1
|
Galal SM, El Kiki SM, Elgazzar EM. The Potential Therapeutic Approach of Ursodeoxycholic Acid as a Potent Activator of ACE-2 on Cerebral Disorders Induced by γ-irradiation in Rats. Cell Biochem Funct 2024; 42:e70024. [PMID: 39660593 DOI: 10.1002/cbf.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/05/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024]
Abstract
The present investigation assesses ursodeoxycholic acid's efficacy (UDCA) as an ACE2 activator against gamma irradiation through activating the renin-angiotensin system's (RAS) beneficial axis, ACE2/Ang-(1-7)/Mas1 via its profitable influence on inflammation, oxidative stress, and neuronal damage caused by irradiation (IRR). Four groups of rats were treated as follows: control group, group receiving UDCA (100 mg/kg/day) for 14 days by gavage, group irradiated at 6 Gy, and group receiving UDCA post-irradiation for 14 days. The results revealed that gamma-irradiation (6 Gy) caused a substantial drop in the cerebral ACE2/Ang-(1-7)/Mas1 axis and remarkably increased the expression of cerebral inflammatory mediators: tumor necrosis factor-α (TNF-α), nuclear factor kappa-B (NF-κB), interleukin-6 (IL-6) and interleukin-1β (IL-1β) combined with significant elevation in cyclooxygenase-II (COX-II), (NADPH) oxidases (NOX4), lipooxygenase (LOX) activities and nitric oxide (NO) content. Moreover, it greatly enhanced the reduction in N-methyl-d-aspartate (NMDA) level, while dramatically increasing gamma-aminobutyric acid (GABA) level and neuronal nitric oxide synthases (nNOS) enzyme activity in cerebral tissue homogenate. Irradiated rats' brain sections underwent histological investigation using hematoxylin and eosin staining, which revealed cellular damage and a pathological appearance. The administration of UDCA inverts these unusual alterations. In conclusion, UDCA treatment efficiently normalizes the above-mentioned pathological abnormalities and avoids the development of IRR-associated neurological dysfunction by upregulating the beneficial axis of RAS in the brain. Hence, ursodeoxycholic acid presents a novel option for patient care during radiotherapy.
Collapse
Affiliation(s)
- Shereen Mohamed Galal
- Health Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Shereen Mohamed El Kiki
- Health Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Eman Mahmoud Elgazzar
- Health Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
2
|
Saha B, Pallatt S, Banerjee A, Banerjee AG, Pathak R, Pathak S. Current Insights into Molecular Mechanisms and Potential Biomarkers for Treating Radiation-Induced Liver Damage. Cells 2024; 13:1560. [PMID: 39329744 PMCID: PMC11429644 DOI: 10.3390/cells13181560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Highly conformal delivery of radiation therapy (RT) has revolutionized the treatment landscape for primary and metastatic liver cancers, yet concerns persist regarding radiation-induced liver disease (RILD). Despite advancements, RILD remains a major dose-limiting factor due to the potential damage to normal liver tissues by therapeutic radiation. The toxicity to normal liver tissues is associated with a multitude of physiological and pathological consequences. RILD unfolds as multifaceted processes, intricately linking various responses, such as DNA damage, oxidative stress, inflammation, cellular senescence, fibrosis, and immune reactions, through multiple signaling pathways. The DNA damage caused by ionizing radiation (IR) is a major contributor to the pathogenesis of RILD. Moreover, current treatment options for RILD are limited, with no established biomarker for early detection. RILD diagnosis often occurs at advanced stages, highlighting the critical need for early biomarkers to adjust treatment strategies and prevent liver failure. This review provides an outline of the diverse molecular and cellular mechanisms responsible for the development of RILD and points out all of the available biomarkers for early detection with the aim of helping clinicians decide on advance treatment strategies from a single literature recourse.
Collapse
Affiliation(s)
- Biki Saha
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Sneha Pallatt
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Antara Banerjee
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| | - Abhijit G. Banerjee
- R&D, Genomic Bio-Medicine Research and Incubation (GBMRI), Durg 491001, Chhattisgarh, India
| | - Rupak Pathak
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Surajit Pathak
- Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai 603103, India
| |
Collapse
|
3
|
Zhang C, Liu J, Wu J, Ranjan K, Cui X, Wang X, Zhang D, Zhu S. Key molecular DNA damage responses of human cells to radiation. Front Cell Dev Biol 2024; 12:1422520. [PMID: 39050891 PMCID: PMC11266142 DOI: 10.3389/fcell.2024.1422520] [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: 04/24/2024] [Accepted: 06/19/2024] [Indexed: 07/27/2024] Open
Abstract
Our understanding of the DNA damage responses of human cells to radiation has increased remarkably over the recent years although some notable signaling events remain to be discovered. Here we provide a brief account of the key molecular events of the responses to reflect the current understanding of the key underlying mechanisms involved.
Collapse
Affiliation(s)
- Chencheng Zhang
- Cancer Research Center, Nantong Tumor Hospital, Nantong, China
- Cancer Research Institute, The Affiliated Tumor Hospital of Nantong University, Nantong, China
- Cancer Research Center, Nantong, China
| | - Jibin Liu
- Cancer Research Center, Nantong Tumor Hospital, Nantong, China
- Cancer Research Institute, The Affiliated Tumor Hospital of Nantong University, Nantong, China
- Cancer Research Center, Nantong, China
| | - Jun Wu
- Nantong Tumor Hospital, Nantong, China
| | - Kamakshi Ranjan
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States
| | - Xiaopeng Cui
- Department of General Surgery, The Affiliated Hospital of Nantong University, Nantong, China
| | - Xingdan Wang
- Department of Radiotherapy, Nantong Tumor Hospital, The Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Dianzheng Zhang
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States
| | - Shudong Zhu
- Cancer Research Center, Nantong Tumor Hospital, Nantong, China
- Cancer Research Institute, The Affiliated Tumor Hospital of Nantong University, Nantong, China
- Cancer Research Center, Nantong, China
- Argus Pharmaceuticals, Changsha, China
| |
Collapse
|
4
|
Kura B, Pavelkova P, Kalocayova B, Pobijakova M, Slezak J. MicroRNAs as Regulators of Radiation-Induced Oxidative Stress. Curr Issues Mol Biol 2024; 46:7097-7113. [PMID: 39057064 PMCID: PMC11276491 DOI: 10.3390/cimb46070423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
microRNAs (miRNAs) represent small RNA molecules involved in the regulation of gene expression. They are implicated in the regulation of diverse cellular processes ranging from cellular homeostasis to stress responses. Unintended irradiation of the cells and tissues, e.g., during medical uses, induces various pathological conditions, including oxidative stress. miRNAs may regulate the expression of transcription factors (e.g., nuclear factor erythroid 2 related factor 2 (Nrf2), nuclear factor kappa B (NF-κB), tumor suppressor protein p53) and other redox-sensitive genes (e.g., mitogen-activated protein kinase (MAPKs), sirtuins (SIRTs)), which trigger and modulate cellular redox signaling. During irradiation, miRNAs mainly act with reactive oxygen species (ROS) to regulate the cell fate. Depending on the pathway involved and the extent of oxidative stress, this may lead to cell survival or cell death. In the context of radiation-induced oxidative stress, miRNA-21 and miRNA-34a are among the best-studied miRNAs. miRNA-21 has been shown to directly target superoxide dismutase (SOD), or NF-κB, whereas miRNA-34a is a direct regulator of NADPH oxidase (NOX), SIRT1, or p53. Understanding the mechanisms underlying radiation-induced injury including the involvement of redox-responsive miRNAs may help to develop novel approaches for modulating the cellular response to radiation exposure.
Collapse
Affiliation(s)
- Branislav Kura
- Centre of Experimental Medicine, Slovak Academy of Sciences, Dubravska Cesta 9, 841 04 Bratislava, Slovakia; (P.P.); (B.K.); (J.S.)
| | - Patricia Pavelkova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Dubravska Cesta 9, 841 04 Bratislava, Slovakia; (P.P.); (B.K.); (J.S.)
| | - Barbora Kalocayova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Dubravska Cesta 9, 841 04 Bratislava, Slovakia; (P.P.); (B.K.); (J.S.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, 832 32 Bratislava, Slovakia
| | - Margita Pobijakova
- Department of Radiation Oncology, Bory Hospital–Penta Hospitals, 841 03 Bratislava, Slovakia;
- Radiological Science, Faculty of Nursing and Medical Professional Studies, Slovak Medical University, 831 01 Bratislava, Slovakia
| | - Jan Slezak
- Centre of Experimental Medicine, Slovak Academy of Sciences, Dubravska Cesta 9, 841 04 Bratislava, Slovakia; (P.P.); (B.K.); (J.S.)
| |
Collapse
|
5
|
El-Sabbagh WA, Fadel NA, El-Hazek RM, Osman AH, Ramadan LA. Ubiquinol attenuates γ-radiation induced coronary and aortic changes via PDGF/p38 MAPK/ICAM-1 related pathway. Sci Rep 2023; 13:22959. [PMID: 38151557 PMCID: PMC10752893 DOI: 10.1038/s41598-023-50218-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/16/2023] [Indexed: 12/29/2023] Open
Abstract
Endothelial vascular injury is one of the most pivotal disorders emerging during radiotherapy. It is crucial to rely on strong antioxidants to defend against vascular damage. The current study was carried out to investigate the ameliorative effect of ubiquinol (Ubq) against gamma (γ)-radiation induced aortic and coronary changes, with highlighting its role in suppression of p38 mitogen activated protein kinase (MAPK). Exposure to γ-radiation was adopted as a potent detrimental model that induces vascular tissue damage. Concisely, male albino rats were irradiated at a dose level of 7 Gy and treated daily with Ubq (10 mg/kg/day, p.o.) for 7 days pre-and post-irradiation. At the end of the experiment, lipid profile, 8-hydroxydeoxyguanosine (8-OHdG), gene expression of intercellular adhesion molecule (ICAM-1), platelet derived growth factor (PDGF), p38 MAPK and matrix metalloproteinase-9 (MMP-9) were estimated. Exposure to radiation significantly deteriorates aortic and coronary tissues. Conversely, administration of Ubq significantly reduced serum t-cholesterol, LDL and triglycerides (p = 0.001). In addition, Ubq prevented oxidative DNA damage (8-OHdG) (p = 0.1) and reduced serum MMP-9 (p = 0.001) which contributed to the endothelial cells damage. The positive impact of Ubq was more apparent in suppression of both PDGF (p = 0.001) and p38 MAPK (p = 0.1) protein concentrations, leading subsequently in reduction of ICAM-1 (p = 0.001) gene expression. As a conclusion, vascular endothelial damage brought on by γ-radiation is one of the leading causes of coronary and aortic deteriorations which could be successfully mitigated by Ubq.
Collapse
Affiliation(s)
- Walaa A El-Sabbagh
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Noha A Fadel
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt.
| | - Rania M El-Hazek
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Ahmed H Osman
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Laila A Ramadan
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Russian University (ERU), Cairo, Egypt
| |
Collapse
|
6
|
A Monte Carlo simulation study of L-band emission upon gamma radiolysis of water. Radiat Phys Chem Oxf Engl 1993 2023. [DOI: 10.1016/j.radphyschem.2023.110883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
7
|
Luo H, Liu X, Liu H, Wang Y, Xu K, Li J, Liu M, Guo J, Qin X. ACT001 Ameliorates ionizing radiation-induced lung injury by inhibiting NLRP3 inflammasome pathway. Biomed Pharmacother 2023; 163:114808. [PMID: 37146417 DOI: 10.1016/j.biopha.2023.114808] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/26/2023] [Accepted: 04/30/2023] [Indexed: 05/07/2023] Open
Abstract
Radiotherapy is a prevalent treatment modality for thoracic tumors; however, it can lead to radiation-induced lung injury (RILI), which currently lacks effective interventions. ACT001, a prodrug of micheliolide, has demonstrated promising clinical application potential, yet its impact on RILI requires further validation. This study aims to investigate the radioprotective effects of ACT001 on RILI and elucidate its underlying mechanism. Sprague-Dawley rats were utilized to induce RILI following 20 Gy X-ray chest irradiation, and lung tissue inflammation and fibrosis were assessed using hematoxylin and eosin (H&E) and Masson staining. Lung injury, inflammation, and oxidative stress markers were evaluated employing commercial kits. Pyroptosis-related differentially expressed genes (DEGs) were analyzed using a microarray dataset from the Gene Expression Omnibus (GEO) database, and their functions and hub genes were identified through protein-protein interaction networks. Pyroptosis-related genes were detected via RT-qPCR, western blotting, immunofluorescence, and immunohistochemistry. The results demonstrated that ACT001 ameliorated RILI, diminished pro-inflammatory cytokine release and fibrosis, and mitigated the activation of the NLRP3 inflammasome while inhibiting pyroptosis in lung tissue. In conclusion, our study reveals that ACT001 can suppress NLRP3 inflammasome-mediated pyroptosis and improve RILI, suggesting its potential as a novel protective agent for RILI.
