1
|
Braun JL, Fajardo VA. Spaceflight increases sarcoplasmic reticulum Ca 2+ leak and this cannot be counteracted with BuOE treatment. NPJ Microgravity 2024; 10:78. [PMID: 39030182 PMCID: PMC11271499 DOI: 10.1038/s41526-024-00419-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024] Open
Abstract
Spending time in a microgravity environment is known to cause significant skeletal muscle atrophy and weakness via muscle unloading, which can be partly attributed to Ca2+ dysregulation. The sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) pump is responsible for bringing Ca2+ from the cytosol into its storage site, the sarcoplasmic reticulum (SR), at the expense of ATP. We have recently demonstrated that, in the soleus of space-flown mice, the Ca2+ uptake ability of the SERCA pump is severely impaired and this may be attributed to increases in reactive oxygen/nitrogen species (RONS), to which SERCA is highly susceptible. The purpose of this study was therefore to investigate whether treatment with the antioxidant, Manganese(III) meso-tetrakis(N-n-butoxyethylpyridinium-2-yl)porphyrin, MnTnBuOE-2-PyP5+ (BuOE), could attenuate muscle atrophy and SERCA dysfunction. We received soleus muscles from the rodent research 18 mission which had male mice housed on the international space station for 35 days and treated with either saline or BuOE. Spaceflight significantly reduced the soleus:body mass ratio and significantly increased SERCA's ionophore ratio, a measure of SR Ca2+ leak, and 4-HNE content (marker of RONS), none of which could be rescued by BuOE treatment. In conclusion, we find that spaceflight induces significant soleus muscle atrophy and SR Ca2+ leak that cannot be counteracted with BuOE treatment. Future work should investigate alternative therapeutics that are specifically aimed at increasing SERCA activation or reducing Ca2+ leak.
Collapse
Affiliation(s)
- Jessica L Braun
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Val A Fajardo
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada.
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada.
| |
Collapse
|
2
|
Mao X, Stanbouly S, Holley J, Pecaut M, Crapo J. Evidence of Spaceflight-Induced Adverse Effects on Photoreceptors and Retinal Function in the Mouse Eye. Int J Mol Sci 2023; 24:ijms24087362. [PMID: 37108526 PMCID: PMC10138634 DOI: 10.3390/ijms24087362] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/06/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
The goal of the present study was to characterize acute oxidative damage in ocular structure and retinal function after exposure to spaceflight, and to evaluate the efficacy of an antioxidant in reducing spaceflight-induced changes in the retina. Ten-week-old adult C57BL/6 male mice were flown aboard the ISS on Space-X 24 over 35 days, and returned to Earth alive. The mice received a weekly injection of a superoxide dismutase mimic, MnTnBuOE-2-PyP 5+ (BuOE), before launch and during their stay onboard the ISS. Ground control mice were maintained on Earth under identical environmental conditions. Before the launch, intraocular pressure (IOP) was measured using a handheld tonometer and retinal function was evaluated using electroretinogram (ERG). ERG signals were recorded when the mouse eye was under dark-adapted conditions in response to ultraviolet monochromatic light flashes. Within 20 h after splashdown, IOP and ERG assessments were repeated before euthanasia. There were significant increases in body weight for habitat control groups post-flight compared to pre-flight measurements. However, the body weights were similar among flight groups before launch and after splashdown. The IOP measurements were similar between pre- and post-flight groups with no significant differences between BuOE-treated and saline controls. Immunofluorescence evaluation showed increases in retinal oxidative stress and apoptotic cell death after spaceflight. BuOE treatment significantly decreased the level of the oxidative stress biomarker. ERG data showed that the average amplitudes of the a- and b-wave were significantly decreased (39% and 32% by spaceflight, respectively) compared to that of habitat ground controls. These data indicate that spaceflight conditions induce oxidative stress in the retina, which may lead to photoreceptor cell damage and retinal function impairment.
Collapse
Affiliation(s)
- Xiaowen Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA 92350, USA
| | - Seta Stanbouly
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA 92350, USA
| | - Jacob Holley
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA 92350, USA
| | - Michael Pecaut
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA 92350, USA
| | - James Crapo
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, University of Colorado Denver, Denver, CO 80204, USA
| |
Collapse
|
3
|
Overbey EG, da Silveira WA, Stanbouly S, Nishiyama NC, Roque-Torres GD, Pecaut MJ, Zawieja DC, Wang C, Willey JS, Delp MD, Hardiman G, Mao XW. Spaceflight influences gene expression, photoreceptor integrity, and oxidative stress-related damage in the murine retina. Sci Rep 2019; 9:13304. [PMID: 31527661 PMCID: PMC6746706 DOI: 10.1038/s41598-019-49453-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/19/2019] [Indexed: 11/08/2022] Open
Abstract
Extended spaceflight has been shown to adversely affect astronaut visual acuity. The purpose of this study was to determine whether spaceflight alters gene expression profiles and induces oxidative damage in the retina. Ten week old adult C57BL/6 male mice were flown aboard the ISS for 35 days and returned to Earth alive. Ground control mice were maintained on Earth under identical environmental conditions. Within 38 (+/-4) hours after splashdown, mice ocular tissues were collected for analysis. RNA sequencing detected 600 differentially expressed genes (DEGs) in murine spaceflight retinas, which were enriched for genes related to visual perception, the phototransduction pathway, and numerous retina and photoreceptor phenotype categories. Twelve DEGs were associated with retinitis pigmentosa, characterized by dystrophy of the photoreceptor layer rods and cones. Differentially expressed transcription factors indicated changes in chromatin structure, offering clues to the observed phenotypic changes. Immunofluorescence assays showed degradation of cone photoreceptors and increased retinal oxidative stress. Total retinal, retinal pigment epithelium, and choroid layer thickness were significantly lower after spaceflight. These results indicate that retinal performance may decrease over extended periods of spaceflight and cause visual impairment.
