1
|
Lotfy M, Khattab A, Shata M, Alhasbani A, Almesmari A, Alsaeedi S, Alyassi S, Kundu B. Destructive effects of UVC radiation on Drosophila melanogaster: Mortality, fertility, mutations, and molecular mechanisms. PLoS One 2024; 19:e0303115. [PMID: 38776353 PMCID: PMC11111075 DOI: 10.1371/journal.pone.0303115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 04/09/2024] [Indexed: 05/24/2024] Open
Abstract
The detrimental effects of ultraviolet C (UVC) radiation on living organisms, with a specific focus on the fruit fly Drosophila melanogaster, were examined. This study investigated the impact of heightened UVC radiation exposure on D. melanogaster by assessing mortality and fertility rates, studying phenotypic mutations, and investigating the associated molecular mechanisms. The findings of this study revealed that UVC radiation increases mortality rates and decreases fertility rates in D. melanogaster. Additionally, phenotypic wing mutations were observed in the exposed flies. Furthermore, the study demonstrated that UVC radiation downregulates the expression of antioxidant genes, including superoxide dismutase (SOD), manganese-dependent superoxide dismutase (Mn-SOD), zinc-dependent superoxide dismutase (Cu-Zn-SOD), and the G protein-coupled receptor methuselah (MTH) gene. These results suggest that UVC radiation exerts a destructive effect on D. melanogaster by inducing oxidative stress, which is marked by the overexpression of harmful oxidative processes and a simultaneous reduction in antioxidant gene expression. In conclusion, this study underscores the critical importance of comprehending the deleterious effects of UVC radiation, not only to safeguard human health on Earth, but also to address the potential risks associated with space missions, such as the ongoing Emirate astronaut program.
Collapse
Affiliation(s)
- Mohamed Lotfy
- Biology Department, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Aalaa Khattab
- Faculty of Dentistry, The British University in Egypt, El Sherouk City, Cairo, Egypt
| | - Mohammed Shata
- Biology Department, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ahmad Alhasbani
- Biology Department, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Abdulla Almesmari
- Biology Department, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Saeed Alsaeedi
- Biology Department, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Saeed Alyassi
- Biology Department, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Biduth Kundu
- Biology Department, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
2
|
Dobney W, Mols L, Mistry D, Tabury K, Baselet B, Baatout S. Evaluation of deep space exploration risks and mitigations against radiation and microgravity. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2023; 3:1225034. [PMID: 39355042 PMCID: PMC11440958 DOI: 10.3389/fnume.2023.1225034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/04/2023] [Indexed: 10/03/2024]
Abstract
Ionizing radiation and microgravity are two considerable health risks encountered during deep space exploration. Both have deleterious effects on the human body. On one hand, weightlessness is known to induce a weakening of the immune system, delayed wound healing and musculoskeletal, cardiovascular, and sensorimotor deconditioning. On the other hand, radiation exposure can lead to long-term health effects such as cancer and cataracts as well as have an adverse effect on the central nervous and cardiovascular systems. Ionizing radiation originates from three main sources in space: galactic cosmic radiation, solar particle events and solar winds. Furthermore, inside the spacecraft and inside certain space habitats on Lunar and Martian surfaces, the crew is exposed to intravehicular radiation, which arises from nuclear reactions between space radiation and matter. Besides the approaches already in use, such as radiation shielding materials (such as aluminium, water or polyethylene), alternative shielding materials (including boron nanotubes, complex hybrids, composite hybrid materials, and regolith) and active shielding (using fields to deflect radiation particles) are being investigated for their abilities to mitigate the effects of ionizing radiation. From a biological point of view, it can be predicted that exposure to ionizing radiation during missions beyond Low Earth Orbit (LEO) will affect the human body in undesirable ways, e.g., increasing the risks of cataracts, cardiovascular and central nervous system diseases, carcinogenesis, as well as accelerated ageing. Therefore, it is necessary to assess the risks related to deep space exploration and to develop mitigation strategies to reduce these risks to a tolerable level. By using biomarkers for radiation sensitivity, space agencies are developing extensive personalised medical examination programmes to determine an astronaut's vulnerability to radiation. Moreover, researchers are developing pharmacological solutions (e.g., radioprotectors and radiomitigators) to proactively or reactively protect astronauts during deep space exploration. Finally, research is necessary to develop more effective countermeasures for use in future human space missions, which can also lead to improvements to medical care on Earth. This review will discuss the risks space travel beyond LEO poses to astronauts, methods to monitor astronauts' health, and possible approaches to mitigate these risks.
