1
|
Liu H, Wei X, He Y, Pan S, Wang C, Cheng J, Zhao Q, Shi K, Si H. Elucidating the antiviral effects of a novel compound throat anti-viral through metabolomics and network pharmacology: A study on infectious bronchitis virus in poultry. Poult Sci 2025; 104:104956. [PMID: 40127563 PMCID: PMC11984592 DOI: 10.1016/j.psj.2025.104956] [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: 01/04/2025] [Revised: 02/09/2025] [Accepted: 02/26/2025] [Indexed: 03/26/2025] Open
Abstract
Infectious bronchitis virus (IBV) is a major pathogen that causes significant economic losses in the global poultry industry. Current vaccination strategies provide only partial protection, highlighting the need for more effective prevention and treatment methods. This study aimed to develop a novel compound throat anti-viral (CTA) from natural plants using data from the Traditional Chinese Medicine Inheritance System and identification through liquid chromatography-mass spectrometry. CTA demonstrated substantial anti-IBV effects both in vitro and in vivo studies. In vitro, CTA significantly inhibited IBV multiplication and alleviated the pathological lesions in chicken embryonic kidney cells, tracheal rings, and chicken embryos. In vivo, a seven-day treatment with CTA obtained much milder clinical signs, enhanced growth performance, and better immune organ indices in infected chickens. Additionally, CTA treatment reduced IBV levels in the trachea and lungs and increased specific antibody titers. CTA also maintained body homeostasis, exhibiting strong antioxidant and anti-inflammatory properties that mitigated respiratory tract damage. Metabolomics and network pharmacology analyses, revealed that CTA's antiviral effects are mediated through the FoxO signaling pathway. This study successfully developed an effective prescription database based on the Traditional Chinese Medicine Inheritance System and validated the antiviral efficacy of CTA through comprehensive in vitro and in vivo experiments. The findings elucidated the mechanisms of CTA's action, particularly through the FoxO signaling pathway, and highlighted its potential for clinical application as a novel antiviral treatment for IBV in the poultry industry.
Collapse
Affiliation(s)
- Huixin Liu
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China
| | - Xiaofang Wei
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China
| | - Yang He
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China
| | - Sijia Pan
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China
| | - Chenchen Wang
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China
| | - Junze Cheng
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China
| | - Qiyuan Zhao
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China
| | - Kaichuang Shi
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China; Guangxi Center for Animal Disease Control and Prevention, Nanning 530001, China
| | - Hongbin Si
- College of Animal Science and Technology, Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Guangxi grass station, Guangxi University, Nanning, 530004, Guangxi, China.
| |
Collapse
|
2
|
Ma J, Huang J, Sun J, Zhou Y, Ji X, Guo D, Liu C, Li J, Zhang J, Song H. L-Se-methylselenocysteine sensitizes lung carcinoma to chemotherapy. Cell Prolif 2021; 54:e13038. [PMID: 33793020 PMCID: PMC8088472 DOI: 10.1111/cpr.13038] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/17/2021] [Accepted: 03/17/2021] [Indexed: 12/14/2022] Open
Abstract
Objectives Organic Selenium (Se) compounds such as L‐Se‐methylselenocysteine (L‐SeMC/SeMC) have been employed as a class of anti‐oxidant to protect normal tissues and organs from chemotherapy‐induced systemic toxicity. However, their comprehensive effects on cancer cell proliferation and tumour progression remain elusive. Materials and Methods CCK‐8 assays were conducted to determine the viabilities of cancer cells after exposure to SeMC, chemotherapeutics or combined treatment. Intracellular reactive oxygen species (ROS) levels and lipid peroxidation levels were assessed via fluorescence staining. The efficacy of free drugs or drug‐loaded hydrogel against tumour growth was evaluated in a xenograft mouse model. Results Among tested cancer cells and normal cells, the A549 lung adenocarcinoma cells showed higher sensitivity to SeMC exposure. In addition, combined treatments with several types of chemotherapeutics induced synergistic lethality. SeMC promoted lipid peroxidation in A549 cells and thereby increased ROS generation. Significantly, the in vivo efficacy of combination therapy was largely potentiated by hydrogel‐mediate drug delivery. Conclusions Our study reveals the selectivity of SeMC in the inhibition of cancer cell proliferation and develops an efficient strategy for local combination therapy.