Collapse
Affiliation(s)
- Eliah G Overbey
- University of Washington, Department of Genome Sciences, Seattle, WA, USA.
| | - Willian Abraham da Silveira
- Queen's University Belfast, Faculty of Medicine, Health and Life Sciences, School of Biological Sciences, Institute for Global Food Security (IGFS), 19 Chlorine Gardens, Belfast, Northern Ireland, BT9 5DL, UK
| | - Seta Stanbouly
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University, Loma Linda, CA, 92350, USA
- Center for Genomics, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Nina C Nishiyama
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University, Loma Linda, CA, 92350, USA
| | | | - Michael J Pecaut
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University, Loma Linda, CA, 92350, USA
| | - David Carl Zawieja
- Department of Medical Physiology, Texas A&M University, College Station, Texas, USA
| | - Charles Wang
- Center for Genomics, School of Medicine, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Jeffrey S Willey
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Michael D Delp
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, 32306, USA
| | - Gary Hardiman
- Queen's University Belfast, Faculty of Medicine, Health and Life Sciences, School of Biological Sciences, Institute for Global Food Security (IGFS), 19 Chlorine Gardens, Belfast, Northern Ireland, BT9 5DL, UK
| | - Xiao Wen Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University, Loma Linda, CA, 92350, USA
| |
Collapse
|
4
|
Kosmacek EA, Chatterjee A, Tong Q, Lin C, Oberley-Deegan RE. MnTnBuOE-2-PyP protects normal colorectal fibroblasts from radiation damage and simultaneously enhances radio/chemotherapeutic killing of colorectal cancer cells. Oncotarget 2018; 7:34532-45. [PMID: 27119354 PMCID: PMC5085174 DOI: 10.18632/oncotarget.8923] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 03/31/2016] [Indexed: 12/28/2022] Open
Abstract
Manganese porphyrins have been shown to be potent radioprotectors in a variety of cancer models. However, the mechanism as to how these porphyrins protect normal tissues from radiation damage still remains largely unknown. In the current study, we determine the effects of the manganese porphyrin, MnTnBuOE-2-PyP, on primary colorectal fibroblasts exposed to irradiation. We found that 2 Gy of radiation enhances the fibroblasts' ability to contract a collagen matrix, increases cell size and promotes cellular senesence. Treating fibroblasts with MnTnBuOE-2-PyP significantly inhibited radiation-induced collagen contraction, preserved cell morphology and also inhibited cellular senescence. We further showed that MnTnBuOE-2-PyP enhanced the overall viability of the fibroblasts following exposure to radiation but did not protect colorectal cancer cell viability. Specifically, MnTnBuOE-2-PyP in combination with irradiation, caused a significant decrease in tumor clonogenicity. Since locally advanced rectal cancers are treated with chemoradiation therapy followed by surgery and non-metastatic anal cancers are treated with chemoradiation therapy, we also investigated the effects of MnTnBuOE-2-PyP in combination with radiation, 5-fluorouracil with and without Mitomycin C. We found that MnTnBuOE-2-PyP in combination with Mitomycin C or 5-fluorouracil further enhances those compounds' ability to suppress tumor cell growth. When MnTnBuOE-2-PyP was combined with the two chemotherapeutics and radiation, we observed the greatest reduction in tumor cell growth. Therefore, these studies indicate that MnTnBuOE-2-PyP could be used as a potent radioprotector for normal tissue, while at the same time enhancing radiation and chemotherapy treatment for rectal and anal cancers.
Collapse
Affiliation(s)
- Elizabeth A Kosmacek
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Arpita Chatterjee
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Qiang Tong
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Chi Lin
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rebecca E Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
5
|
Kleiman NJ, Stewart FA, Hall EJ. Modifiers of radiation effects in the eye. LIFE SCIENCES IN SPACE RESEARCH 2017; 15:43-54. [PMID: 29198313 DOI: 10.1016/j.lssr.2017.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 07/05/2017] [Accepted: 07/17/2017] [Indexed: 06/07/2023]
Abstract
World events, including the threat of radiological terrorism and the fear of nuclear accidents, have highlighted an urgent need to develop medical countermeasures to prevent or reduce radiation injury. Similarly, plans for manned spaceflight to a near-Earth asteroid or journey to Mars raise serious concerns about long-term effects of space radiation on human health and the availability of suitable therapeutic interventions. At the same time, the need to protect normal tissue from the deleterious effects of radiotherapy has driven considerable research into the design of effective radioprotectors. For more than 70 years, animal models of radiation cataract have been utilized to test the short and long-term efficacy of various radiation countermeasures. While some compounds, most notably the Walter Reed (WR) class of radioprotectors, have reported limited effectiveness when given before exposure to low-LET radiation, the human toxicity of these molecules at effective doses limits their usefulness. Furthermore, while there has been considerable testing of eye responses to X- and gamma irradiation, there is limited information about using such models to limit the injurious effects of heavy ions and neutrons on eye tissue. A new class of radioprotector molecules, including the sulfhydryl compound PrC-210, are reported to be effective at much lower doses and with far less side effects. Their ability to modify ocular radiation damage has not yet been examined. The ability to non-invasively measure sensitive, radiation-induced ocular changes over long periods of time makes eye models an attractive option to test the radioprotective and radiation mitigating abilities of new novel compounds.