Collapse
Affiliation(s)
- William Dobney
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- School of Aeronautical, Automotive, Chemical and Materials Engineering, Loughborough University, Loughborough, United Kingdom
| | - Louise Mols
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Department of Physics and Astronomy, KU Leuven, Leuven, Belgium
| | - Dhruti Mistry
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Kevin Tabury
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Department of Biomedical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, SC, United States
| | - Bjorn Baselet
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Department of Physics and Astronomy, KU Leuven, Leuven, Belgium
- Department of Molecular Biotechnology, UGhent, Gent, Belgium
- Department of Human Structure & Repair, UGhent, Gent, Belgium
| |
Collapse
|
3
|
Radioresistance in Prostate Cancer: Focus on the Interplay between NF-κB and SOD. Antioxidants (Basel) 2021; 10:antiox10121925. [PMID: 34943029 PMCID: PMC8750009 DOI: 10.3390/antiox10121925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/15/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022] Open
Abstract
Prostate cancer occurs frequently in men and can often lead to death. Many cancers, including prostate cancer, can be initiated by oxidative insult caused by free radicals and reactive oxygen species. The superoxide dismutase family removes the oxygen-derived reactive oxygen species, and increased superoxide dismutase activity can often be protective against prostate cancer. Prostate cancer can be treated in a variety of ways, including surgery, androgen deprivation therapy, radiation therapy, and chemotherapy. The clinical trajectory of prostate cancer varies from patient to patient, but more aggressive tumors often tend to be radioresistant. This is often due to the free-radical and reactive-oxygen-species-neutralizing effects of the superoxide dismutase family. Superoxide dismutase 2, which is especially important in this regard, can be induced by the NF-κB pathway, which is an important mechanism in radioresistance. This information has enabled the development of interventions that manipulate the NF-κB mechanism to treat prostate cancer.
Collapse
|
4
|
Greenberger JS, Mukherjee A, Epperly MW. Gene Therapy for Systemic or Organ Specific Delivery of Manganese Superoxide Dismutase. Antioxidants (Basel) 2021; 10:1057. [PMID: 34208819 PMCID: PMC8300724 DOI: 10.3390/antiox10071057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 11/16/2022] Open
Abstract
Manganese superoxide dismutase (MnSOD) is a dominant component of the antioxidant defense system in mammalian cells. Since ionizing irradiation induces profound oxidative stress, it was logical to test the effect of overexpression of MnSOD on radioresistance. This task was accomplished by introduction of a transgene for MnSOD into cells in vitro and into organs in vivo, and both paradigms showed clear radioresistance following overexpression. During the course of development and clinical application of using MnSOD as a radioprotector, several prominent observations were made by Larry Oberley, Joel Greenberger, and Michael Epperly which include (1) mitochondrial localization of either manganese superoxide dismutase or copper/zinc SOD was required to provide optimal radiation protection; (2) the time required for optimal expression was 12-18 h, and while acceptable for radiation protection, the time delay was impractical for radiation mitigation; (3) significant increases in intracellular elevation of MnSOD activity were required for effective radioprotection. Lessons learned during the development of MnSOD gene therapy have provided a strategy for delivery of small molecule SOD mimics, which are faster acting and have shown the potential for both radiation protection and mitigation. The purpose of this review is to summarize the current status of using MnSOD-PL and SOD mimetics as radioprotectors and radiomitigators.
