMC8249925 DOI: 10.3389/fonc.2021.689063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/03/2021] [Indexed: 11/25/2022] Open
Abstract
Rearrangements in the Mixed Lineage Leukemia breakpoint cluster region (MLLbcr) are frequently involved in therapy-induced leukemia, a severe side effect of anti-cancer therapies. Previous work unraveled Endonuclease G as the critical nuclease causing initial breakage in the MLLbcr in response to different types of chemotherapeutic treatment. To identify peptides protecting against therapy-induced leukemia, we screened a hemofiltrate-derived peptide library by use of an enhanced green fluorescent protein (EGFP)-based chromosomal reporter of MLLbcr rearrangements. Chromatographic purification of one active fraction and subsequent mass spectrometry allowed to isolate a C-terminal 27-mer of fibrinogen α encompassing amino acids 603 to 629. The chemically synthesized peptide, termed Fα27, inhibited MLLbcr rearrangements in immortalized hematopoietic cells following treatment with the cytostatics etoposide or doxorubicin. We also provide evidence for protection of primary human hematopoietic stem and progenitor cells from therapy-induced MLLbcr breakage. Of note, fibrinogen has been described to activate toll-like receptor 4 (TLR4). Dissecting the Fα27 mode-of action revealed association of the peptide with TLR4 in an antagonistic fashion affecting downstream NFκB signaling and pro-inflammatory cytokine production. In conclusion, we identified a hemofiltrate-derived peptide inhibitor of the genome destabilizing events causing secondary leukemia in patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Julia Eberle
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
| | | | - Boris Gole
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
| | - Liska Jule Mattis
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
| | - Anja Palmer
- Department of Physics, Institute of Biophysics, Ulm University, Ulm, Germany
| | - Ludger Ständker
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, Germany
| | - Wolf-Georg Forssmann
- Pharis Biotec GmbH and Peptide Research Group, Institute of Immunology and Rheumatology, Hannover Medical School, Hannover, Germany
| | - Jan Münch
- Core Facility Functional Peptidomics, Ulm University Medical Center, Ulm, Germany
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | | | - Lisa Wiesmüller
- Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
| |
Collapse
|
32
|
Altered replication stress response due to CARD14 mutations promotes recombination-induced revertant mosaicism. Am J Hum Genet 2021; 108:1026-1039. [PMID: 34004138 DOI: 10.1016/j.ajhg.2021.04.021] [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] [Received: 10/14/2020] [Accepted: 04/22/2021] [Indexed: 01/07/2023] Open
Abstract
Revertant mosaicism, or "natural gene therapy," refers to the spontaneous in vivo reversion of an inherited mutation in a somatic cell. Only approximately 50 human genetic disorders exhibit revertant mosaicism, implicating a distinctive role played by mutant proteins in somatic correction of a pathogenic germline mutation. However, the process by which mutant proteins induce somatic genetic reversion in these diseases remains unknown. Here we show that heterozygous pathogenic CARD14 mutations causing autoinflammatory skin diseases, including psoriasis and pityriasis rubra pilaris, are repaired mainly via homologous recombination. Rather than altering the DNA damage response to exogenous stimuli, such as X-irradiation or etoposide treatment, mutant CARD14 increased DNA double-strand breaks under conditions of replication stress. Furthermore, mutant CARD14 suppressed new origin firings without promoting crossover events in the replication stress state. Together, these results suggest that mutant CARD14 alters the replication stress response and preferentially drives break-induced replication (BIR), which is generally suppressed in eukaryotes. Our results highlight the involvement of BIR in reversion events, thus revealing a previously undescribed role of BIR that could potentially be exploited to develop therapeutics for currently intractable genetic diseases.
Collapse
|
33
|
Age-related activity of Poly (ADP-Ribose) Polymerase (PARP) in men with localized prostate cancer. Mech Ageing Dev 2021; 196:111494. [PMID: 33887280 DOI: 10.1016/j.mad.2021.111494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/01/2021] [Accepted: 04/15/2021] [Indexed: 12/11/2022]
Abstract
Mutations in DNA repair genes have been connected with familial prostate cancer and sensitivity to targeted drugs like PARP-inhibitors. Clinical use of this information is limited by the small fraction of prostate cancer risk gene carriers, variants of unknown pathogenicity and the focus on monogenic disease mechanisms. Functional assays capturing mono- and polygenic defects were shown to detect breast and ovarian cancer risk in blood-derived cells. Here, we comparatively analyzed lymphocytes from prostate cancer patients and controls applying a sensitive DNA double-strand break (DSB) repair assay and a flow cytometrybased assay measuring the activity of Poly(ADP-Ribose)-Polymerase, a target in treatment of metastatic prostate cancer. Contrary to breast and ovarian cancer patients, error-prone DNA double-strand break repair was not activated in prostate cancer patients. Yet, the activity of PARP discriminated between prostate cancer cases and controls. PARylation also correlated with the age of male probands, suggesting male-specific links between mutation-based and aging-associated DNA damage accumulation and PARP. Our work identifies prostate cancer-specific DNA repair phenotypes characterized by increased PARP activities and carboplatin-sensitivities, detected by functional testing of lymphocytes. This provides new insights for further investigation of PARP and carboplatin sensitivity as biomarkers in peripheral cells of men and prostate cancer patients.