Collapse
Affiliation(s)
- Norman J Kleiman
- Department of Environmental Health Sciences, Eye Radiation and Environmental Research Laboratory, Columbia University, Mailman School of Public Health, 722 West 168th St., 11th Floor, New York, NY 10032, USA.
| | - Fiona A Stewart
- Division of Biological Stress Response, Netherlands Cancer Institute, 1006 BE Amsterdam, The Netherlands
| | - Eric J Hall
- Center for Radiological Research, Columbia University, College of Physicians and Surgeons, 630 W. 168th St., New York, NY 10032, USA
| |
Collapse
|
6
|
Slosky LM, Vanderah TW. Therapeutic potential of peroxynitrite decomposition catalysts: a patent review. Expert Opin Ther Pat 2015; 25:443-66. [PMID: 25576197 DOI: 10.1517/13543776.2014.1000862] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Peroxynitrite is a cytotoxic oxidant species implicated in a host of pathologies, including inflammatory and neurodegenerative diseases, cancer, radiation injury and chronic pain. With the recognition of the role of peroxynitrite in disease, numerous experimental and therapeutic tools have arisen to probe peroxyntirite's pathophysiological contribution and attenuate its oxidative damage. Peroxynitrite decomposition catalysts (PNDCs) are redox-active compounds that detoxify peroxynitrite by catalyzing its isomerization or reduction to nitrate or nitrite. AREAS COVERED This review discusses recent research articles and patents published 1995 - 2014 on the development and therapeutic use of PNDCs. Iron and manganese metalloporphyrin PNDCs attenuate the toxic effects of peroxynitrite and are currently being developed for clinical applications. Additionally, some Mn porphyrin-based PNDCs have optimized pharmaceutical properties such that they exhibit greater peroxynitrite selectivity. Other classes of PNDC agents, including bis(hydroxyphenyl)dipyrromethenes and metallocorroles, have demonstrated preclinical efficacy, oral availability and reduced toxicity risk. EXPERT OPINION Interest in the drug-like properties of peroxynitrite-neutralizing agents has grown with the realization that PNDCs will be powerful tools in the treatment of disease. The design of compounds with enhanced oral availability and peroxynitrite selectivity is a critical step toward the availability of safe, effective and selective redox modulators for the treatment of peroxynitrite-associated pathologies.
Collapse
Affiliation(s)
- Lauren M Slosky
- University of Arizona, Department of Pharmacology , Life Science North Rm 621, 1501 North Campbell Ave., Tucson, AZ 85721 , USA
| | | |
Collapse
|
7
|
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
|
8
|
Archambeau JO, Tovmasyan A, Pearlstein RD, Crapo JD, Batinic-Haberle I. Superoxide dismutase mimic, MnTE-2-PyP(5+) ameliorates acute and chronic proctitis following focal proton irradiation of the rat rectum. Redox Biol 2013; 1:599-607. [PMID: 24363995 PMCID: PMC3863774 DOI: 10.1016/j.redox.2013.10.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 10/12/2013] [Accepted: 10/14/2013] [Indexed: 01/05/2023] Open
Abstract
Radiation proctitis, an inflammation and damage to the lower part of colon, is a common adverse event of the radiotherapy of tumors in the abdominal and pelvic region (colon, prostate, cervical). Several Mn(III) porphyrin-based superoxide dismutase mimics have been synthesized and successfully evaluated in preclinical models as radioprotectants. Here we report for the first time the remarkable rectal radioprotection of frequently explored Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin, MnTE-2-PyP(5+). A batch prepared in compliance with good manufacturing practice (GMP), which has good safety/toxicity profile, was used for this study. MnTE-2-PyP(5+) was given subcutaneously at 5 mg/kg, either 1 h before or 1 h after irradiation, with additional drug administered at weekly intervals thereafter. MnTE-2-PyP(5+) ameliorated both acute and chronic radiation proctitis in male Sprague-Dawley rats irradiated with 20-30 Gy protons delivered to 2.5 cm span of rectum using spread-out Bragg peak of a proton treatment beam. Focal irradiation of the rectum produced acute proctitis, which healed, followed by chronic rectal dilation and symptomatic proctitis. MnTE-2-PyP(5+) protected rectal mucosa from radiation-induced crypt loss measured 10 days post-irradiation. Significant effects were observed with both pre- and post-treatment regimens. However, only MnTE-2-PyP(5+) pre-treatment, but not post-treatment, prevented the development of rectal dilation, indicating that proper dosing regimen is critical for radioprotection. The pre-treatment also prevented or delayed the development of chronic proctitis depending on the radiation dose. Further work aimed at developing MnTE-2-PyP(5+) and similar drugs as adjunctive agents for radiotherapy of pelvic tumors is warranted. The present study substantiates the prospects of employing this and similar analogs in preserving normal tissue during cancer radiation as well as any other radiation exposure.