Collapse
Affiliation(s)
- Hao Luo
- Shanxi Provincial Key Laboratory of Drug Toxicology and Radiation Damage Drugs, Department of Radiology and Environmental Medicine, China Institute For Radiation Protection, Taiyuan, China
| | - Xiaoming Liu
- Shanxi Provincial Key Laboratory of Drug Toxicology and Radiation Damage Drugs, Department of Radiology and Environmental Medicine, China Institute For Radiation Protection, Taiyuan, China
| | - Huan Liu
- Shanxi Provincial Key Laboratory of Drug Toxicology and Radiation Damage Drugs, Department of Radiology and Environmental Medicine, China Institute For Radiation Protection, Taiyuan, China
| | - Yong Wang
- Shanxi Provincial Key Laboratory of Drug Toxicology and Radiation Damage Drugs, Department of Radiology and Environmental Medicine, China Institute For Radiation Protection, Taiyuan, China; School of Forensics, Shanxi Medical University, Taiyuan, China
| | - Kai Xu
- Shanxi Provincial Key Laboratory of Drug Toxicology and Radiation Damage Drugs, Department of Radiology and Environmental Medicine, China Institute For Radiation Protection, Taiyuan, China
| | - Jianhua Li
- Shanxi Provincial Key Laboratory of Drug Toxicology and Radiation Damage Drugs, Department of Radiology and Environmental Medicine, China Institute For Radiation Protection, Taiyuan, China
| | - Mengya Liu
- Shanxi Provincial Key Laboratory of Drug Toxicology and Radiation Damage Drugs, Department of Radiology and Environmental Medicine, China Institute For Radiation Protection, Taiyuan, China
| | - Jianshuang Guo
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.
| | - Xiujun Qin
- Shanxi Provincial Key Laboratory of Drug Toxicology and Radiation Damage Drugs, Department of Radiology and Environmental Medicine, China Institute For Radiation Protection, Taiyuan, China.
| |
Collapse
|
8
|
Zhou YJ, Tang Y, Liu SJ, Zeng PH, Qu L, Jing QC, Yin WJ. Radiation-induced liver disease: beyond DNA damage. Cell Cycle 2023; 22:506-526. [PMID: 36214587 PMCID: PMC9928481 DOI: 10.1080/15384101.2022.2131163] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 11/03/2022] Open
Abstract
Radiation-induced liver disease (RILD), also known as radiation hepatitis, is a serious side effect of radiotherapy (RT) for hepatocellular carcinoma. The therapeutic dose of RT can damage normal liver tissue, and the toxicity that accumulates around the irradiated liver tissue is related to numerous physiological and pathological processes. RILD may restrict treatment use or eventually deteriorate into liver fibrosis. However, the research on the mechanism of radiation-induced liver injury has seen little progress compared with that on radiation injury in other tissues, and no targeted clinical pharmacological treatment for RILD exists. The DNA damage response caused by ionizing radiation plays an important role in the pathogenesis and development of RILD. Therefore, in this review, we systematically summarize the molecular and cellular mechanisms involved in RILD. Such an analysis is essential for preventing the occurrence and development of RILD and further exploring the potential treatment of this disease.
Collapse
Affiliation(s)
- Ying Jie Zhou
- Department of Clinical Laboratory Medicine, Institution of microbiology and infectious diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yun Tang
- Department of Clinical Laboratory Medicine, Institution of microbiology and infectious diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Si Jian Liu
- Department of Clinical Laboratory Medicine, Institution of microbiology and infectious diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Peng Hui Zeng
- Department of Clinical Laboratory Medicine, Institution of microbiology and infectious diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Li Qu
- Department of Clinical Laboratory Medicine, Institution of microbiology and infectious diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qian Cheng Jing
- The Affiliated Changsha Central Hospital, Department of Otolaryngology Head and Neck Surgery,Hengyang Medical School, University of South China, Changsha, Hunan, China
- Institute of Otolaryngology Head and Neck Surgery, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Wen Jun Yin
- Department of Clinical Laboratory Medicine, Institution of microbiology and infectious diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory, Changsha Central Hospital, University of South China, Changsha, Hunan, China
| |
Collapse
|
9
|
DiCarlo AL, Cassatt DR, Rios CI, Satyamitra MM, Zhang Y, Golden TG, Taliaferro LP. Making connections: the scientific impact and mentoring legacy of Dr. John E. Moulder. Int J Radiat Biol 2023:1-7. [PMID: 36763099 DOI: 10.1080/09553002.2023.2176563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
PURPOSE The intent of this mini review is to pay homage to Dr. John E. Moulder's long and successful career in radiation science with the Medical College of Wisconsin. This effort will be done from the perspective of his history of U.S. Government funding for research into the biological pathways involved in radiation-induced normal tissue injuries, especially damage to the kidneys and heart, and pharmacological interventions. In addition, the impact of his steady guidance and leadership in the mentoring of junior scientists, and the development of meaningful collaborations with other researchers will be highlighted. CONCLUSION Dr. John E. Moulder's contributions to the field of radiation research, through his strong character and reputation, his consistent and dedicated commitment to his colleagues and students, and his significant scientific advances, have been critical to moving the science forward, and will not be forgotten by those who knew him personally or through publications documenting his important work.
Collapse
Affiliation(s)
- Andrea L DiCarlo
- Radiation and Nuclear Countermeasures Program; Division of Allergy, Immunology, and Transplantation; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - David R Cassatt
- Radiation and Nuclear Countermeasures Program; Division of Allergy, Immunology, and Transplantation; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Carmen I Rios
- Radiation and Nuclear Countermeasures Program; Division of Allergy, Immunology, and Transplantation; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Merriline M Satyamitra
- Radiation and Nuclear Countermeasures Program; Division of Allergy, Immunology, and Transplantation; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Yuji Zhang
- Department of Epidemiology and Public Health, Marlene and Stewart Greenbaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Trevor G Golden
- Radiation and Nuclear Countermeasures Program; Division of Allergy, Immunology, and Transplantation; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Lanyn P Taliaferro
- Radiation and Nuclear Countermeasures Program; Division of Allergy, Immunology, and Transplantation; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
10
|
Cardiac fibrosis in oncologic therapies. CURRENT OPINION IN PHYSIOLOGY 2022; 29. [DOI: 10.1016/j.cophys.2022.100575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
11
|
Liu J, Chen J, Xu B, Lin L, Liu S, Ma X, Liu J. 3,4,5-O-tricaffeoylquinic acid with anti-radiation activity suppresses LPS-induced NLRP3 inflammasome activation via autophagy in THP-1 macrophages. Mol Immunol 2022; 147:187-198. [PMID: 35633614 DOI: 10.1016/j.molimm.2022.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/09/2022] [Accepted: 05/18/2022] [Indexed: 10/18/2022]
Abstract
Damage to normal tissues caused by excessive ionizing radiation (IR) exposure is the major side effect of radiotherapy. Several recent studies have shown that IR-induced damage to tissues leads to a systemic immune response and NLRP3 inflammasome activation in immune cells. 3,4,5-O-tricaffeoylquinic acid (tCQA), extracted from the natural plant Azolla imbricata, relieves inflammation and has radioprotective function. Here, we aimed to investigate the inhibitory effect and molecular mechanism of tCQA on IR-induced NLRP3 inflammasome activation. First, the results of ELISA and qPCR assays showed that tCQA has anti-inflammatory effects in THP-1 cell line and healthy human peripheral blood mononuclear cells. Western blotting and ELISA suggested tCQA could inhibit NF-κB/MAPK signaling pathway, NLRP3 expression and the secretion of IL-1β in lipopolysaccharide (LPS)-stimulated THP-1 macrophages. Then, flow cytometry, LDH assay and western blotting demonstrated that tCQA could inhibit LPS- and nigericin-induced Caspase-1 activation and gasdermin D cleavage, thereby suppressing inflammatory cell death. Furthermore, we found that the autophagy inhibitor chloroquine, not the proteasome inhibitor MG132, could counteract the promoting effect of tCQA on NLRP3 degradation and the inhibitory effect on cell death. Western blotting and autophagosome staining results suggested tCQA could significantly enhance LPS-induced autophagic flux in macrophages and ATG5/ATG7 knockdown reverses the inhibitory effect of tCQA on NLRP3 expression and Caspase-1 activation, indicating that tCQA induces NLRP3 degradation via autophagy. Finally, THP-1 macrophages and BALB/c mice were irradiated with 137Cs γ-rays and tCQA could inhibit IR-induced NLRP3 inflammasome activation both in vitro and in vivo. To conclude, tCQA controls inflammation and NLRP3 inflammasome activation in vitro via NF-κB/MAPK signaling pathway and autophagy, meanwhile inhibits IR-induced NLRP3 inflammasome activation in vivo. Overall, our study provides an experimental and theoretical basis for the application of tCQA as a radioprotectant in clinical radiotherapy.
Collapse
Affiliation(s)
- Jiajun Liu
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Jingyun Chen
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Baixue Xu
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Long Lin
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Shaoqun Liu
- Key Laboratory of Whole-period Monitoring and Precise Intervention of Digestive Cancer (SMHC), Minhang Hosipital & AHS, Fudan University, Shanghai, 201199, PR China; Department of Surgery, Minhang Hospital, Fudan University, Shanghai, 201199, PR China.
| | - Xiaoying Ma
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China.
| | - Jianwen Liu
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, PR China.
| |
Collapse
|
12
|
Zhang DM, Deng JJ, Wu YG, Tang T, Xiong L, Zheng YF, Xu XM. MicroRNA-223-3p Protect Against Radiation-Induced Cardiac Toxicity by Alleviating Myocardial Oxidative Stress and Programmed Cell Death via Targeting the AMPK Pathway. Front Cell Dev Biol 2022; 9:801661. [PMID: 35111759 PMCID: PMC8801819 DOI: 10.3389/fcell.2021.801661] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Objectives: Radiotherapy improves the survival rate of cancer patients, yet it also involves some inevitable complications. Radiation-induced heart disease (RIHD) is one of the most serious complications, especially the radiotherapy of thoracic tumors, which is characterized by cardiac oxidative stress disorder and programmed cell death. At present, there is no effective treatment strategy for RIHD; in addition, it cannot be reversed when it progresses. This study aims to explore the role and potential mechanism of microRNA-223-3p (miR-223-3p) in RIHD.Methods: Mice were injected with miR-223-3p mimic, inhibitor, or their respective controls in the tail vein and received a single dose of 20 Gy whole-heart irradiation (WHI) for 16 weeks after 3 days to construct a RIHD mouse model. To inhibit adenosine monophosphate activated protein kinase (AMPK) or phosphodiesterase 4D (PDE4D), compound C (CompC) and AAV9-shPDE4D were used.Results: WHI treatment significantly inhibited the expression of miR-223-3p in the hearts; furthermore, the levels of miR-223-3p decreased in a radiation time-dependent manner. miR-223-3p mimic significantly relieved, while miR-223-3p inhibitor aggravated apoptosis, oxidative damage, and cardiac dysfunction in RIHD mice. In addition, we found that miR-223-3p mimic improves WHI-induced myocardial injury by activating AMPK and that the inhibition of AMPK by CompC completely blocks these protective effects of miR-223-3p mimic. Further studies found that miR-223-3p lowers the protein levels of PDE4D and inhibiting PDE4D by AAV9-shPDE4D blocks the WHI-induced myocardial injury mediated by miR-223-3p inhibitor.Conclusion: miR-223-3p ameliorates WHI-induced RIHD through anti-oxidant and anti-programmed cell death mechanisms via activating AMPK by PDE4D regulation. miR-223-3p mimic exhibits potential value in the treatment of RIHD.