Collapse
|
34
|
Iqbal S, Shah MA, Rasul A, Saadullah M, Tabassum S, Ali S, Zafar M, Muhammad H, Uddin MS, Batiha GES, Vargas-De-La-Cruz C. Radioprotective Potential of Nutraceuticals and their Underlying Mechanism of Action. Anticancer Agents Med Chem 2021; 22:40-52. [PMID: 33622231 DOI: 10.2174/1871520621666210223101246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 11/22/2022]
Abstract
Radiations are an efficient treatment modality in cancer therapy. Besides the treatment effects of radiations, the ionizing radiations interact with biological systems and generate reactive oxygen species that interfere with the normal cellular process. Previous investigations of synthetic radioprotectors have shown less effectiveness, mainly owing to some limiting effects. The nutraceuticals act as efficient radioprotectors to protect the tissues from the deleterious effects of radiation. The main radioprotection mechanism of nutraceuticals is the scavenging of free radicals while other strategies are involved modulation of signaling transduction of pathways like MAPK (JNK, ERK1/2, ERK5, and P38), NF-kB, cytokines, and their protein regulatory genes expression. The current review is focused on the radioprotective effects of nutraceuticals including vitamin E, -C, organosulphur compounds, phenylpropanoids, and polysaccharides. These natural entities protect against radiation-induced DNA damage. The review mainly entails the antioxidant perspective and mechanism of action of their radioprotective activities on a molecular level, DNA repair pathway, anti-inflammation, immunomodulatory effects, the effect on cellular signaling pathways, and regeneration of hematopoietic cells.
Collapse
Affiliation(s)
- Shabnoor Iqbal
- Department of Zoology, Faculty of Life Sciences, Government College University Faisalabad. Pakistan
| | - Muhammad A Shah
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad. Pakistan
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University Faisalabad. Pakistan
| | - Malik Saadullah
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad. Pakistan
| | - Sobia Tabassum
- Department of Biological Sciences, International Islamic University, Islamabad. Pakistan
| | - Shujat Ali
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013. China
| | - Muhammad Zafar
- Department of Plant Sciences, Quaid-i-Azam University, Islamabad. Pakistan
| | - Haji Muhammad
- Department of Chemistry, Federal Urdu University of Arts, Science & Technology, Karachi. Pakistan
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka. Pakistan
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira. Egypt
| | - Celia Vargas-De-La-Cruz
- Faculty of Pharmacy and Biochemistry, Academic Department of Pharmacology, Bromatology and Toxicology, Centro Latinoamericano de Enseñanza e Investigación en Bacteriología Alimentaria (CLEIBA), Universidad Nacional Mayor de San Marcos, Lima15001. Peru
| |
Collapse
|
35
|
Chi RPA, van der Watt P, Wei W, Birrer MJ, Leaner VD. Inhibition of Kpnβ1 mediated nuclear import enhances cisplatin chemosensitivity in cervical cancer. BMC Cancer 2021; 21:106. [PMID: 33530952 PMCID: PMC7852134 DOI: 10.1186/s12885-021-07819-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
Background Inhibition of nuclear import via Karyopherin beta 1 (Kpnβ1) shows potential as an anti-cancer approach. This study investigated the use of nuclear import inhibitor, INI-43, in combination with cisplatin. Methods Cervical cancer cells were pre-treated with INI-43 before treatment with cisplatin, and MTT cell viability and apoptosis assays performed. Activity and localisation of p53 and NFκB was determined after co-treatment of cells. Results Pre-treatment of cervical cancer cells with INI-43 at sublethal concentrations enhanced cisplatin sensitivity, evident through decreased cell viability and enhanced apoptosis. Kpnβ1 knock-down cells similarly displayed increased sensitivity to cisplatin. Combination index determination using the Chou-Talalay method revealed that INI-43 and cisplatin engaged in synergistic interactions. p53 was found to be involved in the cell death response to combination treatment as its inhibition abolished the enhanced cell death observed. INI-43 pre-treatment resulted in moderately stabilized p53 and induced p53 reporter activity, which translated to increased p21 and decreased Mcl-1 upon cisplatin combination treatment. Furthermore, cisplatin treatment led to nuclear import of NFκB, which was diminished upon pre-treatment with INI-43. NFκB reporter activity and expression of NFκB transcriptional targets, cyclin D1, c-Myc and XIAP, showed decreased levels after combination treatment compared to single cisplatin treatment and this associated with enhanced DNA damage. Conclusions Taken together, this study shows that INI-43 pre-treatment significantly enhances cisplatin sensitivity in cervical cancer cells, mediated through stabilization of p53 and decreased nuclear import of NFκB. Hence this study suggests the possible synergistic use of nuclear import inhibition and cisplatin to treat cervical cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-07819-3.