Collapse
Key Words
- AP-1, activator protein-1
- CGE, cobalt gray equivalent
- GSH, glutathione
- HIF-1α, hypoxia inducible factor-1
- Mn porphyrin
- MnP, Mn(III) porphyrins
- MnTDE-2-ImP5+, Mn(III) meso-tetrakis(N,N’-diethylimidazolium-2-yl)porphyrin (AEOL10150)
- MnTE-2-PyP5+
- MnTE-2-PyP5+, Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin (AEOL10113, BMX-010)
- MnTM-2-PyP5+, Mn(III) meso-tetrakis(N-methylpyridinium-2-yl)porphyrin (AEOL10112)
- MnTnBuOE-2-PyP5+, Mn(III) meso-tetrakis(N-n-butoxyethylpyridinium-2-yl)porphyrin
- MnTnHex-2-PyP5+, Mn(III) meso-tetrakis(N-n-hexylpyridinium-2-yl)porphyrin (BMX-001)
- NF-κB, nuclear factor κB
- PT, proton therapy
- Proton beam therapy
- Radiation proctitis
- Radioprotector
- SOD mimic
- SOD, superoxide dismutase
- SP-1, specificity protein-1
- TF, transcription factor
- kcat(O2−), the rate constant for the catalysis of O2− dismutation by Mn porphyrin or SOD enzyme
Collapse
Affiliation(s)
- John O Archambeau
- Department of Radiation Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Robert D Pearlstein
- Department of Surgery (Neurosurgery), Duke University School of Medicine, Durham, NC 27710, USA
| | - James D Crapo
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Denver, CO 80206, USA
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
9
|
Tumor-related Lipid Exudation and Associated Tumor-related Complications after Plaque Radiotherapy of Posterior Uveal Melanoma. Eur J Ophthalmol 2013; 23:399-409. [DOI: 10.5301/ejo.5000236] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2012] [Indexed: 11/20/2022]
|
10
|
Ozgen SÇ, Dökmeci D, Akpolat M, Karadağ CH, Gündüz O, Erbaş H, Benian O, Uzal C, Turan FN. The Protective Effect of Curcumin on Ionizing Radiation-induced Cataractogenesis in Rats. Balkan Med J 2012; 29:358-63. [PMID: 25207034 DOI: 10.5152/balkanmedj.2012.038] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 05/22/2012] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE The aim of the study was to determine the protective effect of curcumin against ionizing radiation-induced cataract in the lens of rats. MATERIAL AND METHODS Rats were divided into six groups. Group 1: Control, Group 2: Dimethyl sulfoxide (DMSO), Group 3: DMSO+curcumin, Group 4: Irradiation, Group 5: Irradiation+DMSO, Group 6: Irradiation+DMSO+curcumin. A 15 Gy total dose was given to 4, 5, 6 groups for radiation damage. Curcumin (100 mg/kg) was dissolved in DMSO and given by intragastric intubation for 28 days. At the end of the experiment, lenses were graded and enucleated. The lenticular activity of the antioxidant enzymes, total antioxidant and glutathione peroxidase (GSH-Px), and the malondialdehyde (MDA) were measured. RESULTS 100% Cataract was seen in the irradiation group. Cataract rate fell to 40% and was limited at grade 1 and 2 in the curcumin group. In the irradiation group, antioxidant enzyme levels were decreased, MDA levels were increased. There was an increase in antioxidant enzyme levels and a significant decrease in MDA in the group which was given curcumin. CONCLUSION Curcumin has antioxidant and radioprotective properties and is likely to be a valuable agent for protection against ionizing radiation. Hence, it may be used as an antioxidant and radioprotector against radiation-induced cataractogenesis.
Collapse
Affiliation(s)
- Seher Çimen Ozgen
- Department of Pharmacology, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Dikmen Dökmeci
- Department of Pharmacology, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Meryem Akpolat
- Department of Histology and Embryology, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Cetin Hakan Karadağ
- Department of Pharmacology, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Ozgür Gündüz
- Department of Pharmacology, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Hakan Erbaş
- Department of Biochemistry, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Omer Benian
- Department of Ophtalmology, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Cem Uzal
- Department of Radiation Oncology, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Fatma Nesrin Turan
- Department of Biostatistics, Faculty of Medicine, Trakya University, Edirne, Turkey
| |
Collapse
|
11
|
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
|
12
|
Oberley-Deegan RE, Steffan JJ, Rove KO, Pate KM, Weaver MW, Spasojevic I, Frederick B, Raben D, Meacham RB, Crapo JD, Koul HK. The antioxidant, MnTE-2-PyP, prevents side-effects incurred by prostate cancer irradiation. PLoS One 2012; 7:e44178. [PMID: 22984473 PMCID: PMC3440381 DOI: 10.1371/journal.pone.0044178] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 08/02/2012] [Indexed: 11/18/2022] Open
Abstract
Prostate cancer is the most commonly diagnosed cancer, with an estimated 240,000 new cases reported annually in the United States. Due to early detection and advances in therapies, more than 90% of patients will survive 10 years post diagnosis and treatment. Radiation is a treatment option often used to treat localized disease; however, while radiation is very effective at killing tumor cells, normal tissues are damaged as well. Potential side-effects due to prostate cancer-related radiation therapy include bowel inflammation, erectile dysfunction, urethral stricture, rectal bleeding and incontinence. Currently, radiation therapy for prostate cancer does not include the administration of therapeutic agents to reduce these side effects and protect normal tissues from radiation-induced damage. In the current study, we show that the small molecular weight antioxidant, MnTE-2-PyP, protects normal tissues from radiation-induced damage in the lower abdomen in rats. Specifically, MnTE-2-PyP protected skin, prostate, and testes from radiation-induced damage. MnTE-2-PyP also protected from erectile dysfunction, a persistent problem regardless of the type of radiation techniques used because the penile neurovascular bundles lay in the peripheral zones of the prostate, where most prostate cancers reside. Based on previous studies showing that MnTE-2-PyP, in combination with radiation, further reduces subcutaneous tumor growth, we believe that MnTE-2-PyP represents an excellent radioprotectant in combination radiotherapy for cancer in general and specifically for prostate cancer.