Collapse
Affiliation(s)
- Dao-ming Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jun-jian Deng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yao-gui Wu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tian Tang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lin Xiong
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yong-fa Zheng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Yong-fa Zheng, ; Xi-ming Xu,
| | - Xi-ming Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Yong-fa Zheng, ; Xi-ming Xu,
| |
Collapse
|
13
|
Azab KS, Maarouf RE, Abdel-Rafei MK, El Bakary NM, Thabet NM. Withania somnifera (Ashwagandha) root extract counteract acute and chronic impact of γ-radiation on liver and spleen of rats. Hum Exp Toxicol 2022; 41:9603271221106344. [PMID: 35656930 DOI: 10.1177/09603271221106344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The exposure to ionizing radiation has become inescapably because of increased dependence on radiation to execute works in different fields and also its influences on biological systems. Thus, the current study aimed at examination of the radio-protective effect of the natural ashwagandha (Ag) against acute and chronic doses of γ-radiation on liver and spleen of rats. The impact of Ag was inspected in rats exposed to acute exposure of 8 Gy (single dose) or to chronic exposure of 8 Gy (2 Gy every other day for 4 times). The data obtained reveals significant amelioration of the redox status (MDA, GSH and ROS) in spleen and liver tissues of rats treated with Ag and exposed to the 2-different modes of γ-radiation. Besides, the changes in inflammatory responses assessed by measurements of IL-17, IL-10 and α7-nAchR are less pronounced in rats received Ag and γ-radiation compared to irradiated rats. Further, the measurements of tissues structural damage markers (MMP-2, MMP-9 and TIMP-1) pointed to benefit of Ag against γ-radiation. The histopathological investigation of spleen and liver tissues confirmed this ameliorating action of Ag counter to γ-radiation hazards. It could be suggested that Ashwagandha could exerts radio-protective influences because of its antioxidants and anti-inflammatory capabilities.
Collapse
Affiliation(s)
- Khaled Sh Azab
- Department Radiation Biology, National Centre for Radiation Research and Technology (NCRRT), 68892Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Rokaya E Maarouf
- Department Radiation Biology, National Centre for Radiation Research and Technology (NCRRT), 68892Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Mohamed K Abdel-Rafei
- Department Radiation Biology, National Centre for Radiation Research and Technology (NCRRT), 68892Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Nermeen M El Bakary
- Department Radiation Biology, National Centre for Radiation Research and Technology (NCRRT), 68892Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Noura M Thabet
- Department Radiation Biology, National Centre for Radiation Research and Technology (NCRRT), 68892Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
14
|
Wei J, Zhao Q, Zhang Y, Shi W, Wang H, Zheng Z, Meng L, Xin Y, Jiang X. Sulforaphane-Mediated Nrf2 Activation Prevents Radiation-Induced Skin Injury through Inhibiting the Oxidative-Stress-Activated DNA Damage and NLRP3 Inflammasome. Antioxidants (Basel) 2021; 10:antiox10111850. [PMID: 34829721 PMCID: PMC8614868 DOI: 10.3390/antiox10111850] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 11/16/2022] Open
Abstract
This article mainly observed the protective effect of sulforaphane (SFN) on radiation-induced skin injury (RISI). In addition, we will discuss the mechanism of SFN's protection on RISI. The RISI model was established by the irradiation of the left thigh under intravenous anesthesia. Thirty-two C57/BL6 mice were randomly divided into control group (CON), SFN group, irradiation (IR) group, and IR plus SFN (IR/SFN) group. At eight weeks after irradiation, the morphological changes of mouse skin tissues were detected by H&E staining. Then, the oxidative stress and inflammatory response indexes in mouse skin tissues, as well as the expression of Nrf2 and its downstream antioxidant genes, were evaluated by ELISA, real-time PCR, and Western blotting. The H&E staining showed the hyperplasia of fibrous tissue in the mouse dermis and hypodermis of the IR group. Western blotting and ELISA results showed that the inflammasome of NLRP3, caspase-1, and IL-1β, as well as oxidative stress damage indicators ROS, 4-HNE, and 3-NT, in the skin tissues of mice in the IR group were significantly higher than those in the control group (p < 0.05). However, the above pathological changes declined sharply after SFN treatment (p < 0.05). In addition, the expressions of Nrf2 and its regulated antioxidant enzymes, including CAT and HO-1, were higher in the skin tissues of SFN and IR/SFN groups, but lower in the control and IR groups (p < 0.05). SFN may be able to suppress the oxidative stress by upregulating the expression and function of Nrf2, and subsequently inhibiting the activation of NLRP3 inflammasome and DNA damage, so as to prevent and alleviate the RISI.
Collapse
Affiliation(s)
- Jinlong Wei
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (J.W.); (Q.Z.); (Y.Z.); (W.S.); (H.W.); (Z.Z.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Qin Zhao
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (J.W.); (Q.Z.); (Y.Z.); (W.S.); (H.W.); (Z.Z.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Yuyu Zhang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (J.W.); (Q.Z.); (Y.Z.); (W.S.); (H.W.); (Z.Z.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Weiyan Shi
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (J.W.); (Q.Z.); (Y.Z.); (W.S.); (H.W.); (Z.Z.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Huanhuan Wang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (J.W.); (Q.Z.); (Y.Z.); (W.S.); (H.W.); (Z.Z.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Zhuangzhuang Zheng
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (J.W.); (Q.Z.); (Y.Z.); (W.S.); (H.W.); (Z.Z.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Lingbin Meng
- Department of Hematology and Medical Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA;
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
- Correspondence: (Y.X.); ; (X.J.); Tel.: +86-13504310452 (Y.X.); +86-15804302750 (X.J.)
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; (J.W.); (Q.Z.); (Y.Z.); (W.S.); (H.W.); (Z.Z.)
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
- Correspondence: (Y.X.); ; (X.J.); Tel.: +86-13504310452 (Y.X.); +86-15804302750 (X.J.)
| |
Collapse
|
15
|
Zhu W, Zhang X, Yu M, Lin B, Yu C. Radiation-induced liver injury and hepatocyte senescence. Cell Death Discov 2021; 7:244. [PMID: 34531376 PMCID: PMC8446062 DOI: 10.1038/s41420-021-00634-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 08/06/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022] Open
Abstract
Radiation-induced liver injury (RILI) is a major complication of radiotherapy during treatment for liver cancer and other upper abdominal malignant tumors that has poor pharmacological therapeutic options. A series of pathological changes can be induced by radiation. However, the underlying mechanism of RILI remains unclear. Radiation can induce cell damage via direct energy deposition or reactive free radical generation. Cellular senescence can be observed due to the DNA damage response (DDR) caused by radiation. The senescence-associated secretory phenotype (SASP) secreted from senescent cells can cause chronic inflammation and aggravate liver dysfunction for a long time. Oxidative stress further activates the signaling pathway of the inflammatory response and affects cellular metabolism. miRNAs clearly have differential expression after radiation treatment and take part in RILI development. This review aims to systematically profile the overall mechanism of RILI and the effects of radiation on hepatocyte senescence, laying foundations for the development of new therapies.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaofen Zhang
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Mengli Yu
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bingru Lin
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chaohui Yu
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
16
|
Kalekhan F, Kudva AK, Raghu SV, Rao S, Hegde SK, Simon P, Baliga MS. Traditionally Used Natural Products in Preventing Ionizing Radiation-Induced Dermatitis: First Review on the Clinical Studies. Anticancer Agents Med Chem 2021; 22:64-82. [PMID: 33820524 DOI: 10.2174/1871520621666210405093236] [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: 10/21/2020] [Revised: 12/16/2020] [Accepted: 01/15/2021] [Indexed: 11/22/2022]
Abstract
In the treatment of cancer, the use of ionizing radiation is an important modality. However, on the downside, radiation, when used for curative purposes, causes acute dermatitis or radiodermatitis at the site of radiation in most individuals. From a clinical viewpoint, severe dermatitis causes a burning and itching sensation is very painful, and severely affects the quality of life of the individual undergoing treatment. In worse situations, acute radiation dermatitis can cause gaps or breaks in the planned treatment and this can adversely affect the treatment objective and outcome. BACKGROUND In various traditional and folk systems of medicine, plants and plant products have been used since time immemorial for treating various skin ailments. Further, many cosmeceutical creams formulated based on knowledge from ethnomedicinal use are marketed and used to treat various ailments. In the current review, an attempt is made at summarizing the beneficial effects of some plants and plant products in mitigating acute radiation dermatitis in humans undergoing curative radiotherapy. Additionally, the emphasis is also placed on the mechanism/s responsible for the beneficial effects. OBJECTIVE The objective of this review is to summarize the clinical observations on the prevention of radiodermatitis by plant products. In this review, the protective effects of Adlay (Coix lachryma-jobi L.) bran extract, Aloe vera, Calendula officinalis, Cucumis sativus, green tea constituent the epigallocatechin-3-gallate, honey, Achillea millefolium, Matricaria chamomilla, olive oil and some polyherbal creams are addressed by also addressing on the mechanism of action for the beneficial effects. METHODS Two authors' data mined for information in Google Scholar, PubMed, Embase and the Cochrane Library for publications in the field from 1901 up to July 2020. The focus was on acute radiation dermatitis, ionizing radiation, curative radiotherapy, human cancer. The articles were collected and analyzed. RESULTS For the first time, this review addresses the usefulness of natural products like adlay bran, Aloe vera, Calendula officinalis, Cucumis sativus, green tea constituent the epigallocatechin-3-gallate, honey, Achillea millefolium, Matricaria chamomilla, olive oil and some experimentally constituted and commercially available polyherbal creams as skincare agents against the deleterious effects of ionizing radiation on the skin. The protective effects are possibly due to the free radical scavenging, antioxidant, anti-inflammatory, wound healing and skin protective effects. CONCLUSION The authors suggest that these plants have been used since antiquity as medicinal agents and require in-depth investigation with both clinical and preclinical validated models of study. The results of these studies will be extremely useful to cancer patients requiring curative radiotherapy, the dermatology fraternity, agro-based and pharmaceutical sectors at large.
Collapse
Affiliation(s)
- Faizan Kalekhan
- Research Unit, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka. India
| | - Avinash K Kudva
- Department of Biochemistry, Mangalore University, Mangalagangotri, Karnataka. India
| | - Shamprasad V Raghu
- Neurogenetics Laboratory, Department of Applied Zoology, Mangalore University, Mangalagangotri, Karnataka. India
| | - Suresh Rao
- Radiation Oncology, Mangalore Institute of Oncology, Mangalore, Karnataka. India
| | - Sanath K Hegde
- Radiation Oncology, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka. India
| | - Paul Simon
- Research Unit, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka. India
| | - Manjeshwar S Baliga
- Research Unit, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka. India
| |
Collapse
|
17
|
Liu Y, Miao L, Guo Y, Tian H. Preclinical Evaluation of Safety, Pharmacokinetics, Efficacy, and Mechanism of Radioprotective Agent HL-003. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6683836. [PMID: 33688393 PMCID: PMC7914087 DOI: 10.1155/2021/6683836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/01/2021] [Accepted: 02/11/2021] [Indexed: 11/18/2022]
Abstract
Amifostine is a radioprotector with high efficacy but poor safety, short half-life, no oral formulation, and poor compliance, which limits its application. With the increasing risk of exposure to radiation, the development of new radioprotective agents is critical. We previously synthesized a new amifostine derivative, the small molecule compound HL-003. In this study, we focused on evaluating the radioprotective properties of HL-003. Using the in vitro 2,2-diphenyl-1-picrylhydrazyl assay, we initially confirmed HL-003 as a strong antioxidant and demonstrated that its free radical scavenging activity was stronger than that of amifostine. Then, we performed an acute toxicity test, a 28-day toxicity test, a 30-day survival rate test, and a pharmacokinetic study, all of which provided aggregate evidence that HL-003 functioned as a small molecule radioprotector with high efficacy, a favorable safety profile, a long half-life, and oral administration. The intestinal radioprotective mechanism of HL-003 was explored in male C57 mice after abdominal irradiation by analyzing intestinal tissue samples with hematoxylin-eosin staining, immunohistochemistry, TUNEL staining, and immunofluorescence detection. The results showed that HL-003 protected intestinal DNA from radiation damage and suppressed the expression of phosphorylated histone H2AX, phosphorylated p53, and the apoptosis-related proteins caspase-8 and caspase-9, which contributed to maintaining the normal morphology of the small intestine and provided insights into the mechanism of radioprotection. Thus, HL-003 is a small molecule radioprotector with a potential application in radiation medicine.