Collapse
Affiliation(s)
- Ru-Pin Alicia Chi
- Division of Medical Biochemistry & Structural Biology, Department of Integrative Biomedical Sciences, SAMRC/UCT Gynaecological Cancer Research Centre, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Cape Town, 7925, South Africa
| | - Pauline van der Watt
- Division of Medical Biochemistry & Structural Biology, Department of Integrative Biomedical Sciences, SAMRC/UCT Gynaecological Cancer Research Centre, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Cape Town, 7925, South Africa
| | - Wei Wei
- Pfizer, Andover, MA, 01810, USA
| | - Michael J Birrer
- University of Arkansas Medical Sciences, D Winthrop P. Rockefeller Cancer Institute, Little Rock, AR, USA
| | - Virna D Leaner
- Division of Medical Biochemistry & Structural Biology, Department of Integrative Biomedical Sciences, SAMRC/UCT Gynaecological Cancer Research Centre, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Cape Town, 7925, South Africa.
| |
Collapse
|
36
|
Statello L, Ali MM, Reischl S, Mahale S, Kosalai ST, Huarte M, Kanduri C. The DNA damage inducible lncRNA SCAT7 regulates genomic integrity and topoisomerase 1 turnover in lung adenocarcinoma. NAR Cancer 2021; 3:zcab002. [PMID: 34316698 PMCID: PMC8209975 DOI: 10.1093/narcan/zcab002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/26/2020] [Accepted: 01/06/2021] [Indexed: 12/15/2022] Open
Abstract
Despite the rapid improvements in unveiling the importance of lncRNAs in all aspects of cancer biology, there is still a void in mechanistic understanding of their role in the DNA damage response. Here we explored the potential role of the oncogenic lncRNA SCAT7 (ELF3-AS1) in the maintenance of genome integrity. We show that SCAT7 is upregulated in response to DNA-damaging drugs like cisplatin and camptothecin, where SCAT7 expression is required to promote cell survival. SCAT7 silencing leads to decreased proliferation of cisplatin-resistant cells in vitro and in vivo through interfering with cell cycle checkpoints and DNA repair molecular pathways. SCAT7 regulates ATR signaling, promoting homologous recombination. Importantly, SCAT7 also takes part in proteasome-mediated topoisomerase I (TOP1) degradation, and its depletion causes an accumulation of TOP1–cc structures responsible for the high levels of intrinsic DNA damage. Thus, our data demonstrate that SCAT7 is an important constituent of the DNA damage response pathway and serves as a potential therapeutic target for hard-to-treat drug resistant cancers.
Collapse
Affiliation(s)
- Luisa Statello
- Department of Medical Biochemistry and Cell Biology, The Sahlgrenska Academy, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Mohamad M Ali
- Department of Medical Biochemistry and Cell Biology, The Sahlgrenska Academy, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Silke Reischl
- Department of Medical Biochemistry and Cell Biology, The Sahlgrenska Academy, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Sagar Mahale
- Department of Medical Biochemistry and Cell Biology, The Sahlgrenska Academy, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Subazini Thankaswamy Kosalai
- Department of Medical Biochemistry and Cell Biology, The Sahlgrenska Academy, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| | - Maite Huarte
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical Research, University of Navarra, Pamplona 31008, Spain
| | - Chandrasekhar Kanduri
- Department of Medical Biochemistry and Cell Biology, The Sahlgrenska Academy, Institute of Biomedicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
| |
Collapse
|
37
|
Volcic M, Sparrer KMJ, Koepke L, Hotter D, Sauter D, Stürzel CM, Scherer M, Stamminger T, Hofmann TG, Arhel NJ, Wiesmüller L, Kirchhoff F. Vpu modulates DNA repair to suppress innate sensing and hyper-integration of HIV-1. Nat Microbiol 2020; 5:1247-1261. [PMID: 32690953 PMCID: PMC7616938 DOI: 10.1038/s41564-020-0753-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 06/11/2020] [Indexed: 12/19/2022]
Abstract
To avoid innate sensing and immune control, human immunodeficiency virus type 1 (HIV-1) has to prevent the accumulation of viral complementary DNA species. Here, we show that the late HIV-1 accessory protein Vpu hijacks DNA repair mechanisms to promote degradation of nuclear viral cDNA in cells that are already productively infected. Vpu achieves this by interacting with RanBP2-RanGAP1*SUMO1-Ubc9 SUMO E3-ligase complexes at the nuclear pore to reprogramme promyelocytic leukaemia protein nuclear bodies and reduce SUMOylation of Bloom syndrome protein, unleashing end degradation of viral cDNA. Concomitantly, Vpu inhibits RAD52-mediated homologous repair of viral cDNA, preventing the generation of dead-end circular forms of single copies of the long terminal repeat and permitting sustained nucleolytic attack. Our results identify Vpu as a key modulator of the DNA repair machinery. We show that Bloom syndrome protein eliminates nuclear HIV-1 cDNA and thereby suppresses immune sensing and proviral hyper-integration. Therapeutic targeting of DNA repair may facilitate the induction of antiviral immunity and suppress proviral integration replenishing latent HIV reservoirs.
Collapse
Affiliation(s)
- Meta Volcic
- Division of Gynecological Oncology, Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | | | - Lennart Koepke
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Dominik Hotter
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Daniel Sauter
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Christina M Stürzel
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | | | | | - Thomas G Hofmann
- Department of Epigenetics, Deutsches Krebsforschungszentrum, Heidelberg, Germany
- Institute of Toxicology, University of Mainz, Mainz, Germany
| | - Nathalie J Arhel
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
- Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé (CPBS) CNRS FRE3689, Montpellier University, Montpellier, France
| | - Lisa Wiesmüller
- Division of Gynecological Oncology, Department of Obstetrics and Gynecology, Ulm University, Ulm, Germany.