Collapse
Affiliation(s)
- Rebecca E Oberley-Deegan
- Pulmonary Division, Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Mao XW, Crapo JD, Gridley DS. Mitochondrial Oxidative Stress-Induced Apoptosis and Radioprotection in Proton-Irradiated Rat Retina. Radiat Res 2012; 178:118-25. [DOI: 10.1667/rr2821.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
14
|
Mehrotra S, Pecaut MJ, Freeman TL, Crapo JD, Rizvi A, Luo-Owen X, Slater JM, Gridley DS. Analysis of a metalloporphyrin antioxidant mimetic (MnTE-2-PyP) as a radiomitigator: prostate tumor and immune status. Technol Cancer Res Treat 2012; 11:447-57. [PMID: 22475066 DOI: 10.7785/tcrt.2012.500260] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Due to radiation-induced immune depression and development of pathologies such as cancer, there is increasing urgency to identify radiomitigators that are effective when administered after radiation exposure. The main goal of this study was to determine the radiomitigation capacity of MnTE-2-PyP[Mn(III) tetrakis (N-ethylpyridinium-2-yl) porphyrin], a superoxide dismutase (SOD) mimetic, and evaluate leukocyte parameters in spleen and blood. C57BL/6 mice were total-body exposed to 2 Gy γ-rays (Co-60), i.e., well below a lethal dose, followed by subcutaneous implantation of 5 × 10(5) RM-9 prostate tumor cells and initiation of MnTE-2-PyP treatment (day 0); interval between each procedure was 1-2 h. The drug was administered daily (12 times). Tumor progression was monitored and immunological analyses were performed on a subset per group on day 12. Animals treated with MnTE-2-PyP alone had significantly slower tumor growth compared to mice that did not receive the drug (P < 0.05), while radiation alone had no effect. Treatment of tumor-bearing mice with MnTE-2-PyP alone significantly increased spleen mass relative to body mass; the numbers of splenic white blood cells (WBC) and lymphocytes (B and T), as well as circulating WBC, granulocytes, and platelets, were high compared to one of more of the other groups (P < 0.05). The results show that MnTE-2-PyP slowed RM-9 tumor progression and up-regulated immune parameters, but mitigation of the effects of 2 Gy total-body irradiation were minimal.
Collapse
Affiliation(s)
- S Mehrotra
- Department of Basic Sciences, Divisions of Biochemistry and Microbiology, Loma Linda University and Medical Center, Loma Linda, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Stewart FA, Akleyev AV, Hauer-Jensen M, Hendry JH, Kleiman NJ, Macvittie TJ, Aleman BM, Edgar AB, Mabuchi K, Muirhead CR, Shore RE, Wallace WH. ICRP publication 118: ICRP statement on tissue reactions and early and late effects of radiation in normal tissues and organs--threshold doses for tissue reactions in a radiation protection context. Ann ICRP 2012; 41:1-322. [PMID: 22925378 DOI: 10.1016/j.icrp.2012.02.001] [Citation(s) in RCA: 853] [Impact Index Per Article: 65.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
This report provides a review of early and late effects of radiation in normal tissues and organs with respect to radiation protection. It was instigated following a recommendation in Publication 103 (ICRP, 2007), and it provides updated estimates of 'practical' threshold doses for tissue injury defined at the level of 1% incidence. Estimates are given for morbidity and mortality endpoints in all organ systems following acute, fractionated, or chronic exposure. The organ systems comprise the haematopoietic, immune, reproductive, circulatory, respiratory, musculoskeletal, endocrine, and nervous systems; the digestive and urinary tracts; the skin; and the eye. Particular attention is paid to circulatory disease and cataracts because of recent evidence of higher incidences of injury than expected after lower doses; hence, threshold doses appear to be lower than previously considered. This is largely because of the increasing incidences with increasing times after exposure. In the context of protection, it is the threshold doses for very long follow-up times that are the most relevant for workers and the public; for example, the atomic bomb survivors with 40-50years of follow-up. Radiotherapy data generally apply for shorter follow-up times because of competing causes of death in cancer patients, and hence the risks of radiation-induced circulatory disease at those earlier times are lower. A variety of biological response modifiers have been used to help reduce late reactions in many tissues. These include antioxidants, radical scavengers, inhibitors of apoptosis, anti-inflammatory drugs, angiotensin-converting enzyme inhibitors, growth factors, and cytokines. In many cases, these give dose modification factors of 1.1-1.2, and in a few cases 1.5-2, indicating the potential for increasing threshold doses in known exposure cases. In contrast, there are agents that enhance radiation responses, notably other cytotoxic agents such as antimetabolites, alkylating agents, anti-angiogenic drugs, and antibiotics, as well as genetic and comorbidity factors. Most tissues show a sparing effect of dose fractionation, so that total doses for a given endpoint are higher if the dose is fractionated rather than when given as a single dose. However, for reactions manifesting very late after low total doses, particularly for cataracts and circulatory disease, it appears that the rate of dose delivery does not modify the low incidence. This implies that the injury in these cases and at these low dose levels is caused by single-hit irreparable-type events. For these two tissues, a threshold dose of 0.5Gy is proposed herein for practical purposes, irrespective of the rate of dose delivery, and future studies may elucidate this judgement further.