Collapse
Affiliation(s)
- Yahong Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China
| | - Longfei Miao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China
| | - Yuying Guo
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin 300192, China
| |
Collapse
|
18
|
Radiation-Induced Normal Tissue Damage: Oxidative Stress and Epigenetic Mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3010342. [PMID: 31781332 PMCID: PMC6875293 DOI: 10.1155/2019/3010342] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 01/23/2023]
Abstract
Radiotherapy (RT) is currently one of the leading treatments for various cancers; however, it may cause damage to healthy tissue, with both short-term and long-term side effects. Severe radiation-induced normal tissue damage (RINTD) frequently has a significant influence on the progress of RT and the survival and prognosis of patients. The redox system has been shown to play an important role in the early and late effects of RINTD. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are the main sources of RINTD. The free radicals produced by irradiation can upregulate several enzymes including nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase), lipoxygenases (LOXs), nitric oxide synthase (NOS), and cyclooxygenases (COXs). These enzymes are expressed in distinct ways in various cells, tissues, and organs and participate in the RINTD process through different regulatory mechanisms. In recent years, several studies have demonstrated that epigenetic modulators play an important role in the RINTD process. Epigenetic modifications primarily contain noncoding RNA regulation, histone modifications, and DNA methylation. In this article, we will review the role of oxidative stress and epigenetic mechanisms in radiation damage, and explore possible prophylactic and therapeutic strategies for RINTD.
Collapse
|
19
|
The role of NLRP3 inflammasome activation in radiation damage. Biomed Pharmacother 2019; 118:109217. [DOI: 10.1016/j.biopha.2019.109217] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 07/02/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023] Open
|
20
|
Wang H, Wei J, Zheng Q, Meng L, Xin Y, Yin X, Jiang X. Radiation-induced heart disease: a review of classification, mechanism and prevention. Int J Biol Sci 2019; 15:2128-2138. [PMID: 31592122 PMCID: PMC6775290 DOI: 10.7150/ijbs.35460] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 06/19/2019] [Indexed: 12/13/2022] Open
Abstract
With the increasing incidence of thoracic tumors, radiation therapy (RT) has become an important component of comprehensive treatment. RT improves survival in many cancers, but it involves some inevitable complications. Radiation-induced heart disease (RIHD) is one of the most serious complications. RIHD comprises a spectrum of heart disease including cardiomyopathy, pericarditis, coronary artery disease, valvular heart disease and conduction system abnormalities. There are numerous clinical manifestations of RIHD, such as chest pain, palpitation, and dyspnea, even without obvious symptoms. Based on previous studies, the pathogenesis of RIHD is related to the production and effects of various cytokines caused by endothelial injury, inflammatory response, and oxidative stress (OS). Therefore, it is of great importance for clinicians to identify the mechanism and propose interventions for the prevention of RIHD.
Collapse
Affiliation(s)
- Heru Wang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, 130021, China.,Department of Cardiology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jinlong Wei
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Qingshuang Zheng
- Department of Cardiology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Lingbin Meng
- Department of Internal Medicine, Florida Hospital, Orlando, FL 32804,USA
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Xia Yin
- Department of Cardiology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, 130021, China
| |
Collapse
|
21
|
Wei J, Meng L, Hou X, Qu C, Wang B, Xin Y, Jiang X. Radiation-induced skin reactions: mechanism and treatment. Cancer Manag Res 2018; 11:167-177. [PMID: 30613164 PMCID: PMC6306060 DOI: 10.2147/cmar.s188655] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Radiotherapy (RT) is a major treatment for malignant tumors. The latest data show that >70% of patients with malignant tumors need RT at different periods. Skin changes can be experienced by up to 95% of patients who underwent RT. Inflammation and oxidative stress (OS) have been shown to be generally associated with radiation-induced skin reactions (RISRs). Inflammatory response and OS interact and promote each other during RISRs. Severe skin reactions often have a great impact on the progress of RT. The treatment of RISRs is particularly critical because advanced RT technology can also lead to skin reactions. RISRs are classified into acute and chronic reactions. The treatment methods for acute RISRs include steroid treatment, creams, ointments, and hydrocolloid dressings, depending on the reaction grading. Chronic RISRs includes chronic ulcerations, telangiectasias, and fibrosis of the skin, and advanced treatments such as mesenchymal stem cells, hyperbaric oxygen therapy, superoxide dismutase, and low-intensity laser therapy can be considered. Here, we review and summarize the important mechanisms that cause RISRs as well as the standard and advanced treatments for RISRs.
Collapse
Affiliation(s)
- Jinlong Wei
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China,
| | - Lingbin Meng
- Department of Internal Medicine, Florida Hospital, Orlando, FL 32803, USA
| | - Xue Hou
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China,
| | - Chao Qu
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China,
| | - Bin Wang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China,
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China,
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China,
| |
Collapse
|
22
|
Rationally designed mimics of antioxidant manganoenzymes: Role of structural features in the quest for catalysts with catalase and superoxide dismutase activity. Coord Chem Rev 2018. [DOI: 10.1016/j.ccr.2018.03.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
23
|
Kielmann M, Prior C, Senge MO. Porphyrins in troubled times: a spotlight on porphyrins and their metal complexes for explosives testing and CBRN defense. NEW J CHEM 2018. [DOI: 10.1039/c7nj04679k] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A critical perspective on (metallo)porphyrins in security-related applications: the past, present and future of explosives detection, CBRN defense, and beyond.
Collapse
Affiliation(s)
- Marc Kielmann
- School of Chemistry
- SFI Tetrapyrrole Laboratory
- Trinity Biomedical Sciences Institute
- Trinity College Dublin
- The University of Dublin
| | - Caroline Prior
- School of Chemistry
- SFI Tetrapyrrole Laboratory
- Trinity Biomedical Sciences Institute
- Trinity College Dublin
- The University of Dublin
| | - Mathias O. Senge
- Medicinal Chemistry
- Trinity Translational Medicine Institute
- Trinity Centre for Health Sciences
- Trinity College Dublin
- The University of Dublin
| |
Collapse
|
24
|
Nicco C, Batteux F. ROS Modulator Molecules with Therapeutic Potential in Cancers Treatments. Molecules 2017; 23:E84. [PMID: 29301225 PMCID: PMC6016996 DOI: 10.3390/molecules23010084] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 12/28/2017] [Accepted: 12/30/2017] [Indexed: 12/11/2022] Open
Abstract
Reactive Oxygen Species (ROS) are chemically reactive chemical species containing oxygen. The redox status of a cell is function of the relative concentrations of oxidized and reduced forms of proteins, enzymes, ROS, molecules containing thiol and other factors. In the organism, the redox balance is based on the generation and elimination of ROS produced by endogenous and exogenous sources. All living organisms must maintain their redox equilibrium to survive and proliferate. Enzymatic and molecular pathways control ROS levels tightly but differentially depending on the type of cell. This review is an overview of various molecules that modulate ROS production/detoxification and have a synergistic action with the chemotherapies to kill cancer cells while preserving normal cells to avoid anticancer drugs side effects, allowing a better therapeutic index of the anticancer treatments.
Collapse
Affiliation(s)
- Carole Nicco
- Department "Development, Reproduction and Cancer", Cochin Institute, INSERM U1016, University Paris Descartes, Paris 75014, France.
| | - Frédéric Batteux
- Department "Development, Reproduction and Cancer", Cochin Institute, INSERM U1016, University Paris Descartes, Paris 75014, France.
| |
Collapse
|
25
|
Bonetta R. Potential Therapeutic Applications of MnSODs and SOD-Mimetics. Chemistry 2017; 24:5032-5041. [DOI: 10.1002/chem.201704561] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Rosalin Bonetta
- Centre of Molecular Medicine and Biobanking; University of Malta; Msida MSD2080 Malta
| |
Collapse
|
26
|
Prévention médicale et traitement des complications pulmonaires secondaires à la radiothérapie. Cancer Radiother 2017; 21:411-423. [DOI: 10.1016/j.canrad.2017.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/13/2017] [Accepted: 03/24/2017] [Indexed: 12/12/2022]
|
27
|
Klein D, Steens J, Wiesemann A, Schulz F, Kaschani F, Röck K, Yamaguchi M, Wirsdörfer F, Kaiser M, Fischer JW, Stuschke M, Jendrossek V. Mesenchymal Stem Cell Therapy Protects Lungs from Radiation-Induced Endothelial Cell Loss by Restoring Superoxide Dismutase 1 Expression. Antioxid Redox Signal 2017; 26:563-582. [PMID: 27572073 PMCID: PMC5393411 DOI: 10.1089/ars.2016.6748] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
AIMS Radiation-induced normal tissue toxicity is closely linked to endothelial cell (EC) damage and dysfunction (acute effects). However, the underlying mechanisms of radiation-induced adverse late effects with respect to the vascular compartment remain elusive, and no causative radioprotective treatment is available to date. RESULTS The importance of injury to EC for radiation-induced late toxicity in lungs after whole thorax irradiation (WTI) was investigated using a mouse model of radiation-induced pneumopathy. We show that WTI induces EC loss as long-term complication, which is accompanied by the development of fibrosis. Adoptive transfer of mesenchymal stem cells (MSCs) either derived from bone marrow or aorta (vascular wall-resident MSCs) in the early phase after irradiation limited the radiation-induced EC loss and fibrosis progression. Furthermore, MSC-derived culture supernatants rescued the radiation-induced reduction in viability and long-term survival of cultured lung EC. We further identified the antioxidant enzyme superoxide dismutase 1 (SOD1) as a MSC-secreted factor. Importantly, MSC treatment restored the radiation-induced reduction of SOD1 levels after WTI. A similar protective effect was achieved by using the SOD-mimetic EUK134, suggesting that MSC-derived SOD1 is involved in the protective action of MSC, presumably through paracrine signaling. INNOVATION In this study, we explored the therapeutic potential of MSC therapy to prevent radiation-induced EC loss (late effect) and identified the protective mechanisms of MSC action. CONCLUSIONS Adoptive transfer of MSCs early after irradiation counteracts radiation-induced vascular damage and EC loss as late adverse effects. The high activity of vascular wall-derived MSCs for radioprotection may be due to their tissue-specific action. Antioxid. Redox Signal. 26, 563-582.