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
| |
Collapse
|
38
|
Alonso-de Vega I, Paz-Cabrera MC, Rother MB, Wiegant WW, Checa-Rodríguez C, Hernández-Fernaud JR, Huertas P, Freire R, van Attikum H, Smits VAJ. PHF2 regulates homology-directed DNA repair by controlling the resection of DNA double strand breaks. Nucleic Acids Res 2020; 48:4915-4927. [PMID: 32232336 PMCID: PMC7229830 DOI: 10.1093/nar/gkaa196] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 03/12/2020] [Accepted: 03/27/2020] [Indexed: 12/17/2022] Open
Abstract
Post-translational histone modifications and chromatin remodelling play a critical role controlling the integrity of the genome. Here, we identify histone lysine demethylase PHF2 as a novel regulator of the DNA damage response by regulating DNA damage-induced focus formation of 53BP1 and BRCA1, critical factors in the pathway choice for DNA double strand break repair. PHF2 knockdown leads to impaired BRCA1 focus formation and delays the resolution of 53BP1 foci. Moreover, irradiation-induced RPA phosphorylation and focus formation, as well as localization of CtIP, required for DNA end resection, to sites of DNA lesions are affected by depletion of PHF2. These results are indicative of a defective resection of double strand breaks and thereby an impaired homologous recombination upon PHF2 depletion. In accordance with these data, Rad51 focus formation and homology-directed double strand break repair is inhibited in cells depleted for PHF2. Importantly, we demonstrate that PHF2 knockdown decreases CtIP and BRCA1 protein and mRNA levels, an effect that is dependent on the demethylase activity of PHF2. Furthermore, PHF2-depleted cells display genome instability and are mildly sensitive to the inhibition of PARP. Together these results demonstrate that PHF2 promotes DNA repair by homologous recombination by controlling CtIP-dependent resection of double strand breaks.
Collapse
Affiliation(s)
| | | | - Magdalena B Rother
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Wouter W Wiegant
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Pablo Huertas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Sevilla, Spain
| | - Raimundo Freire
- Unidad de Investigación, Hospital Universitario de Canarias, Tenerife, Spain.,Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Tenerife, Spain.,Universidad Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - Haico van Attikum
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Veronique A J Smits
- Unidad de Investigación, Hospital Universitario de Canarias, Tenerife, Spain.,Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Tenerife, Spain.,Universidad Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| |
Collapse
|
39
|
Hsu FT, Chiang I, Wang W. Induction of apoptosis through extrinsic/intrinsic pathways and suppression of ERK/NF-κB signalling participate in anti-glioblastoma of imipramine. J Cell Mol Med 2020; 24:3982-4000. [PMID: 32149465 PMCID: PMC7171418 DOI: 10.1111/jcmm.15022] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 11/27/2019] [Accepted: 01/06/2020] [Indexed: 12/11/2022] Open
Abstract
Glioblastomas are the most aggressive type of brain tumour, with poor prognosis even after standard treatment such as surgical resection, temozolomide and radiation therapy. The overexpression of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in glioblastomas is recognized as an important treatment target. Thus, an urgent need regarding glioblastomas is the development of a new, suitable agent that may show potential for the inhibition of extracellular signal-regulated kinase (ERK)/NF-κB-mediated glioblastoma progression. Imipramine, a tricyclic antidepressant, has anti-inflammatory actions against inflamed glial cells; additionally, imipramine can induce glioblastoma toxicity via the activation of autophagy. However, whether imipramine can suppress glioblastoma progression via the induction of apoptosis and blockage of ERK/NF-κB signalling remains unclear. The main purpose of this study was to investigate the effects of imipramine on apoptotic signalling and ERK/NF-κB-mediated glioblastoma progression by using cell proliferation (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide [MTT] assay), flow cytometry, Western blotting, and cell invasion/migration assay analysis in vitro. The ERK and NF-κB inhibitory capacity of imipramine is detected by NF-κB reporter gene assay and Western blotting. Additionally, a glioblastoma-bearing animal model was used to validate the therapeutic efficacy and general toxicity of imipramine. Our results demonstrated that imipramine successfully triggered apoptosis through extrinsic/intrinsic pathways and suppressed the invasion/migration ability of glioblastoma cells. Furthermore, imipramine effectively suppressed glioblastoma progression in vivo via the inhibition of the ERK/NF-κB pathway. In summary, imipramine is a potential anti-glioblastoma drug which induces apoptosis and has the capacity to inhibit ERK/NF-κB signalling.
Collapse
Affiliation(s)
- Fei-Ting Hsu
- Department of Biological Science and TechnologyChina Medical UniversityTaichungTaiwan
| | - I‐Tsang Chiang
- Department of Radiation OncologyShow Chwan Memorial HospitalChanghuaTaiwan
- Department of Radiation OncologyChang Bing Show Chwan Memorial HospitalLukangTaiwan
- Department of Medical Imaging and Radiological SciencesCentral Taiwan University of Science and TechnologyTaichungTaiwan
| | - Wei‐Shu Wang
- Department of MedicineNational Yang‐Ming University HospitalYilanTaiwan
- School of MedicineNational Yang‐Ming UniversityTaipeiTaiwan
| |
Collapse
|
40
|
Lama-Sherpa TD, Shevde LA. An Emerging Regulatory Role for the Tumor Microenvironment in the DNA Damage Response to Double-Strand Breaks. Mol Cancer Res 2019; 18:185-193. [PMID: 31676722 DOI: 10.1158/1541-7786.mcr-19-0665] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/23/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022]
Abstract
Radiation, alkylating agents, and platinum-based chemotherapy treatments eliminate cancer cells through the induction of excessive DNA damage. The resultant DNA damage challenges the cancer cell's DNA repair capacity. Among the different types of DNA damage induced in cells, double-strand breaks (DSB) are the most lethal if left unrepaired. Unrepaired DSBs in tumor cells exacerbate existing gene deletions, chromosome losses and rearrangements, and aberrant features that characteristically enable tumor progression, metastasis, and drug resistance. Tumor microenvironmental factors like hypoxia, inflammation, cellular metabolism, and the immune system profoundly influence DSB repair mechanisms. Here, we put into context the role of the microenvironment in governing DSB repair mechanisms.