Collapse
|
16
|
Batinic-Haberle I, Rajic Z, Tovmasyan A, Ye X, Leong KW, Dewhirst MW, Vujaskovic Z, Benov L, Spasojevic I. Diverse functions of cationic Mn(III) N-substituted pyridylporphyrins, recognized as SOD mimics. Free Radic Biol Med 2011; 51:1035-53. [PMID: 21616142 PMCID: PMC3178885 DOI: 10.1016/j.freeradbiomed.2011.04.046] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 03/30/2011] [Accepted: 04/29/2011] [Indexed: 10/18/2022]
Abstract
Oxidative stress, a redox imbalance between the endogenous reactive species and antioxidant systems, is common to numerous pathological conditions such as cancer, central nervous system injuries, radiation injury, diabetes etc. Therefore, compounds able to reduce oxidative stress have been actively sought for over 3 decades. Superoxide is the major species involved in oxidative stress either in its own right or through its progeny, such as ONOO⁻, H₂O₂, •OH, CO₃•⁻, and •NO₂. Hence, the very first compounds developed in the late 1970-ies were the superoxide dismutase (SOD) mimics. Thus far the most potent mimics have been the cationic meso Mn(III) N-substituted pyridylporphyrins and N,N'-disubstituted imidazolylporphyrins (MnPs), some of them with k(cat)(O₂·⁻) similar to the k(cat) of SOD enzymes. Most frequently studied are ortho isomers MnTE-2-PyP⁵⁺, MnTnHex-2-PyP⁵⁺, and MnTDE-2-ImP⁵⁺. The ability to disproportionate O₂·⁻ parallels their ability to remove the other major oxidizing species, peroxynitrite, ONOO⁻. The same structural feature that gives rise to the high k(cat)(O₂·⁻) and k(red)(ONOO⁻), allows MnPs to strongly impact the activation of the redox-sensitive transcription factors, HIF-1α, NF-κB, AP-1, and SP-1, and therefore modify the excessive inflammatory and immune responses. Coupling with cellular reductants and other redox-active endogenous proteins seems to be involved in the actions of Mn porphyrins. While hydrophilic analogues, such as MnTE-2-PyP⁵⁺ and MnTDE-2-ImP⁵⁺ are potent in numerous animal models of diseases, the lipophilic analogues, such as MnTnHex-2-PyP⁵⁺, were developed to cross blood brain barrier and target central nervous system and critical cellular compartments, mitochondria. The modification of its structure, aimed to preserve the SOD-like potency and lipophilicity, and diminish the toxicity, has presently been pursued. The pulmonary radioprotection by MnTnHex-2-PyP⁵⁺ was the first efficacy study performed successfully with non-human primates. The Phase I toxicity clinical trials were done on amyotrophic lateral sclerosis patients with N,N'-diethylimidazolium analogue, MnTDE-2-ImP⁵⁺ (AEOL10150). Its aggressive development as a wide spectrum radioprotector by Aeolus Pharmaceuticals has been supported by USA Federal government. The latest generation of compounds, bearing oxygens in pyridyl substituents is presently under aggressive development for cancer and CNS injuries at Duke University and is supported by Duke Translational Research Institute, The Wallace H. Coulter Translational Partners Grant Program, Preston Robert Tisch Brain Tumor Center at Duke, and National Institute of Allergy and Infectious Diseases. Metal center of cationic MnPs easily accepts and donates electrons as exemplified in the catalysis of O₂·⁻ dismutation. Thus such compounds may be equally good anti- and pro-oxidants; in either case the beneficial therapeutic effects may be observed. Moreover, while the in vivo effects may appear antioxidative, the mechanism of action of MnPs that produced such effects may be pro-oxidative; the most obvious example being the inhibition of NF-κB. The experimental data therefore teach us that we need to distinguish between the mechanism/s of action/s of MnPs and the effects we observe. A number of factors impact the type of action of MnPs leading to favorable therapeutic effects: levels of reactive species and oxygen, levels of endogenous antioxidants (enzymes and low-molecular compounds), levels of MnPs, their site of accumulation, and the mutual encounters of all of those species. The complexity of in vivo redox systems and the complex redox chemistry of MnPs challenge and motivate us to further our understanding of the physiology of the normal and diseased cell with ultimate goal to successfully treat human diseases.