Collapse
Affiliation(s)
- Jia Ma
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, China.,State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Huang
- Department of Neurology, Xuhui District Central Hospital, Shanghai, China
| | - Jinli Sun
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanfeng Zhou
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyuan Ji
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Daoxia Guo
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chang Liu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiyu Li
- Henan Xibaikang Health Industry Co., Ltd, Jiyuan, China
| | - Jiye Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Haiyun Song
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Zhang X, Fisher R, Hou W, Shields D, Epperly MW, Wang H, Wei L, Leibowitz BJ, Yu J, Alexander LM, VAN Pijkeren JP, Watkins S, Wipf P, Greenberger JS. Second-generation Probiotics Producing IL-22 Increase Survival of Mice After Total Body Irradiation. In Vivo 2020; 34:39-50. [PMID: 31882461 PMCID: PMC6984118 DOI: 10.21873/invivo.11743] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/21/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND/AIM Intestinal damage induced by total body irradiation (TBI) reduces leucine-rich repeat-containing G-protein-coupled receptor 5 (Lgr5)-expressing stem cells, goblet, and Paneth cells, breaching the epithelial lining, and facilitating bacterial translocation, sepsis, and death. MATERIALS AND METHODS Survival was measured after TBI in animals that received wild-type or recombinant bacteria producing interleukin-22 (IL-22). Changes in survival due to microbially delivered IL-22 were measured. Lactobacillus reuteri producing IL-22, or Escherichia coli-IL-22 were compared to determine which delivery system is better. RESULTS C57BL/6 mice receiving IL-22 probiotics at 24 h after 9.25 Gy TBI, demonstrated green fluorescent protein-positive bacteria in the intestine, doubled the number of Lgr5+ intestinal stem cells, and increased 30-day survival. Bacteria were localized to the jejunum, ileum, and colon. CONCLUSION Second-generation probiotics appear to be valuable for mitigation of TBI, and radiation protection during therapeutic total abdominal irradiation.
Collapse
Affiliation(s)
- Xichen Zhang
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Wen Hou
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Donna Shields
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Michael W Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Liang Wei
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Brian J Leibowitz
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Jian Yu
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Laura M Alexander
- Department of Food Science, University of Wisconsin-Madison, Madison, WI, U.S.A
| | | | - Simon Watkins
- Center for Imaging, Department of Pathology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Joel S Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A.
| |
Collapse
|
4
|
Varlamova EG, Cheremushkina IV. Contribution of mammalian selenocysteine-containing proteins to carcinogenesis. J Trace Elem Med Biol 2017; 39:76-85. [PMID: 27908428 DOI: 10.1016/j.jtemb.2016.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 07/28/2016] [Accepted: 08/09/2016] [Indexed: 12/17/2022]
Abstract
Oxidative stress caused by a sharp growth of free radicals in the organism is a major cause underlying the occurrence of all kinds of malignant formations. Selenium is an important essential trace element found in selenoproteins in the form of selenocysteine, an amino acid differing from cysteine for the presence of selenium instead of sulfur and making such proteins highly active. To date the role of selenium has been extensively investigated through studying the functions of selenoproteins in carcinogenesis. Analysis of the obtained results clearly demonstrates that selenoproteins can act as oncosuppressors, but can also, on the contrary, favor the formation of malignant tumors.
Collapse
Affiliation(s)
- Elena Gennadyevna Varlamova
- Federal State Institution of Science Institute of Cell Biophysics, Russian Academy of Sciences, Moscow Region, Institutskaya st. 3, 142290, Pushchino, Russia.
| | - Irina Valentinovna Cheremushkina
- Federal State Educational Institution of Higher Education Voronezh State University of Engineering Technology, Prospect revolution st. 19, 394000, Voronezh, Russia.
| |
Collapse
|
5
|
Greenberger J, Kagan V, Bayir H, Wipf P, Epperly M. Antioxidant Approaches to Management of Ionizing Irradiation Injury. Antioxidants (Basel) 2015; 4:82-101. [PMID: 26785339 PMCID: PMC4665573 DOI: 10.3390/antiox4010082] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 01/12/2015] [Indexed: 11/25/2022] Open
Abstract
Ionizing irradiation induces acute and chronic injury to tissues and organs. Applications of antioxidant therapies for the management of ionizing irradiation injury fall into three categories: (1) radiation counter measures against total or partial body irradiation; (2) normal tissue protection against acute organ specific ionizing irradiation injury; and (3) prevention of chronic/late radiation tissue and organ injury. The development of antioxidant therapies to ameliorate ionizing irradiation injury began with initial studies on gene therapy using Manganese Superoxide Dismutase (MnSOD) transgene approaches and evolved into applications of small molecule radiation protectors and mitigators. The understanding of the multiple steps in ionizing radiation-induced cellular, tissue, and organ injury, as well as total body effects is required to optimize the use of antioxidant therapies, and to sequence such approaches with targeted therapies for the multiple steps in the irradiation damage response.
Collapse
Affiliation(s)
- Joel Greenberger
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Rm. 533, Pittsburgh, PA 15232, USA.
| | - Valerian Kagan
- Department of Environmental/Occupational Health, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| | - Hulya Bayir
- Department of Critical Care Medicine, Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA.
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, USA.
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| | - Michael Epperly
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Rm. 533, Pittsburgh, PA 15232, USA.