Collapse
Affiliation(s)
| | - Lalita A Shevde
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, Alabama. .,O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
41
|
Mortezaee K, Potes Y, Mirtavoos-Mahyari H, Motevaseli E, Shabeeb D, Musa AE, Najafi M, Farhood B. Boosting immune system against cancer by melatonin: A mechanistic viewpoint. Life Sci 2019; 238:116960. [PMID: 31629760 DOI: 10.1016/j.lfs.2019.116960] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/05/2019] [Accepted: 10/11/2019] [Indexed: 12/15/2022]
Abstract
Cancer is a disease of high complexity. Resistance to therapy is a major challenge in cancer targeted therapies. Overcoming this resistance requires a deep knowledge of the cellular interactions within tumor. Natural killer (NK) cells and cytotoxic T lymphocytes (CTLs) are the main anti-cancer immune cells, while T regulatory cells (Tregs) and cancer associated fibroblasts (CAFs) facilitate immune escape of cancer cells. Melatonin is a natural agent with anti-cancer functions that has also been suggested as an adjuvant in combination with cancer therapy modalities such as chemotherapy, radiotherapy, immunotherapy and tumor vaccination. One of the main effects of melatonin is regulation of immune responses against cancer cells. Melatonin has been shown to potentiate the activities of anti-cancer immune cells, as well as attenuating the activities of Tregs and CAFs. It also has a potent effect on the mitochondria, which may change immune responses against cancer. In this review, we explain the mechanisms of immune regulation by melatonin involved in its anti-cancer effects.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Yaiza Potes
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, C/ Julián Clavería 6, 33006, Oviedo, Spain
| | - Hanifeh Mirtavoos-Mahyari
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology, College of Medicine, University of Misan, Misan, Iraq
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences (International Campus), Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
42
|
BRCA-1 depletion impairs pro-inflammatory polarization and activation of RAW 264.7 macrophages in a NF-κB-dependent mechanism. Mol Cell Biochem 2019; 462:11-23. [DOI: 10.1007/s11010-019-03605-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/08/2019] [Indexed: 12/15/2022]
|
43
|
Kay J, Thadhani E, Samson L, Engelward B. Inflammation-induced DNA damage, mutations and cancer. DNA Repair (Amst) 2019; 83:102673. [PMID: 31387777 DOI: 10.1016/j.dnarep.2019.102673] [Citation(s) in RCA: 255] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 06/15/2019] [Accepted: 07/18/2019] [Indexed: 12/22/2022]
Abstract
The relationships between inflammation and cancer are varied and complex. An important connection linking inflammation to cancer development is DNA damage. During inflammation reactive oxygen and nitrogen species (RONS) are created to combat pathogens and to stimulate tissue repair and regeneration, but these chemicals can also damage DNA, which in turn can promote mutations that initiate and promote cancer. DNA repair pathways are essential for preventing DNA damage from causing mutations and cytotoxicity, but RONS can interfere with repair mechanisms, reducing their efficacy. Further, cellular responses to DNA damage, such as damage signaling and cytotoxicity, can promote inflammation, creating a positive feedback loop. Despite coordination of DNA repair and oxidative stress responses, there are nevertheless examples whereby inflammation has been shown to promote mutagenesis, tissue damage, and ultimately carcinogenesis. Here, we discuss the DNA damage-mediated associations between inflammation, mutagenesis and cancer.
Collapse
Affiliation(s)
- Jennifer Kay
- Department of Biological Engineering, United States.
| | | | - Leona Samson
- Department of Biological Engineering, United States; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, United States
| | | |
Collapse
|
44
|
Tomicic MT, Steigerwald C, Rasenberger B, Brozovic A, Christmann M. Functional mismatch repair and inactive p53 drive sensitization of colorectal cancer cells to irinotecan via the IAP antagonist BV6. Arch Toxicol 2019; 93:2265-2277. [PMID: 31289894 DOI: 10.1007/s00204-019-02513-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/04/2019] [Indexed: 11/26/2022]
Abstract
A common strategy to overcome acquired chemotherapy resistance is the combination of a specific anticancer drug (e.g., topoisomerase I inhibitor irinotecan) together with a putative sensitizer. The purpose of this study was to analyze the cytostatic/cytotoxic response of colorectal carcinoma (CRC) cells to irinotecan, depending on the mismatch repair (MMR) and p53 status and to examine the impact of BV6, a bivalent antagonist of inhibitors of apoptosis c-IAP1/c-IAP2, alone or combined with irinotecan. Therefore, several MSH2- or MSH6-deficient cell lines were complemented for MMR deficiency, or MSH6 was knocked out/down in MMR-proficient cells. Upon irinotecan, MMR-deficient/p53-mutated lines repaired DNA double-strand breaks by homologous recombination less efficiently than MMR-proficient/p53-mutated lines and underwent elevated caspase-9-dependent apoptosis. Opposite, BV6-mediated sensitization was achieved only in MMR-proficient/p53-mutated cells. In those cells, c-IAP1 and c-IAP2 were effectively degraded by BV6, caspase-8 was fully activated, and both canonical and non-canonical NF-κB signaling were triggered. The results were confirmed ex vivo in tumor organoids from CRC patients. Therefore, the particular MMR+/p53mt signature, often found in non-metastasizing (stage II) CRC might be used as a prognostic factor for an adjuvant therapy using low-dose irinotecan combined with a bivalent IAP antagonist.