Collapse
Affiliation(s)
- Diana Klein
- 1 Institute of Cell Biology (Cancer Research), University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Jennifer Steens
- 1 Institute of Cell Biology (Cancer Research), University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Alina Wiesemann
- 1 Institute of Cell Biology (Cancer Research), University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Florian Schulz
- 2 Department of Chemical Biology, Faculty of Biology, Center for Medical Biotechnology, University of Duisburg-Essen , Essen, Germany
| | - Farnusch Kaschani
- 2 Department of Chemical Biology, Faculty of Biology, Center for Medical Biotechnology, University of Duisburg-Essen , Essen, Germany
| | - Katharina Röck
- 3 Institute for Pharmacology, University Hospital, Heinrich-Heine-University , Düsseldorf, Germany
| | | | - Florian Wirsdörfer
- 1 Institute of Cell Biology (Cancer Research), University Hospital, University of Duisburg-Essen , Essen, Germany
| | - Markus Kaiser
- 2 Department of Chemical Biology, Faculty of Biology, Center for Medical Biotechnology, University of Duisburg-Essen , Essen, Germany
| | - Jens W Fischer
- 3 Institute for Pharmacology, University Hospital, Heinrich-Heine-University , Düsseldorf, Germany
| | - Martin Stuschke
- 5 Department of Radiotherapy, University of Duisburg-Essen, University Hospital , Essen, Germany
| | - Verena Jendrossek
- 1 Institute of Cell Biology (Cancer Research), University Hospital, University of Duisburg-Essen , Essen, Germany
| |
Collapse
|
28
|
Mathieu E, Bernard AS, Delsuc N, Quévrain E, Gazzah G, Lai B, Chain F, Langella P, Bachelet M, Masliah J, Seksik P, Policar C. A Cell-Penetrant Manganese Superoxide Dismutase (MnSOD) Mimic Is Able To Complement MnSOD and Exerts an Antiinflammatory Effect on Cellular and Animal Models of Inflammatory Bowel Diseases. Inorg Chem 2017; 56:2545-2555. [PMID: 28198622 DOI: 10.1021/acs.inorgchem.6b02695] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Inorganic complexes are increasingly used for biological and medicinal applications, and the question of the cell penetration and distribution of metallodrugs is key to understanding their biological activity. Oxidative stress is known to be involved in inflammation and in inflammatory bowel diseases for which antioxidative defenses are weakened. We report here the study of the manganese complex Mn1 mimicking superoxide dismutase (SOD), a protein involved in cell protection against oxidative stress, using an approach in inorganic cellular chemistry combining the investigation of Mn1 intracellular speciation using mass spectrometry and of its quantification and distribution using electron paramagnetic resonance and spatially resolved X-ray fluorescence with evaluation of its biological activity. More precisely, we have looked for and found the MS signature of Mn1 in cell lysates and quantified the overall manganese content. Intestinal epithelial cells activated by bacterial lipopolysaccharide were taken as a cellular model of oxidative stress and inflammation. DNBS-induced colitis in mice was used to investigate Mn1 activity in vivo. Mn1 exerts an intracellular antiinflammatory activity, remains at least partially coordinated, with diffuse distribution over the whole cell, and functionally complements mitochondrial MnSOD.
Collapse
Affiliation(s)
- Emilie Mathieu
- Département de Chimie, Ecole Normale Superieure, PSL Research University, UPMC Univ Paris 06, CNRS, Laboratoire des Biomolecules (LBM), 24 rue Lhomond, 75005 Paris, France
| | - Anne-Sophie Bernard
- Département de Chimie, Ecole Normale Superieure, PSL Research University, UPMC Univ Paris 06, CNRS, Laboratoire des Biomolecules (LBM), 24 rue Lhomond, 75005 Paris, France
| | - Nicolas Delsuc
- Département de Chimie, Ecole Normale Superieure, PSL Research University, UPMC Univ Paris 06, CNRS, Laboratoire des Biomolecules (LBM), 24 rue Lhomond, 75005 Paris, France
| | - Elodie Quévrain
- Département de Chimie, Ecole Normale Superieure, PSL Research University, UPMC Univ Paris 06, CNRS, Laboratoire des Biomolecules (LBM), 24 rue Lhomond, 75005 Paris, France
| | - Géraldine Gazzah
- Département de Chimie, Ecole Normale Superieure, PSL Research University, UPMC Univ Paris 06, CNRS, Laboratoire des Biomolecules (LBM), 24 rue Lhomond, 75005 Paris, France
| | - Barry Lai
- X-ray Science Division, Argonne National Laboratory (ANL) , Argonne, Illinois 60439, United States
| | - Florian Chain
- Commensal and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France
| | - Philippe Langella
- Commensal and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France
| | - Maria Bachelet
- Sorbonne Universites, UPMC Univ Paris 06 - Département de Chimie, Ecole Normale Superieure, PSL Research University - CNRS, INSERM, APHP, INRA, Laboratoire des Biomolecules (LBM), 27 rue de Chaligny, 75012 Paris, France.,Commensal and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France
| | - Joelle Masliah
- Département de Chimie, Ecole Normale Superieure, PSL Research University, UPMC Univ Paris 06, CNRS, Laboratoire des Biomolecules (LBM), 24 rue Lhomond, 75005 Paris, France.,Sorbonne Universites, UPMC Univ Paris 06 - Département de Chimie, Ecole Normale Superieure, PSL Research University - CNRS, INSERM, APHP, INRA, Laboratoire des Biomolecules (LBM), 27 rue de Chaligny, 75012 Paris, France
| | - Philippe Seksik
- Sorbonne Universites, UPMC Univ Paris 06 - Département de Chimie, Ecole Normale Superieure, PSL Research University - CNRS, INSERM, APHP, INRA, Laboratoire des Biomolecules (LBM), 27 rue de Chaligny, 75012 Paris, France.,Commensal and Probiotics-Host Interactions Laboratory, Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, F-78350 Jouy-en-Josas, France
| | - Clotilde Policar
- Département de Chimie, Ecole Normale Superieure, PSL Research University, UPMC Univ Paris 06, CNRS, Laboratoire des Biomolecules (LBM), 24 rue Lhomond, 75005 Paris, France
| |
Collapse
|
29
|
Zhang X, Hadley C, Jackson IL, Zhang Y, Zhang A, Spasojevic I, Haberle IB, Vujaskovic Z. Hypo-CpG methylation controls PTEN expression and cell apoptosis in irradiated lung. Free Radic Res 2016; 50:875-86. [PMID: 27367846 DOI: 10.1080/10715762.2016.1189078] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE The current study was designed to address our hypothesis that oxidative stress secondary to the ionizing event upregulates phosphatase and tensin homolog (PTEN) mRNA and protein in the lungs of C57BL/6J mice through oxidative DNA damage resulting in CpG hypomethylation in the PTEN promoter. METHODS Fibrosis-prone C57BL/6J mice were exposed to 0 or 15 Gy of 320 kVp X-rays to the whole thorax. Lung tissue was serially harvested at time points between one day and six months postirradiation. Tissue levels of PTEN mRNA, total protein, and phosphorylated PTEN, as well as CpG methylation of the PTEN promoter, expression of DNA methyltransferases 1 (Dnmt1) and 3a (Dnmt3a), NADPH oxidase 4 (Nox4) protein expression, and DNA damage levels were measured. The induction of DNA damage and global methylation changes were also examined in hydrogen peroxide (H2O2)-treated human umbilical vein endothelial cells (HUVECs) and human bronchial epithelial cells in vitro. RESULTS These experiments demonstrate that PTEN mRNA and protein, Nox4 protein, and DNA damage levels increase continuously from one day to six months following radiation exposure. Elevated PTEN transcription and translation are likely the result of the observed decrease in CpG methylation of the PTEN promoter region. This finding is not consistent with the observed increase in Dnmt1 and Dnmt3a protein expression, implicating an alternative mechanism as the driving force behind hypomethylation. In vitro results provide evidence that H2O2 can induce DNA damage and affect DNA methylation status. The Mn porphyrin-based superoxide dismutase (SOD) mimic MnTnHEx-2-PyP(5+ )exhibited partial rescue from radiation-induced hypomethylation. CONCLUSIONS Taken together, these data suggest that reactive oxygen species (ROS)-induced DNA damage results in hypomethylation of the PTEN promoter, upregulation of PTEN mRNA and protein, and a subsequent increase in apoptosis in irradiated lung tissue.
Collapse
Affiliation(s)
- Xiuwu Zhang
- a Division of Translational Radiation Sciences, Department of Radiation Oncology , University of Maryland, School of Medicine , Baltimore , MD , USA
| | | | - Isabel L Jackson
- a Division of Translational Radiation Sciences, Department of Radiation Oncology , University of Maryland, School of Medicine , Baltimore , MD , USA
| | - Yi Zhang
- a Division of Translational Radiation Sciences, Department of Radiation Oncology , University of Maryland, School of Medicine , Baltimore , MD , USA
| | - Angel Zhang
- a Division of Translational Radiation Sciences, Department of Radiation Oncology , University of Maryland, School of Medicine , Baltimore , MD , USA
| | - Ivan Spasojevic
- c Department of Radiation Oncology , Duke University Medical Center , Durham , NC , USA
| | - Ines Batinic Haberle
- a Division of Translational Radiation Sciences, Department of Radiation Oncology , University of Maryland, School of Medicine , Baltimore , MD , USA
| | - Zeljko Vujaskovic
- a Division of Translational Radiation Sciences, Department of Radiation Oncology , University of Maryland, School of Medicine , Baltimore , MD , USA ;,c Department of Radiation Oncology , Duke University Medical Center , Durham , NC , USA
| |
Collapse
|
30
|
Mishra OP, Popov AV, Pietrofesa RA, Christofidou-Solomidou M. Gamma-irradiation produces active chlorine species (ACS) in physiological solutions: Secoisolariciresinol diglucoside (SDG) scavenges ACS - A novel mechanism of DNA radioprotection. Biochim Biophys Acta Gen Subj 2016; 1860:1884-97. [PMID: 27261092 PMCID: PMC5253237 DOI: 10.1016/j.bbagen.2016.05.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 05/13/2016] [Accepted: 05/26/2016] [Indexed: 11/28/2022]
Abstract
Background Secoisolariciresinol diglucoside (SDG), the main lignan in whole grain flaxseed, is a potent antioxidant and free radical scavenger with known radioprotective properties. However, the exact mechanism of SDG radioprotection is not well understood. The current study identified a novel mechanism of DNA radioprotection by SDG in physiological solutions by scavenging active chlorine species (ACS) and reducing chlorinated nucleobases. Methods The ACS scavenging activity of SDG was determined using two highly specific fluoroprobes: hypochlorite-specific 3′-(p-aminophenyl) fluorescein (APF) and hydroxyl radical-sensitive 3′-(p-hydroxyphenyl) fluorescein (HPF). Dopamine, an SDG structural analog, was used for proton 1H NMR studies to trap primary ACS radicals. Taurine N-chlorination was determined to demonstrate radiation-induced generation of hypochlorite, a secondary ACS. DNA protection was assessed by determining the extent of DNA fragmentation and plasmid DNA relaxation following exposure to ClO− and radiation. Purine base chlorination by ClO− and γ-radiation was determined by using 2-aminopurine (2-AP), a fluorescent analog of 6-aminopurine. Results: Chloride anions (Cl−) consumed >90% of hydroxyl radicals in physiological solutions produced by γ-radiation resulting in ACS formation, which was detected by 1H NMR. Importantly, SDG scavenged hypochlorite- and γ-radiation-induced ACS. In addition, SDG blunted ACS-induced fragmentation of calf thymus DNA and plasmid DNA relaxation. SDG treatment before or after ACS exposure decreased the ClO− or γ-radiation-induced chlorination of 2-AP. Exposure to γ-radiation resulted in increased taurine chlorination, indicative of ClO− generation. NMR studies revealed formation of primary ACS radicals (chlorine atoms (Cl•) and dichloro radical anions (Cl2−•)), which were trapped by SDG and its structural analog dopamine. Conclusion We demonstrate that γ-radiation induces the generation of ACS in physiological solutions. SDG treatment scavenged ACS and prevented ACS-induced DNA damage and chlorination of 2-aminopurine. This study identified a novel and unique mechanism of SDG radioprotection, through ACS scavenging, and supports the potential usefulness of SDG as a radioprotector and mitigator for radiation exposure as part of cancer therapy or accidental exposure.