| |
Collapse
|
6
|
Greenberger JS, Clump D, Kagan V, Bayir H, Lazo JS, Wipf P, Li S, Gao X, Epperly MW. Strategies for discovery of small molecule radiation protectors and radiation mitigators. Front Oncol 2012; 1:59. [PMID: 22655254 PMCID: PMC3356036 DOI: 10.3389/fonc.2011.00059] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 12/20/2011] [Indexed: 01/01/2023] Open
Abstract
Mitochondrial targeted radiation damage protectors (delivered prior to irradiation) and mitigators (delivered after irradiation, but before the appearance of symptoms associated with radiation syndrome) have been a recent focus in drug discovery for (1) normal tissue radiation protection during fractionated radiotherapy, and (2) radiation terrorism counter measures. Several categories of such molecules have been discovered: nitroxide-linked hybrid molecules, including GS-nitroxide, GS-nitric oxide synthase inhibitors, p53/mdm2/mdm4 inhibitors, and pharmaceutical agents including inhibitors of the phosphoinositide-3-kinase pathway and the anti-seizure medicine, carbamazepine. Evaluation of potential new radiation dose modifying molecules to protect normal tissue includes: clonogenic radiation survival curves, assays for apoptosis and DNA repair, and irradiation-induced depletion of antioxidant stores. Studies of organ specific radioprotection and in total body irradiation-induced hematopoietic syndrome in the mouse model for protection/mitigation facilitate rational means by which to move candidate small molecule drugs along the drug discovery pipeline into clinical development.
Collapse
Affiliation(s)
- Joel S. Greenberger
- Radiation Oncology Department, University of Pittsburgh Cancer InstitutePittsburgh, PA, USA
| | - David Clump
- Radiation Oncology Department, University of Pittsburgh Cancer InstitutePittsburgh, PA, USA
| | - Valerian Kagan
- Environmental and Occupational Health Department, University of PittsburghPittsburgh, PA, USA
| | - Hülya Bayir
- Critical Care Medicine Department, University of Pittsburgh Medical CenterPittsburgh, PA, USA
| | - John S. Lazo
- Pharmacology Department, University of VirginiaCharlottesville, VA, USA
| | - Peter Wipf
- Department of Chemistry, Accelerated Chemical Discovery Center, University of PittsburghPittsburgh, PA, USA
| | - Song Li
- Pharmaceutical Science Department, University of PittsburghPittsburgh, PA, USA
| | - Xiang Gao
- Pharmaceutical Science Department, University of PittsburghPittsburgh, PA, USA
| | - Michael W. Epperly
- Radiation Oncology Department, University of Pittsburgh Cancer InstitutePittsburgh, PA, USA
| |
Collapse
|
7
|
Gonzalez-Moreno O, Boque N, Redrado M, Milagro F, Campion J, Endermann T, Takahashi K, Saito Y, Catena R, Schomburg L, Calvo A. Selenoprotein-P is down-regulated in prostate cancer, which results in lack of protection against oxidative damage. Prostate 2011; 71:824-834. [PMID: 21456065 DOI: 10.1002/pros.21298] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Accepted: 09/26/2010] [Indexed: 02/05/2023]
Abstract
BACKGROUND Oxidative stress plays a role in prostate cancer (PrCa) initiation and development. Selenoprotein-P (SepP; a protein involved in antioxidant defence) mRNA levels are down-regulated in PrCa. The main goal of our study was to assess whether SepP protects prostate cells from reactive oxygen species (ROS) in prostate carcinogenesis. METHODS Modification of SepP levels and ROS conditions in C3(1)/Tag-derived cell lines representing prostate epithelial neoplasia (PIN) lesions (Pr-111, with high SepP expression); and invasive tumors (Pr-14, with very low SepP expression). RESULTS Both Pr-111 and Pr-14 cells express ApoER2 (SepP receptor), which suggests that they may uptake SepP. Pr-14 cells had much higher ROS levels than Pr-111 cells and were highly sensitive to H(2)O(2)-mediated cytotoxicity. When SepP mRNA levels were knocked down with siRNAs in Pr-111 cells, a significant increase in ROS and cell growth inhibition upon H(2)O(2) exposure was found. Subsequent administration of purified SepP in the culture medium of these cells was able to rescue the original phenotype. Similarly, administration of SepP to Pr-14 cells was able to reduce ROS concentrations. Administration of flutamide decreased SepP mRNA levels whereas dihydrotestosterone or synthetic androgens induced SepP expression, indicating the importance of androgens for SepP expression. Immunohistochemical analysis using a PrCa tissue microarray further revealed that SepP protein was reduced in 60.8% prostate tumors compared to benign prostates. CONCLUSIONS Levels of SepP in prostate cells determine basal ROS levels and sensitivity to H(2)O(2)-induced cytotoxicity. Deregulation of SepP during prostate carcinogenesis may increase free radicals, thus promoting tumor development and de-differentiation.