Collapse
Affiliation(s)
- Maja T Tomicic
- Department of Toxicology, University Medical Center Mainz, Obere Zahlbacher Str. 67, 55130, Mainz, Germany.
| | - Christian Steigerwald
- Department of Toxicology, University Medical Center Mainz, Obere Zahlbacher Str. 67, 55130, Mainz, Germany
| | - Birgit Rasenberger
- Department of Toxicology, University Medical Center Mainz, Obere Zahlbacher Str. 67, 55130, Mainz, Germany
| | - Anamaria Brozovic
- Department of Toxicology, University Medical Center Mainz, Obere Zahlbacher Str. 67, 55130, Mainz, Germany
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000, Zagreb, Croatia
| | - Markus Christmann
- Department of Toxicology, University Medical Center Mainz, Obere Zahlbacher Str. 67, 55130, Mainz, Germany
| |
Collapse
|
45
|
Chen X, Chen F, Ren Y, Weng G, Xu L, Xue X, Keng PC, Lee SO, Chen Y. IL-6 signaling contributes to radioresistance of prostate cancer through key DNA repair-associated molecules ATM, ATR, and BRCA 1/2. J Cancer Res Clin Oncol 2019; 145:1471-1484. [PMID: 31020420 DOI: 10.1007/s00432-019-02917-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/15/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE To study an association between IL-6 signaling and resistance to radiotherapy of prostate cancer (PCa) and explore the molecular mechanisms involved. METHODS IL-6 expressing C4-2 and CWR22Rv1 (C4-2IL-6/CWRIL-6) and vector control (C4-2vec/CWRvec) cell lines were developed. Radiation-sensitivities of these cells were compared in clonogenic assay, Comet assay, and γH2AX staining. In xenograft animal studies, radiation-sensitivity of C4-2IL-6 cell-derived tumors vs. C4-2vec cell-derived tumors was investigated. To reveal IL-6 downstream molecules involved in DNA repair after radiation, qPCR and Western blot analyses as well as immunofluorescence staining were performed. Transcriptional control of IL-6 on ATM and ATR molecules was also investigated. RESULTS We found C4-2IL-6 and CWRIL-6 cells survived better than their vector control cells after irradiation, and animal studies confirmed such in vitro results. We discovered that DNA repair-related molecules such as ATM, ATR, BRCA1, and BRCA2 were significantly upregulated in irradiated IL-6 expressing cells compared with vector control cells. We further defined that IL-6 signaling regulated cellular expressions of ATM and ATR at the transcriptional level through the activation of Stat3 signaling pathway. CONCLUSIONS IL-6 leads to PCa resistance to radiation through upregulation of DNA repair associated molecules ATM, ATR, BRCA1, and BRCA2.
Collapse
Affiliation(s)
- Xiaodong Chen
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, 315100, Zhejiang, People's Republic of China
| | - Feng Chen
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, 315100, Zhejiang, People's Republic of China
| | - Yu Ren
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, 315100, Zhejiang, People's Republic of China
| | - Guobin Weng
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, 315100, Zhejiang, People's Republic of China
| | - Lijun Xu
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, 315100, Zhejiang, People's Republic of China
| | - Xiang Xue
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA
| | - Peter C Keng
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA
| | - Soo Ok Lee
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA.
| | - Yuhchyau Chen
- Department of Radiation Oncology, James P. Wilmot Cancer Center, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Box 647, Rochester, NY, 14642, USA.
| |
Collapse
|
46
|
A Role for NF-κB in Organ Specific Cancer and Cancer Stem Cells. Cancers (Basel) 2019; 11:cancers11050655. [PMID: 31083587 PMCID: PMC6563002 DOI: 10.3390/cancers11050655] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/02/2019] [Accepted: 05/08/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) account for tumor initiation, invasiveness, metastasis, and recurrence in a broad range of human cancers. Although being a key player in cancer development and progression by stimulating proliferation and metastasis and preventing apoptosis, the role of the transcription factor NF-κB in cancer stem cells is still underestimated. In the present review, we will evaluate the role of NF-κB in CSCs of glioblastoma multiforme, ovarian cancer, multiple myeloma, lung cancer, colon cancer, prostate cancer, as well as cancer of the bone. Next to summarizing current knowledge regarding the presence and contribution of CSCs to the respective types of cancer, we will emphasize NF-κB-mediated signaling pathways directly involved in maintaining characteristics of cancer stem cells associated to tumor progression. Here, we will also focus on the status of NF-κB-activity predominantly in CSC populations and the tumor mass. Genetic alterations leading to NF-κB activity in glioblastoma, ependymoma, and multiple myeloma will be discussed.