Collapse
Affiliation(s)
- Om P Mishra
- Department of Medicine, Pulmonary, Allergy and Critical Care Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States.
| | - Anatoliy V Popov
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States.
| | - Ralph A Pietrofesa
- Department of Medicine, Pulmonary, Allergy and Critical Care Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States.
| | - Melpo Christofidou-Solomidou
- Department of Medicine, Pulmonary, Allergy and Critical Care Division, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, United States.
| |
Collapse
|
31
|
Abstract
In the event of a nuclear disaster, the individuals proximal to the source of radiation will be exposed to combined radiation injury. As irradiation delays cutaneous repair, the purpose of this study was to elucidate the effect of combined radiation and burn injury (CRBI) on apoptosis and inflammation at the site of skin injury. Male C57Bl/6 mice were exposed to no injury, thermal injury only, radiation only (1 and 6 Gy) and CRBI (1 and 6 Gy) and euthanized at various times after for skin collection. TUNEL staining revealed that the CRBI 6 Gy group had a delayed and increased apoptotic response. This correlated with decreased recovery of live cells as compared to the other injuries. Similar response was observed when cleaved-caspase-3 immunohistochemical staining was compared between CRBI 6 Gy and thermal injury. TNFR1, caspase 8, Bax and IL-6 mRNA expression revealed that the higher CRBI group had delayed increase in mRNA expression as compared to thermal injury alone. RIPK1 mRNA expression and necrotic cell counts were delayed in the CRBI 6 Gy group to day 5. TNF-α and NFκB expression peaked in the CRBI 6 Gy group at day 1 and was much higher than the other injuries. Also, inflammatory cell counts in the CRBI 6 Gy group were lower at early time points as compared to thermal injury by itself. These data suggest that CRBI delays and exacerbates apoptosis and inflammation in skin as well as increases necrosis thus resulting in delayed wound healing.
Collapse
|
32
|
Singh V, Gupta D, Arora R. NF-kB as a key player in regulation of cellular radiation responses and identification of radiation countermeasures. Discoveries (Craiova) 2015; 3:e35. [PMID: 32309561 PMCID: PMC7159829 DOI: 10.15190/d.2015.27] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Nuclear factor (NF)-κB is a transcription factor that plays significant role in immunity, cellular survival and inhibition of apoptosis, through the induction of genetic networks. Depending on the stimulus and the cell type, the members of NF-κB related family (RelA, c-Rel, RelB, p50, and p52), forms different combinations of homo and hetero-dimers. The activated complexes (Es) translocate into the nucleus and bind to the 10bp κB site of promoter region of target genes in stimulus specific manner. In response to radiation, NF-κB is known to reduce cell death by promoting the expression of anti-apoptotic proteins and activation of cellular antioxidant defense system. Constitutive activation of NF-κB associated genes in tumour cells are known to enhance radiation resistance, whereas deletion in mice results in hypersensitivity to IR-induced GI damage. NF-κB is also known to regulate the production of a wide variety of cytokines and chemokines, which contribute in enhancing cell proliferation and tissue regeneration in various organs, such as the GI crypts stem cells, bone marrow etc., following exposure to IR. Several other cytokines are also known to exert potent pro-inflammatory effects that may contribute to the increase of tissue damage following exposure to ionizing radiation. Till date there are a series of molecules or group of compounds that have been evaluated for their radio-protective potential, and very few have reached clinical trials. The failure or less success of identified agents in humans could be due to their reduced radiation protection efficacy.
In this review we have considered activation of NF-κB as a potential marker in screening of radiation countermeasure agents (RCAs) and cellular radiation responses. Moreover, we have also focused on associated mechanisms of activation of NF-κB signaling and their specified family member activation with respect to stimuli. Furthermore, we have categorized their regulated gene expressions and their function in radiation response or modulation. In addition, we have discussed some recently developed radiation countermeasures in relation to NF-κB activation
Collapse
Affiliation(s)
- Vijay Singh
- Division of Radiation Biosciences, Institute of Nuclear Medicine & Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi, India
| | - Damodar Gupta
- Division of Radiation Biosciences, Institute of Nuclear Medicine & Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi, India
| | - Rajesh Arora
- Division of Radiation Biosciences, Institute of Nuclear Medicine & Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi, India
| |
Collapse
|
33
|
Moulder JE. 2013 Dade W. Moeller lecture: medical countermeasures against radiological terrorism. HEALTH PHYSICS 2014; 107:164-71. [PMID: 24978287 PMCID: PMC4076685 DOI: 10.1097/hp.0000000000000082] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Soon after the 9-11 attacks, politicians and scientists began to question our ability to cope with a large-scale radiological terrorism incident. The outline of what was needed was fairly obvious: the ability to prevent such an attack, methods to cope with the medical consequences, the ability to clean up afterward, and the tools to figure out who perpetrated the attack and bring them to justice. The medical response needed three components: the technology to determine rapidly the radiation doses received by a large number of people, methods for alleviating acute hematological radiation injuries, and therapies for mitigation and treatment of chronic radiation injuries. Research done to date has shown that a realistic medical response plan is scientifically possible, but the regulatory and financial barriers to achieving this may currently be insurmountable.
Collapse
Affiliation(s)
- John E. Moulder
- Center for Medical Countermeasures Against Radiological Terrorism, Radiation Oncology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226 U. S. A
| |
Collapse
|
34
|
Batinic-Haberle I, Tovmasyan A, Roberts ERH, Vujaskovic Z, Leong KW, Spasojevic I. SOD therapeutics: latest insights into their structure-activity relationships and impact on the cellular redox-based signaling pathways. Antioxid Redox Signal 2014; 20:2372-415. [PMID: 23875805 PMCID: PMC4005498 DOI: 10.1089/ars.2012.5147] [Citation(s) in RCA: 169] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 06/30/2013] [Accepted: 07/22/2013] [Indexed: 01/23/2023]
Abstract
SIGNIFICANCE Superoxide dismutase (SOD) enzymes are indispensable and ubiquitous antioxidant defenses maintaining the steady-state levels of O2·(-); no wonder, thus, that their mimics are remarkably efficacious in essentially any animal model of oxidative stress injuries thus far explored. RECENT ADVANCES Structure-activity relationship (half-wave reduction potential [E1/2] versus log kcat), originally reported for Mn porphyrins (MnPs), is valid for any other class of SOD mimics, as it is dominated by the superoxide reduction and oxidation potential. The biocompatible E1/2 of ∼+300 mV versus normal hydrogen electrode (NHE) allows powerful SOD mimics as mild oxidants and antioxidants (alike O2·(-)) to readily traffic electrons among reactive species and signaling proteins, serving as fine mediators of redox-based signaling pathways. Based on similar thermodynamics, both SOD enzymes and their mimics undergo similar reactions, however, due to vastly different sterics, with different rate constants. CRITICAL ISSUES Although log kcat(O2·(-)) is a good measure of therapeutic potential of SOD mimics, discussions of their in vivo mechanisms of actions remain mostly of speculative character. Most recently, the therapeutic and mechanistic relevance of oxidation of ascorbate and glutathionylation and oxidation of protein thiols by MnP-based SOD mimics and subsequent inactivation of nuclear factor κB has been substantiated in rescuing normal and killing cancer cells. Interaction of MnPs with thiols seems to be, at least in part, involved in up-regulation of endogenous antioxidative defenses, leading to the healing of diseased cells. FUTURE DIRECTIONS Mechanistic explorations of single and combined therapeutic strategies, along with studies of bioavailability and translational aspects, will comprise future work in optimizing redox-active drugs.
Collapse
Affiliation(s)
- Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University Medical School, Durham, North Carolina
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University Medical School, Durham, North Carolina
| | - Emily R. H. Roberts
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, Duke University Medical School, Durham, North Carolina
| | - Kam W. Leong
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
- King Abdulaziz University, Jeddah, Saudi Arabia Kingdom
| | - Ivan Spasojevic
- Department of Medicine, Duke University Medical School, Durham, North Carolina
| |
Collapse
|
35
|
Radioprotection of normal tissue cells. Strahlenther Onkol 2014; 190:745-52. [DOI: 10.1007/s00066-014-0637-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 02/05/2014] [Indexed: 12/13/2022]
|
36
|
Zhao X, Yang H, Jiang G, Ni M, Deng Y, Cai J, Li Z, Shen F, Tao X. Simvastatin attenuates radiation-induced tissue damage in mice. JOURNAL OF RADIATION RESEARCH 2014; 55:257-64. [PMID: 24105712 PMCID: PMC3951077 DOI: 10.1093/jrr/rrt115] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 08/17/2013] [Accepted: 08/21/2013] [Indexed: 05/18/2023]
Abstract
The aim of this study was to investigate the protective effect of simvastatin against radiation-induced tissue injury in mice. Mice were radiated with 4 Gy or 8 Gy after 20 mg/kg/d simvastatin treatment over 2 weeks. Morphological changes were observed in the jejunum and bone marrow, and apoptotic cells were determined in both tissues. Peripheral blood cells were counted, and the superoxide dismutase (SOD) activity and the malondialdehyde (MDA) level in tissues of both thymus and spleen were measured. Compared with the radiation-only group, 20 mg/kg/d simvastatin administration significantly increased the mean villi height and decreased apoptotic cells in jejunum tissue, and stimulated regeneration and reduced apoptotic cells in bone marrow. Peripheral blood cell analysis revealed that simvastatin treatment induced a larger number of red blood cells and increased the hemoglobin level present after 4 Gy of radiation. Interestingly, it was also found that the number of peripheral endothelial progenitor cells was markedly increased following simvastatin administration. Antioxidant determination for tissues displayed that simvastatin therapy increased the SOD activity after both 4 and 8 Gy of radiation, but only decreased the MDA level after 4 Gy. Simvastatin ameliorated radiation-induced tissue damage in mice. The radioprotective effect of simvastatin was possibly related to inhibition of apoptosis and improvement of oxygen-carrying and antioxidant activities.
Collapse
Affiliation(s)
- Xinbin Zhao
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, 415 Feng-Yang Road, Shanghai 200003, China
| | - Hong Yang
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, 415 Feng-Yang Road, Shanghai 200003, China
| | - Guojun Jiang
- Department of Pharmacy, Xiaoshan Hospital, 728 Yu-Cai-Bei Road, Hangzhou, Zhejiang 311202, China
| | - Min Ni
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, 415 Feng-Yang Road, Shanghai 200003, China
| | - Yaping Deng
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, 325 Guo-He Road, Shanghai 200433, China
| | - Jian Cai
- Department of Pharmacy, Xiaoshan Hospital, 728 Yu-Cai-Bei Road, Hangzhou, Zhejiang 311202, China
| | - Zhangpeng Li
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, 325 Guo-He Road, Shanghai 200433, China
| | - Fuming Shen
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, 325 Guo-He Road, Shanghai 200433, China
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University, 301 Yan-Chang-Zhong Road, Shanghai 200072, China
| | - Xia Tao
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, 415 Feng-Yang Road, Shanghai 200003, China
- Corresponding author. Tel: +86-21-8188-6182; Fax: +86-21-6549-3951;
| |
Collapse
|
37
|
Kash JC, Xiao Y, Davis AS, Walters KA, Chertow DS, Easterbrook JD, Dunfee RL, Sandouk A, Jagger BW, Schwartzman LM, Kuestner RE, Wehr NB, Huffman K, Rosenthal RA, Ozinsky A, Levine RL, Doctrow SR, Taubenberger JK. Treatment with the reactive oxygen species scavenger EUK-207 reduces lung damage and increases survival during 1918 influenza virus infection in mice. Free Radic Biol Med 2014; 67:235-47. [PMID: 24140866 PMCID: PMC3927540 DOI: 10.1016/j.freeradbiomed.2013.10.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 10/07/2013] [Accepted: 10/08/2013] [Indexed: 12/16/2022]
Abstract
The 1918 influenza pandemic caused over 40 million deaths worldwide, with 675,000 deaths in the United States alone. Studies in several experimental animal models showed that 1918 influenza virus infection resulted in severe lung pathology associated with dysregulated immune and cell death responses. To determine if reactive oxygen species produced by host inflammatory responses play a central role in promoting severity of lung pathology, we treated 1918 influenza virus-infected mice with the catalytic catalase/superoxide dismutase mimetic, salen-manganese complex EUK-207 beginning 3 days postinfection. Postexposure treatment of mice infected with a lethal dose of the 1918 influenza virus with EUK-207 resulted in significantly increased survival and reduced lung pathology without a reduction in viral titers. In vitro studies also showed that EUK-207 treatment did not affect 1918 influenza viral replication. Immunohistochemical analysis showed a reduction in the detection of the apoptosis marker cleaved caspase-3 and the oxidative stress marker 8-oxo-2'-deoxyguanosine in lungs of EUK-207-treated animals compared to vehicle controls. High-throughput sequencing and RNA expression microarray analysis revealed that treatment resulted in decreased expression of inflammatory response genes and increased lung metabolic and repair responses. These results directly demonstrate that 1918 influenza virus infection leads to an immunopathogenic immune response with excessive inflammatory and cell death responses that can be limited by treatment with the catalytic antioxidant EUK-207.