Collapse
Affiliation(s)
- Oscar Gonzalez-Moreno
- Division of Oncology, Center for Applied Medical Research, Department of Histology and Pathology, University of Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Lipid replacement therapy: a nutraceutical approach for reducing cancer-associated fatigue and the adverse effects of cancer therapy while restoring mitochondrial function. Cancer Metastasis Rev 2010; 29:543-52. [DOI: 10.1007/s10555-010-9245-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
9
|
Adaramoye OA, Adedara IA, Popoola B, Farombi EO. Extract of Xylopia aethiopica (Annonaceae) protects against gamma-radiation induced testicular damage in Wistar rats. J Basic Clin Physiol Pharmacol 2010; 21:295-313. [PMID: 21305847 DOI: 10.1515/jbcpp.2010.21.4.295] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Ionizing radiation is an important environmental risk factor and, a major therapeutic agent for cancer treatment. This study was designed to evaluate the protective effect of extract of Xylopia aethiopica (XA) on gamma-radiation-induced testicular damage in rats. Vitamin C (VC) served as the reference antioxidant during the study. The study consists of 4 groups of 11 rats each. Group I received corn oil (vehicle), groups II and IV were pretreated with XA (250 mg/kg) and VC (250mg/kg) for 6 weeks before and 8 weeks after exposure to gamma-radiation; group III was exposed to a single dose of gamma-radiation (5 Gy). Biochemical analysis revealed that gamma-irradiation caused a significant increase (p < .05) in serum and testicular lipid peroxidation (LPO) levels by 217% and 221%, respectively. Irradiated rats had markedly decreased testicular catalase (CAT), superoxide dismutase (SOD), glutathione-S-transferase (GST), and reduced glutathione (GSH) levels. Irradiation resulted in 59% and 40% decreases in spermatozoa motility and live/dead sperm count, respectively, and a 161% increase in total sperm abnormalities. Histologically, testes of the irradiated rats showed extensive degenerative changes in the seminiferous tubules and defoliation of spermatocytes. Supplementation of XA and VC reversed the adverse effects of gamma-radiation on biochemical and histological indices of the rats. These findings demonstrated that Xylopia aethiopica has a protective effect by inhibiting oxidative damage in testes of irradiated rats.
Collapse
Affiliation(s)
- Oluwatosin Adekunle Adaramoye
- Drug Metabolism Research Laboratory, College of Medicine, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Nigeria.
| | | | | | | |
Collapse
|
10
|
Chandrasekharan DK, Kagiya TV, Nair CKK. Radiation protection by 6-palmitoyl ascorbic acid-2-glucoside: studies on DNA damage in vitro, ex vivo, in vivo and oxidative stress in vivo. JOURNAL OF RADIATION RESEARCH 2009; 50:203-212. [PMID: 19384055 DOI: 10.1269/jrr.08090] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
A palmitoyl derivative of ascorbic acid 2-glucoside, 6-palmitoyl ascorbic acid-2-glucoside (PAsAG), which possess good antioxidant properties, is examined for radioprotection in vitro, ex vivo and in vivo models. PAsAG protected plasmid DNA from gamma-radiation induced damages under in vitro conditions. Presence of 1.6 mM PAsAG inhibited the disappearance of ccc (covalently closed circular) form of plasmid pBR322 with a dose modifying factor of 1.5. Comet assay studies on mouse spleen cells exposed to 6 Gy gamma-radiation (ex vivo) in presence and absence of PAsAG revealed that cellular DNA was effectively protected by this compound from radiation induced damages. Oral administration of 80 mg/kg body weight of PAsAG to mice 1 hour prior to 6 Gy whole body gamma-radiation exposure, efficiently protected cellular DNA in tissues such as spleen, bone marrow and blood, from radiation induced damages as indicated by alkaline comet assay. Oxidative stress in tissues such as liver and brain of mice, following whole body exposure to various doses of gamma-radiation (2-8 Gy), monitored as levels of GSH and peroxidation of lipids, were found considerably reduced when PAsAG was orally administered (80 mg/kg body weight) to the mice one hour prior to the radiation exposure. PAsAG administration improved the per cent survival of mice following exposure to 10 Gy whole body gamma-radiation. Thus PAsAG could act as a radioprotector under in vitro, ex vivo and in vivo conditions of ionizing-radiation exposure.
Collapse
|
11
|
Zabbarova I, Kanai A. Targeted delivery of radioprotective agents to mitochondria. Mol Interv 2009; 8:294-302. [PMID: 19144902 DOI: 10.1124/mi.8.6.7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Adverse effects of ionizing radiation are mediated through reactive oxygen and nitrogen species. Mitochondria are the principal source of these species in the cell and play an important role in irradiation-induced apoptosis. The use of free radical scavengers and nitric oxide synthase inhibitors has proven to protect normal tissues and, in some cases, to sensitize tumor tissues to radiation damage. Dual molecules that combine radical-scavenging and NOS-inhibitory functions may be particularly effective. Drugging strategies that target mitochondria can enhance the effectiveness of such agents, in comparison to systemic administration, and circumvent side effects.
Collapse
Affiliation(s)
- Irina Zabbarova
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | | |
Collapse
|
12
|
Agostinelli E, Tempera G, Dalla Vedova L, Condello M, Arancia G. MDL 72527 and spermine oxidation products induce a lysosomotropic effect and mitochondrial alterations in tumour cells. Biochem Soc Trans 2007; 35:343-8. [PMID: 17371275 DOI: 10.1042/bst0350343] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Cytotoxic products of polyamines generated in situ by an enzyme-catalysed reaction may be useful as a new avenue in combating cancer. This study demonstrated that MDR (multidrug-resistant) cancer cells (colon adenocarcinoma and melanoma) are significantly more sensitive than the corresponding WT (wild-type) ones to H(2)O(2) and aldehydes, the products of BSAO (bovine serum amine oxidase)-catalysed oxidation of spermine. Moreover, cytotoxicity was considerably greater when the treatment was carried out at 42 degrees C than at 37 degrees C. TEM (transmission electron microscopy) observations showed major ultrastructural alterations of the mitochondria. These were more pronounced in MDR than in WT cells. After treatment with BSAO/spermine, a higher mitochondrial membrane depolarization and an increased mitochondrial activity in drug-resistant cells were observed.