Collapse
|
47
|
Kraya AA, Maxwell KN, Wubbenhorst B, Wenz BM, Pluta J, Rech AJ, Dorfman LM, Lunceford N, Barrett A, Mitra N, Morrissette JJD, Feldman M, Nayak A, Domchek SM, Vonderheide RH, Nathanson KL. Genomic Signatures Predict the Immunogenicity of BRCA-Deficient Breast Cancer. Clin Cancer Res 2019; 25:4363-4374. [PMID: 30914433 DOI: 10.1158/1078-0432.ccr-18-0468] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 12/08/2018] [Accepted: 03/15/2019] [Indexed: 12/19/2022]
Abstract
PURPOSE Breast cancers with BRCA1/2 alterations have a relatively high mutational load, suggesting that immune checkpoint blockade may be a potential treatment option. However, the degree of immune cell infiltration varies widely, and molecular features contributing to this variability remain unknown. EXPERIMENTAL DESIGN We hypothesized that genomic signatures might predict immunogenicity in BRCA1/2 breast cancers. Using The Cancer Genome Atlas (TCGA) genomic data, we compared breast cancers with (89) and without (770) either germline or somatic BRCA1/2 alterations. We also studied 35 breast cancers with germline BRCA1/2 mutations from Penn using WES and IHC. RESULTS We found that homologous recombination deficiency (HRD) scores were negatively associated with expression-based immune indices [cytolytic index (P = 0.04), immune ESTIMATE (P = 0.002), type II IFN signaling (P = 0.002)] despite being associated with a higher mutational/neoantigen burden, in BRCA1/2 mutant breast cancers. Further, absence of allele-specific loss of heterozygosity (LOH negative; P = 0.01) or subclonality (P = 0.003) of germline and somatic BRCA1/2 mutations, respectively, predicted for heightened cytolytic activity. Gene set analysis found that multiple innate and adaptive immune pathways that converge on NF-κB may contribute to this heightened immunogenicity. IHC of Penn breast cancers demonstrated increased CD45+ (P = 0.039) and CD8+ infiltrates (P = 0.037) and increased PDL1 expression (P = 0.012) in HRD-low or LOH-negative cancers. Triple-negative cancers with low HRD had far greater CD8+ T cells (P = 0.0011) and Perforin 1 expression (P = 0.014) compared with hormone receptor-positive HRD-high cancers. CONCLUSIONS HRD scores and hormone receptor subtype are predictive of immunogenicity in BRCA1/2 breast cancers and may inform the design of optimal immune therapeutic strategies.
Collapse
Affiliation(s)
- Adam A Kraya
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kara N Maxwell
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bradley Wubbenhorst
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brandon M Wenz
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - John Pluta
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andrew J Rech
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Liza M Dorfman
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nicole Lunceford
- Division of Surgical Pathology, Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia Pennsylvania
| | - Amanda Barrett
- Division of Surgical Pathology, Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia Pennsylvania
| | - Nandita Mitra
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jennifer J D Morrissette
- Division of Precision and Computational Diagnostics, Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael Feldman
- Division of Surgical Pathology, Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia Pennsylvania.,Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anupma Nayak
- Division of Surgical Pathology, Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia Pennsylvania
| | - Susan M Domchek
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Basser Center for BRCA, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert H Vonderheide
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Basser Center for BRCA, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Katherine L Nathanson
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania. .,Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Basser Center for BRCA, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
48
|
Mortezaee K, Najafi M, Farhood B, Ahmadi A, Shabeeb D, Musa AE. NF‐κB targeting for overcoming tumor resistance and normal tissues toxicity. J Cell Physiol 2019; 234:17187-17204. [DOI: 10.1002/jcp.28504] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/22/2019] [Accepted: 03/05/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy School of Medicine, Kurdistan University of Medical Sciences Sanandaj Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department School of Paramedical Sciences, Kermanshah University of Medical Sciences Kermanshah Iran
| | - Bagher Farhood
- Departments of Medical Physics and Radiology Faculty of Paramedical Sciences, Kashan University of Medical Sciences Kashan Iran
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Center Faculty of Pharmacy, Mazandaran University of Medical Sciences Sari Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology College of Medicine, University of Misan Misan Iraq
| | - Ahmed E. Musa
- Department of Medical Physics Tehran University of Medical Sciences (International Campus) Tehran Iran
| |
Collapse
|
49
|
Cardozo TR, De Carli RF, Seeber A, Flores WH, da Rosa JAN, Kotzal QSG, Lehmann M, da Silva FR, Dihl RR. Genotoxicity of zinc oxide nanoparticles: an in vivo and in silico study. Toxicol Res (Camb) 2019; 8:277-286. [PMID: 30997027 PMCID: PMC6430083 DOI: 10.1039/c8tx00255j] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/28/2019] [Indexed: 12/28/2022] Open
Abstract
Zinc oxide (ZnO) NPs are being used worldwide in consumer products and industrial applications. Based on predefined pathways, this study synthesized and characterized the nanostructures of ZnO NPs. The genotoxic effects of these nanomaterials were evaluated using a short-term in vivo bioassay, the somatic mutation and recombination test (SMART) in Drosophila melanogaster. In addition, a systems biology approach was used to search for known and predicted interaction networks between ZnO and proteins. The results observed in this study after in vivo exposure indicate that ZnO NPs are genotoxic and that homologous recombination (HR) was the main mechanism inducing loss of heterozygosis in the somatic cells of D. melanogaster. The results of in silico analysis indicated that ZnO is associated with the nuclear factor-kappa-beta (NFKB) protein family. In accordance with this model, ZnO exposure decreases the levels of NFKB inhibitory protein in the cell, consequently increasing NFKB dimers in the nucleus and inducing DNA double strand breaks (DSB) repair via HR. This excess level of HR can be observed in the SMART results. Assessing the mutagenic/recombinagenic effect of nanomaterials is essential in the development of strategies to protect human and environmental integrity.