Collapse
Affiliation(s)
- John C Kash
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Yongli Xiao
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - A Sally Davis
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Daniel S Chertow
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Judith D Easterbrook
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rebecca L Dunfee
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aline Sandouk
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brett W Jagger
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Louis M Schwartzman
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Nancy B Wehr
- Laboratory of Biochemistry, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Karl Huffman
- Pulmonary Center, Department of Medicine, Boston University Medical School, Boston, MA 02118, USA
| | - Rosalind A Rosenthal
- Pulmonary Center, Department of Medicine, Boston University Medical School, Boston, MA 02118, USA
| | | | - Rodney L Levine
- Laboratory of Biochemistry, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Susan R Doctrow
- Pulmonary Center, Department of Medicine, Boston University Medical School, Boston, MA 02118, USA
| | - Jeffery K Taubenberger
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
38
|
Lee YW, Cho HJ, Lee WH, Sonntag WE. Whole brain radiation-induced cognitive impairment: pathophysiological mechanisms and therapeutic targets. Biomol Ther (Seoul) 2013; 20:357-70. [PMID: 24009822 PMCID: PMC3762274 DOI: 10.4062/biomolther.2012.20.4.357] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 07/04/2012] [Indexed: 12/19/2022] Open
Abstract
Radiation therapy, the most commonly used for the treatment of brain tumors, has been shown to be of major significance in tu-mor control and survival rate of brain tumor patients. About 200,000 patients with brain tumor are treated with either partial large field or whole brain radiation every year in the United States. The use of radiation therapy for treatment of brain tumors, however, may lead to devastating functional deficits in brain several months to years after treatment. In particular, whole brain radiation therapy results in a significant reduction in learning and memory in brain tumor patients as long-term consequences of treatment. Although a number of in vitro and in vivo studies have demonstrated the pathogenesis of radiation-mediated brain injury, the cel-lular and molecular mechanisms by which radiation induces damage to normal tissue in brain remain largely unknown. Therefore, this review focuses on the pathophysiological mechanisms of whole brain radiation-induced cognitive impairment and the iden-tification of novel therapeutic targets. Specifically, we review the current knowledge about the effects of whole brain radiation on pro-oxidative and pro-inflammatory pathways, matrix metalloproteinases (MMPs)/tissue inhibitors of metalloproteinases (TIMPs) system and extracellular matrix (ECM), and physiological angiogenesis in brain. These studies may provide a foundation for defin-ing a new cellular and molecular basis related to the etiology of cognitive impairment that occurs among patients in response to whole brain radiation therapy. It may also lead to new opportunities for therapeutic interventions for brain tumor patients who are undergoing whole brain radiation therapy.
Collapse
Affiliation(s)
- Yong Woo Lee
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA 24061, USA ; School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | | | | | | |
Collapse
|
39
|
Mahmood J, Jelveh S, Zaidi A, Doctrow SR, Hill RP. Mitigation of radiation-induced lung injury with EUK-207 and genistein: effects in adolescent rats. Radiat Res 2012; 179:125-34. [PMID: 23237541 DOI: 10.1667/rr2954.1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Exposure of civilian populations to radiation due to accident, war or terrorist act is an increasing concern. The lung is one of the more radiosensitive organs that may be affected in people receiving partial-body irradiation and radiation injury in lung is thought to be associated with the development of a prolonged inflammatory response. Here we examined how effectively damage to the lung can be mitigated by administration of drugs initiated at different times after radiation exposure and examined response in adolescent animals for comparison with the young adult animals that we had studied previously. We studied the mitigation efficacy of the isoflavone genistein (50 mg/kg) and the salen-Mn superoxide dismutase-catalase mimetic EUK-207 (8 mg/kg), both of which have been reported to scavenge reactive oxygen species and reduce activity of the NFkB pathway. The drugs were given by subcutaneous injection to 6- to 7-week-old Fisher rats daily starting either immediately or 2 weeks after irradiation with 12 Gy to the whole thorax. The treatment was stopped at 28 weeks post irradiation and the animals were assessed for levels of inflammatory cytokines, activated macrophages, oxidative damage and fibrosis at 48 weeks post irradiation. We demonstrated that both genistein and EUK-207 delayed and suppressed the increased breathing rate associated with pneumonitis. These agents also reduced levels of oxidative damage (50-100%), levels of TGF-β1 expression (75-100%), activated macrophages (20-60%) and fibrosis (60-80%). The adolescent rats developed pneumonitis earlier following irradiation of the lung than did the adult rats leading to greater severe morbidity requiring euthanasia (∼37% in adolescents vs. ∼10% in young adults) but the extent of the mitigation of the damage was similar or slightly greater.
Collapse
Affiliation(s)
- J Mahmood
- Ontario Cancer Institute/Princess Margaret Cancer Center, University Health Network, and The Campbell Family Institute for Cancer Research, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
40
|
Otterson MF, Nie L, Schmidt JL, Link BJ, Jovanovic N, Lyros O, Rafiee P. EUK-207 protects human intestinal microvascular endothelial cells (HIMEC) against irradiation-induced apoptosis through the Bcl2 pathway. Life Sci 2012; 91:771-82. [PMID: 22940617 PMCID: PMC3520131 DOI: 10.1016/j.lfs.2012.08.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 08/04/2012] [Accepted: 08/08/2012] [Indexed: 01/16/2023]
Abstract
AIM To elucidate the signaling mechanisms involved in the protective effect of EUK-207 against irradiation-induced cellular damage and apoptosis in human intestinal microvasculature endothelial cells (HIMEC). METHODS HIMECs were irradiated and treated with EUK-207. Using hydroethidine and DCF-DA fluorescent probe the intracellular superoxide and reactive oxygen species (ROS) were determined. By real-time PCR and western blotting caspase-3, Bcl2 and Bax genes and proteins were analyzed. Proliferation was determined by [(3)H]-thymidine uptake. Immunofluorescence staining was used for translocation of p65 NFκB subunit. KEY FINDING Irradiation increased ROS production, apoptosis, Bax, Caspase3 and NFkB activity in HIMEC and inhibited cell survival/growth/proliferation. EUK-207 restored the endothelial functions, markedly inhibited the ROS, up-regulated the Bcl2 and down-regulated Bax and prevented NFκB caspase 3 activity in HIMEC. SIGNIFICANCE HIMEC provide a novel model to define the effect of irradiation induced endothelial dysfunction. Our findings suggest that EUK-207 effectively inhibits the damaging effect of irradiation.
Collapse
Affiliation(s)
- Mary F. Otterson
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Linghui Nie
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jamie L. Schmidt
- Division of Gastroenterology and Hepathology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Benjamin J. Link
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Nebojsa Jovanovic
- Division of Gastroenterology and Hepathology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Orestis Lyros
- Division of Gastroenterology and Hepathology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Parvaneh Rafiee
- Department of Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin
- Division of Gastroenterology and Hepathology, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
41
|
Tovmasyan A, Sheng H, Weitner T, Arulpragasam A, Lu M, Warner DS, Vujaskovic Z, Spasojevic I, Batinic-Haberle I. Design, mechanism of action, bioavailability and therapeutic effects of mn porphyrin-based redox modulators. Med Princ Pract 2012; 22:103-30. [PMID: 23075911 PMCID: PMC3640855 DOI: 10.1159/000341715] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 07/01/2012] [Indexed: 12/18/2022] Open
Abstract
Based on aqueous redox chemistry and simple in vivo models of oxidative stress, Escherichia coli and Saccharomyces cerevisiae, the cationic Mn(III) N-substituted pyridylporphyrins (MnPs) have been identified as the most potent cellular redox modulators within the porphyrin class of drugs; their efficacy in animal models of diseases that have oxidative stress in common is based on their high ability to catalytically remove superoxide, peroxynitrite, carbonate anion radical, hypochlorite, nitric oxide, lipid peroxyl and alkoxyl radicals, thus suppressing the primary oxidative event. While doing so MnPs could couple with cellular reductants and redox-active proteins. Reactive species are widely accepted as regulators of cellular transcriptional activity: minute, nanomolar levels are essential for normal cell function, while submicromolar or micromolar levels impose oxidative stress, which is evidenced in increased inflammatory and immune responses. By removing reactive species, MnPs affect redox-based cellular transcriptional activity and consequently secondary oxidative stress, and in turn inflammatory processes. The equal ability to reduce and oxidize superoxide during the dismutation process and recently accumulated results suggest that pro-oxidative actions of MnPs may also contribute to their therapeutic effects. All our data identify the superoxide dismutase-like activity, estimated by log k(cat)O2-*), as a good measure for the therapeutic efficacy of MnPs. Their accumulation in mitochondria and their ability to cross the blood-brain barrier contribute to their remarkable efficacy. We summarize herein the therapeutic effects of MnPs in cancer, central nervous system injuries, diabetes, their radioprotective action and potential for imaging. Few of the most potent modulators of cellular redox-based pathways, MnTE2-PyP5+, MnTDE-2-ImP5+, MnTnHex-2-PyP5+ and MnTnBuOE-2-PyP5+, are under preclinical and clinical development.
Collapse
Affiliation(s)
- Artak Tovmasyan
- Department of Radiation Oncology, Duke University Medical
Center, Durham, N.C., USA
| | - Huaxin Sheng
- Department of Anesthesiology, Duke University Medical Center,
Durham, N.C., USA
- Department of Multidisciplinary Neuroprotection Laboratories,
Duke University Medical Center, Durham, N.C., USA
| | - Tin Weitner
- Department of Radiation Oncology, Duke University Medical
Center, Durham, N.C., USA
| | - Amanda Arulpragasam
- Department of Duke University Neuroscience Undergraduate
Program, Duke University Medical Center, Durham, N.C., USA
| | - Miaomiao Lu
- Department of Anesthesiology, Duke University Medical Center,
Durham, N.C., USA
- Department of Multidisciplinary Neuroprotection Laboratories,
Duke University Medical Center, Durham, N.C., USA
- Department of Department of Anesthesiology, Second Affiliated
Hospital, Zhengzhou University, Zhengzhou, China
| | - David S. Warner
- Department of Anesthesiology, Duke University Medical Center,
Durham, N.C., USA
- Department of Multidisciplinary Neuroprotection Laboratories,
Duke University Medical Center, Durham, N.C., USA
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, Duke University Medical
Center, Durham, N.C., USA
| | - Ivan Spasojevic
- Department of Medicine, Duke University Medical Center, Durham,
N.C., USA
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University Medical
Center, Durham, N.C., USA
| |
Collapse
|
42
|
Gao F, Fish BL, Szabo A, Doctrow SR, Kma L, Molthen RC, Moulder JE, Jacobs ER, Medhora M. Short-term treatment with a SOD/catalase mimetic, EUK-207, mitigates pneumonitis and fibrosis after single-dose total-body or whole-thoracic irradiation. Radiat Res 2012; 178:468-80. [PMID: 23020094 DOI: 10.1667/rr2953.1] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
In the event of a radiological accident or terrorist attack, whole- or partial-body exposure can injure the lungs. To simulate such an incident, we used a single fraction of total-body irradiation (TBI) or whole-thoracic irradiation to induce pneumonitis or pulmonary fibrosis, respectively, in a rat model. The superoxide dismutase and catalase mimetic EUK-207 was given by subcutaneous injection (20 mg/kg/day, 5 days per week, once daily) starting at 7 days after irradiation and stopping before pneumonitis developed. After TBI, morbidity and the increase in breathing rates associated with pneumonitis were significantly improved in rats treated with EUK-207 compared to rats receiving irradiation alone. At 42 days after TBI (the peak of pneumonitis) changes in vascular end points including pulmonary hemodynamics ex vivo and relative arterial density in lungs were also mitigated by EUK-207. At 7 months after whole-thoracic irradiation, EUK-207 reduced synthesis of collagen as assessed by the Sircol collagen assay and Masson's trichrome staining. Our results demonstrate promise for EUK-207 as a mitigator of radiation pneumonitis and fibrosis. We also demonstrate for the first time mitigation of multiple vascular injuries in the irradiated lung in vivo by EUK-207.