Collapse
Affiliation(s)
- E Agostinelli
- Department of Biochemical Sciences A Rossi Fanelli, University of Rome La Sapienza and CNR, Biology and Molecular Pathology Institutes, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | | | | | | | | |
Collapse
|
13
|
Agostinelli E, Seiler N. Non-irradiation-derived reactive oxygen species (ROS) and cancer: therapeutic implications. Amino Acids 2006; 31:341-55. [PMID: 16680401 DOI: 10.1007/s00726-005-0271-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2005] [Accepted: 10/11/2005] [Indexed: 12/21/2022]
Abstract
Owing to their chemical reactivity, radicals have cytocidal properties. Destruction of cells by irradiation-induced radical formation is one of the most frequent interventions in cancer therapy. An alternative to irradiation-induced radical formation is in principle drug-induced formation of radicals, and the formation of toxic metabolites by enzyme catalysed reactions. Although these developments are currently still in their infancy, they nevertheless deserve consideration. There are now numerous examples known of conventional anti-cancer drugs that may at least in part exert cytotoxicity by induction of radical formation. Some drugs, such as arsenic trioxide and 2-methoxy-estradiol, were shown to induce programmed cell death due to radical formation. Enzyme-catalysed radical formation has the advantage that cytotoxic products are produced continuously over an extended period of time in the vicinity of tumour cells. Up to now the enzymatic formation of toxic metabolites has nearly exclusively been investigated using bovine serum amine oxidase (BSAO), and spermine as substrate. The metabolites of this reaction, hydrogen peroxide and aldehydes are cytotoxic. The combination of BSAO and spermine is not only able to prevent tumour cell growth, but prevents also tumour growth, particularly well if the enzyme has been conjugated with a biocompatible gel. Since the tumour cells release substrates of BSAO, the administration of spermine is not required. Combination with cytotoxic drugs, and elevation of temperature improves the cytocidal effect of spermine metabolites. The fact that multidrug resistant cells are more sensitive to spermine metabolites than their wild type counterparts makes this new approach especially attractive, since the development of multidrug resistance is one of the major problems of conventional cancer therapy.
Collapse
Affiliation(s)
- E Agostinelli
- Department of Biochemical Sciences A. Rossi Fanelli, University of Rome La Sapienza, Rome, Italy.
| | | |
Collapse
|
14
|
Brea-Calvo G, Rodríguez-Hernández A, Fernández-Ayala DJM, Navas P, Sánchez-Alcázar JA. Chemotherapy induces an increase in coenzyme Q10 levels in cancer cell lines. Free Radic Biol Med 2006; 40:1293-302. [PMID: 16631519 DOI: 10.1016/j.freeradbiomed.2005.11.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2005] [Revised: 10/13/2005] [Accepted: 11/16/2005] [Indexed: 11/17/2022]
Abstract
Free radicals have been implicated in the action of many chemotherapeutic drugs. Here we tested the hypothesis that camptothecin and other chemotherapeutic drugs, such as etoposide, doxorubicin, and methotrexate, induce an increase in coenzyme Q(10) levels as part of the antioxidant defense against free radical production under these anticancer treatments in cancer cell lines. Chemotherapy treatment induced both free radical production and an increase in coenzyme Q(10) levels in all the cancer cell lines tested. Reduced coenzyme Q(10) form levels were particularly enhanced. Coenzyme Q(10)-increased levels were associated with up-regulation of COQ genes expression, involved in coenzyme Q(10) biosynthesis. At the translational level, COQ7 protein expression levels were also increased. Furthermore, coenzyme Q(10) biosynthesis inhibition blocked camptothecin-induced coenzyme Q(10) increase, and enhanced camptothecin cytotoxicity. Our findings suggest that coenzyme Q(10) increase is implicated in the cellular defense under chemotherapy treatment and may contribute to cell survival.
Collapse
Affiliation(s)
- Gloria Brea-Calvo
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Carretera de Utrera Km 1, Sevilla, Spain
| | | | | | | | | |
Collapse
|
15
|
Skrzydlewska E, Sulkowska M, Koda M, Sulkowski S. Proteolytic-antiproteolytic balance and its regulation in carcinogenesis. World J Gastroenterol 2005; 11:1251-66. [PMID: 15761961 PMCID: PMC4250670 DOI: 10.3748/wjg.v11.i9.1251] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer development is essentially a tissue remodeling process in which normal tissue is substituted with cancer tissue. A crucial role in this process is attributed to proteolytic degradation of the extracellular matrix (ECM). Degradation of ECM is initiated by proteases, secreted by different cell types, participating in tumor cell invasion and increased expression or activity of every known class of proteases (metallo-, serine-, aspartyl-, and cysteine) has been linked to malignancy and invasion of tumor cells. Proteolytic enzymes can act directly by degrading ECM or indirectly by activating other proteases, which then degrade the ECM. They act in a determined order, resulting from the order of their activation. When proteases exert their action on other proteases, the end result is a cascade leading to proteolysis. Presumable order of events in this complicated cascade is that aspartyl protease (cathepsin D) activates cysteine proteases (e.g., cathepsin B) that can activate pro-uPA. Then active uPA can convert plasminogen into plasmin. Cathepsin B as well as plasmin are capable of degrading several components of tumor stroma and may activate zymogens of matrix metalloproteinases, the main family of ECM degrading proteases. The activities of these proteases are regulated by a complex array of activators, inhibitors and cellular receptors. In physiological conditions the balance exists between proteases and their inhibitors. Proteolytic-antiproteolytic balance may be of major significance in the cancer development. One of the reasons for such a situation is enhanced generation of free radicals observed in many pathological states. Free radicals react with main cellular components like proteins and lipids and in this way modify proteolytic-antiproteolytic balance and enable penetration damaging cellular membrane. All these lead to enhancement of proteolysis and destruction of ECM proteins and in consequence to invasion and metastasis.