Collapse
Affiliation(s)
- Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
- Corresponding authors: Ines Batinic-Haberle, Ph. D. Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, Tel: 919-684-2101, Fax: 919-684-8718, . Ivan Spasojevic, Ph. D. Department of Medicine, Duke University Medical Center, Durham, NC 27710, Tel: 919-684-8311, Fax: 919-684-8380,
| | - Zrinka Rajic
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Artak Tovmasyan
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Xiaodong Ye
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Kam W. Leong
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Mark W. Dewhirst
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Zeljko Vujaskovic
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ludmil Benov
- Department of Biochemistry, Faculty of Medicine, Kuwait School of Medicine, Kuwait
| | - Ivan Spasojevic
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- Corresponding authors: Ines Batinic-Haberle, Ph. D. Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, Tel: 919-684-2101, Fax: 919-684-8718, . Ivan Spasojevic, Ph. D. Department of Medicine, Duke University Medical Center, Durham, NC 27710, Tel: 919-684-8311, Fax: 919-684-8380,
| |
Collapse
|
17
|
Li H, Wang Y, Pazhanisamy SK, Shao L, Batinic-Haberle I, Meng A, Zhou D. Mn(III) meso-tetrakis-(N-ethylpyridinium-2-yl) porphyrin mitigates total body irradiation-induced long-term bone marrow suppression. Free Radic Biol Med 2011; 51:30-7. [PMID: 21565268 PMCID: PMC3390209 DOI: 10.1016/j.freeradbiomed.2011.04.016] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2010] [Revised: 04/05/2011] [Accepted: 04/08/2011] [Indexed: 12/28/2022]
Abstract
Our recent studies showed that total body irradiation (TBI) induces long-term bone marrow (BM) suppression in part by induction of hematopoietic stem cell (HSC) senescence through reactive oxygen species (ROS). In this study, we examined if Mn(III) meso-tetrakis-(N-ethylpyridinium-2-yl) porphyrin (MnTE), a superoxide dismutase mimetic and potent antioxidant, can mitigate TBI-induced long-term BM injury in a mouse model. Our results showed that post-TBI treatment with MnTE significantly inhibited the increases in ROS production and DNA damage in HSCs and the reduction in HSC frequency and clonogenic function induced by TBI. In fact, the clonogenic function of HSCs from irradiated mice after MnTE treatment was comparable to that of HSCs from normal controls on a per-HSC basis, suggesting that MnTE treatment inhibited the induction of HSC senescence by TBI. This suggestion is supported by the finding that MnTE treatment also reduced the expression of p16(Ink4a) (p16) mRNA in HSCs induced by TBI and improved the long-term and multilineage engraftment of irradiated HSCs after transplantation. Therefore, the results from this study demonstrate that MnTE has the potential to be used as a therapeutic agent to mitigate TBI-induced long-term BM suppression by inhibiting ionizing radiation-induced HSC senescence through the ROS-p16 pathway.
Collapse
Affiliation(s)
- Hongliang Li
- Department of Biochemistry and Molecular Biology, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Pathology, Medical University of South Carolina, Charleston, SC
- Division of Radiation Health, Department of Pharmaceutical Sciences and Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Yong Wang
- Department of Pathology, Medical University of South Carolina, Charleston, SC
| | - Senthil K. Pazhanisamy
- Department of Pathology, Medical University of South Carolina, Charleston, SC
- Division of Radiation Health, Department of Pharmaceutical Sciences and Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Lijian Shao
- Department of Pathology, Medical University of South Carolina, Charleston, SC
- Division of Radiation Health, Department of Pharmaceutical Sciences and Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR
| | | | - Aimin Meng
- Department of Biochemistry and Molecular Biology, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Daohong Zhou
- Department of Pathology, Medical University of South Carolina, Charleston, SC
- Division of Radiation Health, Department of Pharmaceutical Sciences and Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR
- Corresponding author: Daohong Zhou, MD, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W Markham, #607, Little Rock, AR 72205; Tel: (501) 526-5272; Fax: (501) 686-6517;
| |
Collapse
|
18
|
Keir ST, Dewhirst MW, Kirkpatrick JP, Bigner DD, Batinic-Haberle I. Cellular redox modulator, ortho Mn(III) meso-tetrakis(N-n-hexylpyridinium-2-yl)porphyrin, MnTnHex-2-PyP(5+) in the treatment of brain tumors. Anticancer Agents Med Chem 2011; 11:202-12. [PMID: 21291403 PMCID: PMC3357315 DOI: 10.2174/187152011795255957] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2010] [Accepted: 02/04/2011] [Indexed: 01/11/2023]
Abstract