Collapse
Affiliation(s)
- Feng Gao
- Departments of Radiation Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ajami M, Davoodi SH, Habibey R, Namazi N, Soleimani M, Pazoki-Toroudi H. Effect of DHA+EPA on oxidative stress and apoptosis induced by ischemia-reperfusion in rat kidneys. Fundam Clin Pharmacol 2012; 27:593-602. [PMID: 22943605 DOI: 10.1111/j.1472-8206.2012.01066.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2012] [Revised: 06/04/2012] [Accepted: 07/10/2012] [Indexed: 12/17/2022]
Abstract
Apoptosis, as well as necrosis, has an important role in post-ischemic renal pathology. The effect of pretreatment with Docosahexaenoic acid+Eicosapentaenoic acid (DHA+EPA) on renal injury and apoptotic protein expression was evaluated. Right nephrectomy was completed on male Wistar rats (255-300 g). The rats received DHA+EPA (200 mg/kg/day) of distilled water orally for 14 days before ischemia reperfusion (IR) or sham operation. A total of 81 rats were divided into three main groups with 6, 24 and 48 h of post-operation or reperfusion period. Serum creatinine (SCr), BUN, creatinine clearance (CCr) and fractional excretion of sodium (FEN a ) were measured. Tissue levels of malondialdehyde (MDA), superoxide dismutase (SOD) and catalase (CAT) activities, Bax and Bcl-2 protein expressions and renal histological injury were determined. SCr, BUN and FEN a increased 6-48 h of reperfusion (P < 0.01). Tissue MDA content and Bax expression increased (P < 0.01) and CAT and SOD activities decreased (P < 0.05) in the IR group. DHA+EPA decreased SCr and BUN, FEN a , tissue MDA levels (P < 0.05 vs. IR) and increased CAT and SOD activities and Bcl-2 expression (P < 0.05 vs. IR) for 6-48 h after ischemia. IR induced mild (6 h, P < 0.05) and severe (24-48 h, P < 0.01) tissue damage. Mild-to-moderate tissue damage was observed in DHA+EPA groups from 6 to 48 h of reperfusion period (P < 0.05 vs. IR, 24-48 h). In conclusion, the results suggest that pre-ischemic exposure to DHA+EPA could improve the outcome of early graft function by inhibition of IR-induced oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Marjan Ajami
- Faculty of Nutrition Sciences & Food Technology, Shahid Beheshti University of Medical Sciences & Health Services, 19395-4741, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
44
|
Wang G, Zhang J, Dewilde AH, Pal AK, Bello D, Therrien JM, Braunhut SJ, Marx KA. Understanding and correcting for carbon nanotube interferences with a commercial LDH cytotoxicity assay. Toxicology 2012; 299:99-111. [DOI: 10.1016/j.tox.2012.05.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 04/26/2012] [Accepted: 05/14/2012] [Indexed: 10/28/2022]
|
45
|
Matés JM, Segura JA, Alonso FJ, Márquez J. Oxidative stress in apoptosis and cancer: an update. Arch Toxicol 2012; 86:1649-65. [PMID: 22811024 DOI: 10.1007/s00204-012-0906-3] [Citation(s) in RCA: 246] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 07/03/2012] [Indexed: 02/07/2023]
Abstract
The oxygen paradox tells us that oxygen is both necessary for aerobic life and toxic to all life forms. Reactive oxygen species (ROS) touch every biological and medical discipline, especially those involving proliferative status, supporting the idea that active oxygen may be increased in tumor cells. In fact, metabolism of oxygen and the resulting toxic byproducts can cause cancer and death. Efforts to counteract the damage caused by ROS are gaining acceptance as a basis for novel therapeutic approaches, and the field of prevention of cancer is experiencing an upsurge of interest in medically useful antioxidants. Apoptosis is an important means of regulating cell numbers in the developing cell system, but it is so important that it must be controlled. Normal cell death in homeostasis of multicellular organisms is mediated through tightly regulated apoptotic pathways that involve oxidative stress regulation. Defective signaling through these pathways can contribute to both unbalance in apoptosis and development of cancer. Finally, in this review, we discuss new knowledge about recent tools that provide powerful antioxidant strategies, and designing methods to deliver to target cells, in the prevention and treatment of cancer.
Collapse
Affiliation(s)
- José M Matés
- Department of Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Campus de Teatinos, Málaga, Spain.
| | | | | | | |
Collapse
|
46
|
Sepp T, Sild E, Blount JD, Männiste M, Karu U, Hõrak P. Individual Consistency and Covariation of Measures of Oxidative Status in Greenfinches. Physiol Biochem Zool 2012; 85:299-307. [DOI: 10.1086/664827] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
47
|
Miriyala S, Spasojevic I, Tovmasyan A, Salvemini D, Vujaskovic Z, St. Clair D, Batinic-Haberle I. Manganese superoxide dismutase, MnSOD and its mimics. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1822:794-814. [PMID: 22198225 PMCID: PMC3304004 DOI: 10.1016/j.bbadis.2011.12.002] [Citation(s) in RCA: 294] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 12/02/2011] [Accepted: 12/02/2011] [Indexed: 12/20/2022]
Abstract
Increased understanding of the role of mitochondria under physiological and pathological conditions parallels increased exploration of synthetic and natural compounds able to mimic MnSOD - endogenous mitochondrial antioxidant defense essential for the existence of virtually all aerobic organisms from bacteria to humans. This review describes most successful mitochondrially-targeted redox-active compounds, Mn porphyrins and MitoQ(10) in detail, and briefly addresses several other compounds that are either catalysts of O(2)(-) dismutation, or its non-catalytic scavengers, and that reportedly attenuate mitochondrial dysfunction. While not a true catalyst (SOD mimic) of O(2)(-) dismutation, MitoQ(10) oxidizes O(2)(-) to O(2) with a high rate constant. In vivo it is readily reduced to quinol, MitoQH(2), which in turn reduces ONOO(-) to NO(2), producing semiquinone radical that subsequently dismutes to MitoQ(10) and MitoQH(2), completing the "catalytic" cycle. In MitoQ(10), the redox-active unit was coupled via 10-carbon atom alkyl chain to monocationic triphenylphosphonium ion in order to reach the mitochondria. Mn porphyrin-based SOD mimics, however, were designed so that their multiple cationic charge and alkyl chains determine both their remarkable SOD potency and carry them into the mitochondria. Several animal efficacy studies such as skin carcinogenesis and UVB-mediated mtDNA damage, and subcellular distribution studies of Saccharomyces cerevisiae and mouse heart provided unambiguous evidence that Mn porphyrins mimic the site and action of MnSOD, which in turn contributes to their efficacy in numerous in vitro and in vivo models of oxidative stress. Within a class of Mn porphyrins, lipophilic analogs are particularly effective for treating central nervous system injuries where mitochondria play key role. This article is part of a Special Issue entitled: Antioxidants and Antioxidant Treatment in Disease.
Collapse
Affiliation(s)
- Sumitra Miriyala
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY, 40536
| | - Ivan Spasojevic
- Department of Medicine, Duke University Medical Center, Durham, NC 27710
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710
| | - Daniela Salvemini
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, 1402 South Grand Blvd, St. Louis, MO 63104
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710
| | - Daret St. Clair
- Graduate Center for Toxicology, University of Kentucky, Lexington, KY, 40536
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
48
|
Williams JP, Jackson IL, Shah JR, Czarniecki CW, Maidment BW, DiCarlo AL. Animal models and medical countermeasures development for radiation-induced lung damage: report from an NIAID Workshop. Radiat Res 2012; 177:e0025-39. [PMID: 22468702 DOI: 10.1667/rrol04.1] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since 9/11, there have been concerns that terrorists may detonate a radiological or nuclear device in an American city. Aside from several decorporation and blocking agents for use against internal radionuclide contamination, there are currently no medications within the Strategic National Stockpile that are approved to treat the immediate or delayed complications resulting from accidental exposure to radiation. Although the majority of research attention has focused on developing countermeasures that target the bone marrow and gastrointestinal tract, since they represent the most acutely radiosensitive organs, individuals who survive early radiation syndromes will likely suffer late effects in the months that follow. Of particular concern are the delayed effects seen in the lung that play a major role in late mortality seen in radiation-exposed patients and accident victims. To address these concerns, the National Institute of Allergy and Infectious Diseases convened a workshop to discuss pulmonary model development, mechanisms of radiation-induced lung injury, targets for medical countermeasures development, and end points to evaluate treatment efficacy. Other topics covered included guidance on the challenges of developing and licensing drugs and treatments specific to a radiation lung damage indication. This report reviews the data presented, as well as key points from the ensuing discussion.
Collapse
|
49
|
Arsenic trioxide enhances the radiation sensitivity of androgen-dependent and -independent human prostate cancer cells. PLoS One 2012; 7:e31579. [PMID: 22363680 PMCID: PMC3282747 DOI: 10.1371/journal.pone.0031579] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 01/09/2012] [Indexed: 01/07/2023] Open
Abstract
Prostate cancer is the most common malignancy in men. In the present study, LNCaP (androgen-sensitive human prostate cancer cells) and PC-3 cells (androgen-independent human prostate cancer cells) were used to investigate the anti-cancer effects of ionizing radiation (IR) combined with arsenic trioxide (ATO) and to determine the underlying mechanisms in vitro and in vivo. We found that IR combined with ATO increases the therapeutic efficacy compared to individual treatments in LNCaP and PC-3 human prostate cancer cells. In addition, combined treatment showed enhanced reactive oxygen species (ROS) generation compared to treatment with ATO or IR alone in PC-3 cells. Combined treatment induced autophagy and apoptosis in LNCaP cells, and mainly induced autophagy in PC-3 cells. The cell death that was induced by the combined treatment was primarily the result of inhibition of the Akt/mTOR signaling pathways. Furthermore, we found that the combined treatment of cells pre-treated with 3-MA resulted in a significant change in AO-positive cells and cytotoxicity. In an in vivo study, the combination treatment had anti-tumor growth effects. These novel findings suggest that combined treatment is a potential therapeutic strategy not only for androgen-dependent prostate cancer but also for androgen-independent prostate cancer.
Collapse
|
50
|
Tominaga T, Hachiya M, Shibata T, Sakamoto Y, Taki K, Akashi M. Exogenously-added copper/zinc superoxide dismutase rescues damage of endothelial cells from lethal irradiation. J Clin Biochem Nutr 2011; 50:78-83. [PMID: 22247605 PMCID: PMC3246187 DOI: 10.3164/jcbn.11-15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Accepted: 04/13/2011] [Indexed: 01/27/2023] Open
Abstract
The vascular endothelium is important for the early and late effects observed in lethally irradiated tissue and organs. We examined the effects of exogenously added superoxide dismutase on cell survival and angiogenesis in lethally irradiated human primary umbilical vein endothelial cells. Cell survival was significantly improved in superoxide dismutase-treated cells; the addition of superoxide dismutase to cells after irradiation was also effective for increased survival, as it was before irradiation. Moreover, treatment of cells with superoxide dismutase enhanced the phosphorylation of mitogen-activated protein/extracellular signal-regulated kinase/extracellular signal regulated kinases 1 and 2 in human primary umbilical vein endothelial cells. The addition of superoxide dismutase to cells after irradiation attenuated the reduction of angiogenesis by irradiation, and inhibition of the mitogen-activated protein/extracellular signal-regulated kinase/extracellular signal regulated kinases signaling pathway abrogated the rescue effect of superoxide dismutase. Our results suggest that superoxide dismutase rescues human primary umbilical vein endothelial cells from endothelial dysfunction caused by irradiation via a pathway requiring activation of mitogen-activated protein/extracellular signal-regulated kinase/extracellular signal regulated kinases 1 and 2.
Collapse
Affiliation(s)
- Takako Tominaga
- Department of Radiation Emergency Medicine, The Research Center for Radiation Emergency Medicine, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | | | | | | | | | | |
Collapse
|