Collapse
Affiliation(s)
- Elzbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2, 15-230 Bialystok, Poland.
| | | | | | | |
Collapse
|
16
|
Murley JS, Kataoka Y, Cao D, Li JJ, Oberley LW, Grdina DJ. Delayed radioprotection by NFkappaB-mediated induction of Sod2 (MnSOD) in SA-NH tumor cells after exposure to clinically used thiol-containing drugs. Radiat Res 2005; 162:536-46. [PMID: 15624308 DOI: 10.1667/rr3256] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The ability of thiol-containing reducing agents to activate transcription factors leading to changes in gene expression and enzyme activities provides an additional mechanism to potentially protect against radiation-induced cell killing. Manganese superoxide dismutase (Sod2) is one such gene whose expression levels have been shown to be elevated after exposure to the thiol compounds WR-1065 and N-acetyl-L-cysteine (NAC), resulting in an increase in radiation resistance. To further characterize this effect, SA-NH sarcoma cells, both wild-type and a clone stably transfected with a plasmid containing an IkappaBalpha gene mutated at serines 32 and 36, which prevents the inducible phosphorylation of these residues and the subsequent activation of NFkappaB (SA-NH+mIkappaBalpha1), were grown to confluence and then exposed to amifostine's free thiol WR-1065 at a concentration of 4 mM for 30 min. Effects of thiol exposure on NFKB activation in SA-NH+mIkappaBalpha1 cells were determined by a gel shift assay, and changes in Sod2 protein levels in these cells 24 h after exposure to 40 microM or 4 mM WR-1065 were measured by Western blot analysis and compared with wild-type cells exposed to the NFkappaB inhibitor BAY 11-7082. Changes in radiation response, measured immediately after thiol exposure or 24 h later, were determined using a colony-forming assay and were correlated with NFKB activation and Sod2 protein levels. The effects of captopril, mesna and NAC, each at a dose of 4 mM, on radiation response were also determined and contrasted with those of WR-1065. Only WR-1065 and captopril protected SA-NH cells when present during irradiation, i.e. 1.57 and 1.31 times increase in survival at 2 Gy, respectively. All four thiols were protective if irradiation with 2 Gy occurred 24 h later; i.e. increases in survival of 1.40, 1.22, 1.35, and 1.25 times were found for WR-1065, captopril, mesna and NAC, respectively. This delayed radioprotective effect correlated with elevated Sod2 protein levels in wild-type SA-NH tumor cells but was not observed in SA-NH+mIkappaBalpha1 cells, indicating that interference with thiol-induced NFKB activation abrogates this delayed radioprotective effect. Because the delayed radioprotective effect is readily demonstrable at a radiation dose of 2 Gy 24 h after exposure to clinically approved thiol-containing drugs such as amifostine, captopril, mesna and NAC, it suggests a new potential concern regarding the issue of tumor protection and the use of these agents in cancer therapy.
Collapse
Affiliation(s)
- Jeffrey S Murley
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, Illinois 60637, USA.
| | | | | | | | | | | |
Collapse
|
17
|
Yang J, Li H, Chen YY, Wang XJ, Shi GY, Hu QS, Kang XL, Lu Y, Tang XM, Guo QS, Yi J. Anthraquinones sensitize tumor cells to arsenic cytotoxicity in vitro and in vivo via reactive oxygen species-mediated dual regulation of apoptosis. Free Radic Biol Med 2004; 37:2027-41. [PMID: 15544921 DOI: 10.1016/j.freeradbiomed.2004.09.016] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2004] [Accepted: 09/17/2004] [Indexed: 01/03/2023]
Abstract
Cellular oxidation/reduction state affects the cytotoxicity of a number of chemotherapeutic agents, including arsenic trioxide. Reactive oxygen species (ROS), the major intracellular oxidants, may be a determinant of cellular susceptibility to arsenic. Our previous studies showed that a naphthoquinone and an anthraquinone (emodin) displayed the capability of producing ROS and facilitating arsenic cytotoxicity in both leukemia and solid tumor cell lines. We therefore attempted to test emodin and several other kinds of anthraquinone derivatives on EC/CUHK1, a cell line derived from esophageal carcinoma, and on a nude mouse model, with regard to their effects and mechanisms. Results showed that anthraquinones could produce ROS and sensitize tumor cells to arsenic both in vivo and in vitro. The combination of emodin and arsenic promoted the major apoptotic signaling events, i.e., the collapse of the mitochondrial transmembrane potential, the release of cytochrome c, and the activation of caspases 9 and 3. Meanwhile a combination of emodin and arsenic suppressed the activation of transcription factor NF-kappaB and downregulated the expression of a NF-kappaB-specific antiapoptotic protein, survivin. These two aspects could be antagonized by the antioxidant N-acetyl-L-cysteine. Therefore anthraquinones exert their effects via a ROS-mediated dual regulation, i.e., the enhancement of proapoptosis and the simultaneous inhibition of antiapoptosis. In vivo study showed that emodin made the EC/CUHK1 cell-derived tumors more sensitive to arsenic trioxide with no additional systemic toxicity and side effects. Taken together, these results suggest an innovative and safe chemotherapeutic strategy that uses natural anthraquinone derivatives as ROS generators to increase the susceptibility of tumor cells to cytotoxic therapeutic agents.