Despite intensive efforts to improve multimodal treatment of brain tumor, survival remains limited. Current therapy consists of a combination of surgery, irradiation and chemotherapy with predisposition to long-term complications. Identifying novel targeted therapies is therefore at the forefront of brain tumor research. This study explores the utility of a manganese porphyrin in a brain tumor model. The compound used is ortho isomer, mangnese(III) meso-tetrakis(N-n-hexylpyridinium-2-yl)porphyrin, MnTnHex-2-PyP(5+). It is a powerful SOD mimic and peroxynitrite scavenger and a potent modulator of redox-based cellular transcriptional activity, able to suppress excessive immune and inflammatory responses and in turn proliferative pathways. It is further one of the most lipophilic compound among cationic Mn(III) N-alkylpyridylporphyrins, and thus accumulates predominantly in mitochondria relative to cytosol. In mitochondria, MnTnHex-2-PyP(5+) mimics our key antioxidant system, mitochondrial superoxide dismutase, MnSOD, whose overexpression has been widely shown to suppress tumor growth. Importantly, MnTnHex-2-PyP(5+) crosses blood brain barrier in sufficient amounts to demonstrate efficacy in treating CNS injuries. For those reasons we elected to test its effects in inhibiting brain tumor growth. This study is the first report of the antitumor properties of MnTnHex-2-PyP(5+) as a single agent in adult and pediatric glioblastoma multiforme (D-54 MG, D-245 MG, D-256 MG, D-456 MG) and pediatric medulloblastoma (D-341 MED), and is the first case where a redox-able metal complex has been used in glioma therapy. When given subcutaneously to mice bearing subcutaneous and intracranial xenografts, MnTnHex-2-PyP(5+) caused a significant (P ≤ 0.001) growth delay in D 245 MG, D-256 MG, D-341 MED, and D-456 MG tumors. Growth delay for mice bearing subcutaneous xenografts ranged from 3 days in D-54 MG to 34 days in D-341 MED. With mice bearing intracranial xenografts, MnTnHex-2-PyP(5+) increases median survival by 33% in adult glioblastoma multiforme (D-256 MG; p≤ 0.001) and 173% in pediatric medulloblastoma (D-341 MED, <0.001). The beneficial effects of MnTnHex-2-PyP(5+) are presumably achieved either (1) indirectly via elimination of signaling reactive oxygen and nitrogen species (in particular superoxide and peroxynitrite) which in turn would prevent activation of transcription factors; or (2) directly by coupling with cellular reductants and redox-sensitive signaling proteins. The former action is antioxidative while the latter action is presumably pro-oxidative in nature. Our findings suggest that the use of Mn porphyrin-based SOD mimics, and in particular lipophilic analogues such as MnTnHex-2-PyP(5+), is a promising approach for brain tumor therapy.
Collapse
Affiliation(s)
- Stephen T. Keir
- The Preston Robert Tisch Brain Tumor Center at Duke, Duke University, Durham NC, 27710, USA
- Department of Surgery, Duke University, Durham NC, 27710, USA
| | - Mark W. Dewhirst
- Department of Radiation Oncology, Duke University, Durham NC, 27710, USA
| | | | - Darell D. Bigner
- The Preston Robert Tisch Brain Tumor Center at Duke, Duke University, Durham NC, 27710, USA
- Department of Pathology, Duke University, Durham NC, 27710, USA
| | | |
Collapse
|
19
|
Batinić-Haberle I, Rebouças JS, Spasojević I. Superoxide dismutase mimics: chemistry, pharmacology, and therapeutic potential. Antioxid Redox Signal 2010; 13:877-918. [PMID: 20095865 PMCID: PMC2935339 DOI: 10.1089/ars.2009.2876] [Citation(s) in RCA: 407] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Oxidative stress has become widely viewed as an underlying condition in a number of diseases, such as ischemia-reperfusion disorders, central nervous system disorders, cardiovascular conditions, cancer, and diabetes. Thus, natural and synthetic antioxidants have been actively sought. Superoxide dismutase is a first line of defense against oxidative stress under physiological and pathological conditions. Therefore, the development of therapeutics aimed at mimicking superoxide dismutase was a natural maneuver. Metalloporphyrins, as well as Mn cyclic polyamines, Mn salen derivatives and nitroxides were all originally developed as SOD mimics. The same thermodynamic and electrostatic properties that make them potent SOD mimics may allow them to reduce other reactive species such as peroxynitrite, peroxynitrite-derived CO(3)(*-), peroxyl radical, and less efficiently H(2)O(2). By doing so SOD mimics can decrease both primary and secondary oxidative events, the latter arising from the inhibition of cellular transcriptional activity. To better judge the therapeutic potential and the advantage of one over the other type of compound, comparative studies of different classes of drugs in the same cellular and/or animal models are needed. We here provide a comprehensive overview of the chemical properties and some in vivo effects observed with various classes of compounds with a special emphasis on porphyrin-based compounds.
Collapse
Affiliation(s)
- Ines Batinić-Haberle
- Department of Radiation Oncology, Duke University Medical School, Durham, North Carolina 27710, USA.
| | | | | |
Collapse
|
20
|
Gridley DS, Grover RS, Loredo LN, Wroe AJ, Slater JD. Proton-beam therapy for tumors of the CNS. Expert Rev Neurother 2010; 10:319-30. [PMID: 20136386 DOI: 10.1586/ern.09.150] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The focus of this review is proton radiotherapy for primary neoplasms of the brain. Although glial cells are among the most radioresistant in the body, the presence of sensitive critical structures and the high doses needed to control CNS tumors present a formidable challenge to the treating radiation oncologist. Treatment with conventional photon radiation at doses required to control disease progression all too often results in unacceptable toxicity. Protons have intrinsic properties that often allow radiation oncologists to deliver a higher dose to the tumor compared with photons, while at the same time offering better sparing of normal tissues. Recognition of these advantages has resulted in development of many new proton treatment facilities worldwide.
Collapse
Affiliation(s)
- Daila S Gridley
- Department of Radiation Medicine, Chan Shun Pavilion, 11175 Campus Street, Loma Linda University, Loma Linda, CA 92354, USA.
| | | | | | | | | |
Collapse
|