Collapse
Affiliation(s)
- Tatiane R Cardozo
- Laboratory of Genetic Toxicity , Graduate Program in Molecular and Cellular Biology Applied to Health , Lutheran University of Brazil (ULBRA) , Canoas , RS , Brazil . ; ; Tel: + 55 51 34779219
- Federal University of Pampa - Research Group on Nanostructured Materials , Campus Bagé , RS , Brazil
| | - Raíne F De Carli
- Laboratory of Genetic Toxicity , Graduate Program in Molecular and Cellular Biology Applied to Health , Lutheran University of Brazil (ULBRA) , Canoas , RS , Brazil . ; ; Tel: + 55 51 34779219
| | - Allan Seeber
- Federal University of Pampa - Research Group on Nanostructured Materials , Campus Bagé , RS , Brazil
| | - Wladimir H Flores
- Federal University of Pampa - Research Group on Nanostructured Materials , Campus Bagé , RS , Brazil
| | - Jordana A N da Rosa
- Laboratory of Genetic Toxicity , Graduate Program in Molecular and Cellular Biology Applied to Health , Lutheran University of Brazil (ULBRA) , Canoas , RS , Brazil . ; ; Tel: + 55 51 34779219
| | - Queila S G Kotzal
- Laboratory of Genetic Toxicity , Graduate Program in Molecular and Cellular Biology Applied to Health , Lutheran University of Brazil (ULBRA) , Canoas , RS , Brazil . ; ; Tel: + 55 51 34779219
| | - Mauricio Lehmann
- Laboratory of Genetic Toxicity , Graduate Program in Molecular and Cellular Biology Applied to Health , Lutheran University of Brazil (ULBRA) , Canoas , RS , Brazil . ; ; Tel: + 55 51 34779219
| | - Fernanda R da Silva
- La Salle University , Master's Degree in Environmental Impact Evaluation , Canoas , RS , Brazil
| | - Rafael R Dihl
- Laboratory of Genetic Toxicity , Graduate Program in Molecular and Cellular Biology Applied to Health , Lutheran University of Brazil (ULBRA) , Canoas , RS , Brazil . ; ; Tel: + 55 51 34779219
| |
Collapse
|
50
|
Cognition-Enhancing Vagus Nerve Stimulation Alters the Epigenetic Landscape. J Neurosci 2019; 39:3454-3469. [PMID: 30804093 DOI: 10.1523/jneurosci.2407-18.2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 02/15/2019] [Accepted: 02/16/2019] [Indexed: 01/27/2023] Open
Abstract
Vagus nerve stimulation (VNS) has been shown to enhance learning and memory, yet the mechanisms behind these enhancements are unknown. Here, we present evidence that epigenetic modulation underlies VNS-induced improvements in cognition. We show that VNS enhances novelty preference (NP); alters the hippocampal, cortical, and blood epigenetic transcriptomes; and epigenetically modulates neuronal plasticity and stress-response signaling genes in male Sprague Dawley rats. Brain-behavior analysis revealed structure-specific relationships between NP test performance (NPTP) and epigenetic alterations. In the hippocampus, NPTP correlated with decreased histone deacetylase 11 (HDAC11), a transcriptional repressor enriched in CA1 cells important for memory consolidation. In the cortex, the immediate early gene (IEG) ARC was increased in VNS rats and correlated with transcription of plasticity genes and epigenetic regulators, including HDAC3. For rats engaged in NPTP, ARC correlated with performance. Interestingly, blood ARC transcripts decreased in VNS rats performing NPTP, but increased in VNS-only rats. Because DNA double-strand breaks (DSBs) facilitate transcription of IEGs, we investigated phosphorylated H2A.X (γH2A.X), a histone modification known to colocalize with DSBs. In agreement with reduced cortical stress-response transcription factor NF-κB1, chromatin immunoprecipitation revealed reduced γH2A.X in the ARC promoter. Surprisingly, VNS did not significantly reduce transcription of cortical or hippocampal proinflammatory cytokines. However, TNFRSF11B (osteoprotegerin) correlated with NPTP as well as plasticity, stress-response signaling, and epigenetic regulation transcripts in both hippocampus and cortex. Together, our findings provide the first evidence that VNS induces widespread changes in the cognitive epigenetic landscape and specifically affects epigenetic modulators associated with NPTP, stress-response signaling, memory consolidation, and cortical neural remodeling.SIGNIFICANCE STATEMENT Recent studies have implicated vagus nerve stimulation (VNS) in enhanced learning and memory. However, whereas epigenetic modifications are known to play an important role in memory, the particular mechanisms involved in VNS-enhanced cognition are unknown. In this study, we examined brain and behavior changes in VNS and sham rats performing a multiday novelty preference (NP) task. We found that VNS activated specific histone modifications and DNA methylation changes at important stress-response signaling and plasticity genes. Both cortical and hippocampal plasticity changes were predictive of NP test performance. Our results reveal important epigenetic alterations associated with VNS cognitive improvements, as well as new potential pharmacological targets for enhancing cortical and hippocampal plasticity.
Collapse
|