Collapse
Affiliation(s)
- Jie Yang
- Department of Cell Biology, Shanghai Second Medical University, 280 Chongqing Road, Shanghai 200025, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Wiegman EM, van Gameren MM, Kampinga HH, Szabó BG, Coppes RP. Post-irradiation dietary vitamin E does not affect the development of radiation-induced lung damage in rats. Radiother Oncol 2004; 72:67-70. [PMID: 15236876 DOI: 10.1016/j.radonc.2004.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2004] [Revised: 03/22/2004] [Accepted: 04/02/2004] [Indexed: 10/26/2022]
Abstract
The purpose of this study was to investigate whether application of post-irradiation vitamin E, an anti-oxidant, could prevent the development of radiation induced lung damage. Wistar rats were given vitamin E enriched or vitamin E deprived food starting from 4 weeks after 18Gy single dose irradiation of the right thorax. Neither breathing frequencies nor CT density measurements revealed differences between the groups. It is concluded that post-irradiation vitamin E does not influence radiation-induced fibrosis to the lung.
Collapse
Affiliation(s)
- Erwin M Wiegman
- Department of Radiation Oncology, University Hospital Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
19
|
Yi J, Yang J, He R, Gao F, Sang H, Tang X, Ye RD. Emodin enhances arsenic trioxide-induced apoptosis via generation of reactive oxygen species and inhibition of survival signaling. Cancer Res 2004; 64:108-16. [PMID: 14729614 DOI: 10.1158/0008-5472.can-2820-2] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although arsenic trioxide (As(2)O(3)) induces apoptosis in a relatively wide spectrum of tumors, the sensitivity of different cell types to this treatment varies to a great extent. Because reactive oxygen species (ROS) are critically involved in As(2)O(3)-induced apoptosis, we attempted to explore the possibility that elevating the cellular ROS level might be an approach to facilitate As(2)O(3)-induced apoptosis. Emodin, a natural anthraquinone derivative, was selected because its semiquinone structure is likely to increase the generation of intracellular ROS. Its independent and synergistic effects with As(2)O(3) in cytotoxicity were studied, and the plausible signaling mechanism was investigated in HeLa cells. Cell Proliferation Assay and flow cytometry were used to assess cell viability and apoptosis. Electrophoretic mobility shift assay, luciferase reporter assay, and Western blotting were performed to analyze signaling alteration. The results demonstrated that coadministration of emodin, at low doses of 0.5-10 micro M, with As(2)O(3) enhanced As(2)O(3)-rendered cytotoxicity on tumor cells, whereas these treatments caused no detectable proproliferative or proapoptotic effects on nontumor cells. ROS generation was increased, and activation of nuclear factor kappaB and activator protein 1 was suppressed by coadministration. All enhancements by emodin could be abolished by the antioxidant N-acetyl-L-cysteine. Therefore, we concluded that emodin sensitized HeLa cells to As(2)O(3) via generation of ROS and ROS-mediated inhibition on two major prosurvival transcription factors, nuclear factor kappaB and activator protein 1. This result allows us to propose a novel strategy in chemotherapy that uses mild ROS generators to facilitate apoptosis-inducing drugs whose efficacy depends on ROS.
Collapse
Affiliation(s)
- Jing Yi
- Department of Cell Biology, Shanghai Second Medical University, Shanghai, People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
20
|
Epperly MW, Osipov AN, Martin I, Kawai KK, Borisenko GG, Tyurina YY, Jefferson M, Bernarding M, Greenberger JS, Kagan VE. Ascorbate as a “redox sensor” and protector against irradiation-induced oxidative stress in 32D CL 3 hematopoietic cells and subclones overexpressing human manganese superoxide dismutase. Int J Radiat Oncol Biol Phys 2004; 58:851-61. [PMID: 14967442 DOI: 10.1016/j.ijrobp.2003.10.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2003] [Revised: 10/06/2003] [Accepted: 10/15/2003] [Indexed: 12/01/2022]
Abstract
PURPOSE To determine whether increased expression of manganese superoxide dismutase (MnSOD) protects cells from irradiation by preventing the production of reactive oxygen species (ROS), a new approach to detecting free radical intermediates using ascorbate as an endogenous spin trap was used. MATERIALS AND METHODS Cells from the 32D cl 3 hematopoietic cell line or a subclone overexpressing MnSOD (2C6) were incubated with dehydroascorbate for 30 min and irradiated to doses from 0 to 50 Gy. Radical intermediates reacting with spin traps or ascorbate were measured by electron spin resonance spectroscopy. Results were compared to irradiation-induced changes in thiol levels, irradiation survival curves, and accumulation of 8-OHdG as a measurement of DNA oxidative damage. RESULTS Manganese superoxide dismutase-overexpressing 2C6 cells maintained higher levels of ascorbate (5.4 +/- 0.5 and 2.6 +/- 0.5 nmol/10(6) cells, respectively) and thiols (14.0 +/- 0.1 and 11.1 +/- 0.2 nmol/10(6) cells) compared to 32D cl 3 parent cells. Cells overexpressing MnSOD produced lower levels of ROS than did the parental 32D cl 3 cells, as evidenced by lower expenditure of ascorbate and GSH after irradiation. Increased ascorbate levels protected both 32D cl 3 and 2C6 cells from irradiation killing, as demonstrated by an increased shoulder on survival curves and decreased DNA 8-OHdG accumulation. CONCLUSIONS Manganese superoxide dismutase overexpression protects 2C6 cells from irradiation damage by scavenging ROS that readily interact with major endogenous antioxidants--ascorbate and GSH--in nontransfected hematopoietic 32D cl 3 cells.
Collapse
Affiliation(s)
- Michael W Epperly
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Epperly MW, Bernarding M, Gretton J, Jefferson M, Nie S, Greenberger JS. Overexpression of the transgene for manganese superoxide dismutase (MnSOD) in 32D cl 3 cells prevents apoptosis induction by TNF-alpha, IL-3 withdrawal, and ionizing radiation. Exp Hematol 2003; 31:465-74. [PMID: 12829021 DOI: 10.1016/s0301-472x(03)00041-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Stabilization of the mitochondria in IL-3-dependent hematopoietic progenitor cell line 32D cl 3 by overexpression of the transgene for manganese superoxide dismutase (MnSOD) prior to ionizing radiation prevents apoptosis. We now demonstrate that overexpression of the MnSOD transgene also protects 32D cl 3 cells from apoptosis caused by exposure to tumor necrosis factor-alpha (TNF-alpha) or withdrawal of interleukin (IL)-3. MATERIALS AND METHODS The hematopoietic progenitor cell line, 32D cl 3, and subclones overexpressing the human MnSOD transgene, 1F2 or 2C6, were radiated to 1000 cGy or were exposed to TNF-alpha (0 to 100 etag/mL) or were subjected to IL-3 withdrawal. The cells were then examined at several time points for DNA strand breaks using a comet assay, depolarization of the mitochondrial membrane, activation of caspase-3, PARP cleavage, and apoptosis, and also for changes in cell cycle distribution. RESULTS Overexpression of the transgene for MnSOD resulted in increased survival following exposure to radiation, exposure to TNF-alpha, or IL-3 withdrawal. The cell lines overexpressing MnSOD (1F2 or 2C6) displayed decreased radiation-induced, TNF-alpha-induced, or IL-3 withdrawal-induced mitochondrial membrane permeability, caspase-3 and PARP activation, and apoptosis. CONCLUSIONS Overexpression of the human MnSOD transgene in 32D cl 3 cells results in stabilization of the mitochondria and reduction in radiation-, TNF-alpha-, or IL-3 withdrawal-induced damage. Thus, MnSOD stabilization of the mitochondrial membrane is relevant to reduction of apoptosis by several classes of oxidative stress inducers.
Collapse
Affiliation(s)
- Michael W Epperly
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Hillman Cancer Center, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | | | | | | | | | | |
Collapse
|
22
|
Liu B, Rotenberg SA, Mirkin MV. Scanning electrochemical microscopy of living cells. 4. Mechanistic study of charge transfer reactions in human breast cells. Anal Chem 2002; 74:6340-8. [PMID: 12510757 DOI: 10.1021/ac020564g] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Scanning electrochemical microscopy (SECM) has recently been employed for probing the redox properties of individual mammalian cells. It was shown that intracellular redox activity can be probed noninvasively by measuring the rate of mediator regeneration by the cell. Depending on the properties of the mediator species (e.g., formal potential, ionic charge, and hydrophobicity), different steps can limit the rate of the mediator regeneration reaction. This paper describes the evaluation of several factors that determine the rates of different steps of the process. These include intracellular concentration of redox centers, mixed redox potential inside the cell, and the rate of membrane permeation by mediator species. The kinetic analysis has been carried out to clarify the origins of different rates of the overall charge-transfer reaction in different cell types and with different redox mediators. The results can be used to facilitate differentiation between different types of cells, for example, normal and metastatic breast cells, on the basis of differences in redox responses.
Collapse
Affiliation(s)
- Biao Liu
- Department of Chemistry and Biochemistry, Queens College-CUNY, Flushing, New York 11367, USA
| | | | | |
Collapse
|