Collapse
Affiliation(s)
- Joel S. Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.M.); (M.W.E.)
| | | | | |
Collapse
|
5
|
Obrador E, Salvador R, Villaescusa JI, Soriano JM, Estrela JM, Montoro A. Radioprotection and Radiomitigation: From the Bench to Clinical Practice. Biomedicines 2020; 8:E461. [PMID: 33142986 PMCID: PMC7692399 DOI: 10.3390/biomedicines8110461] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
The development of protective agents against harmful radiations has been a subject of investigation for decades. However, effective (ideal) radioprotectors and radiomitigators remain an unsolved problem. Because ionizing radiation-induced cellular damage is primarily attributed to free radicals, radical scavengers are promising as potential radioprotectors. Early development of such agents focused on thiol synthetic compounds, e.g., amifostine (2-(3-aminopropylamino) ethylsulfanylphosphonic acid), approved as a radioprotector by the Food and Drug Administration (FDA, USA) but for limited clinical indications and not for nonclinical uses. To date, no new chemical entity has been approved by the FDA as a radiation countermeasure for acute radiation syndrome (ARS). All FDA-approved radiation countermeasures (filgrastim, a recombinant DNA form of the naturally occurring granulocyte colony-stimulating factor, G-CSF; pegfilgrastim, a PEGylated form of the recombinant human G-CSF; sargramostim, a recombinant granulocyte macrophage colony-stimulating factor, GM-CSF) are classified as radiomitigators. No radioprotector that can be administered prior to exposure has been approved for ARS. This differentiates radioprotectors (reduce direct damage caused by radiation) and radiomitigators (minimize toxicity even after radiation has been delivered). Molecules under development with the aim of reaching clinical practice and other nonclinical applications are discussed. Assays to evaluate the biological effects of ionizing radiations are also analyzed.
Collapse
Affiliation(s)
- Elena Obrador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (E.O.); (R.S.); (J.M.E.)
| | - Rosario Salvador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (E.O.); (R.S.); (J.M.E.)
| | - Juan I. Villaescusa
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain;
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| | - José M. Soriano
- Food & Health Lab, Institute of Materials Science, University of Valencia, 46980 Valencia, Spain;
- Joint Research Unit in Endocrinology, Nutrition and Clinical Dietetics, University of Valencia-Health Research Institute IISLaFe, 46026 Valencia, Spain
| | - José M. Estrela
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (E.O.); (R.S.); (J.M.E.)
| | - Alegría Montoro
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain;
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| |
Collapse
|
6
|
Zimmermann A, Hercher D, Regner B, Frischer A, Sperger S, Redl H, Hacobian A. Evaluation of BMP-2 Minicircle DNA for Enhanced Bone Engineering and Regeneration. Curr Gene Ther 2020; 20:55-63. [PMID: 32338217 DOI: 10.2174/1566523220666200427121350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND To date, the significant osteoinductive potential of bone morphogenetic protein 2 (BMP-2) non-viral gene therapy cannot be fully exploited therapeutically. This is mainly due to weak gene delivery and brief expression peaks restricting the therapeutic effect. OBJECTIVE Our objective was to test the application of minicircle DNA, allowing prolonged expression potential. It offers notable advantages over conventional plasmid DNA. The lack of bacterial sequences and the resulting reduction in size, enables safe usage and improved performance for tissue regeneration. METHODS We inserted an optimized BMP-2 gene cassette with minicircle plasmid technology. BMP-2 minicircle plasmids were produced in E. coli yielding plasmids lacking bacterial backbone elements. Comparative studies of these BMP-2 minicircles and conventional BMP-2 plasmids were performed in vitro in cell systems, including bone marrow derived stem cells. Tests performed included gene expression profiles and cell differentiation assays. RESULTS A C2C12 cell line transfected with the BMP-2-Advanced minicircle showed significantly elevated expression of osteocalcin, alkaline phosphatase (ALP) activity, and BMP-2 protein amount when compared to cells transfected with conventional BMP-2-Advanced plasmid. Furthermore, the plasmids show suitability for stem cell approaches by showing significantly higher levels of ALP activity and mineralization when introduced into human bone marrow stem cells (BMSCs). CONCLUSION We have designed a highly bioactive BMP-2 minicircle plasmid with the potential to fulfil clinical requirements for non-viral gene therapy in the field of bone regeneration.
Collapse
Affiliation(s)
- Alice Zimmermann
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - David Hercher
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - Benedikt Regner
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - Amelie Frischer
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - Simon Sperger
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - Ara Hacobian
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| |
Collapse
|
7
|
Farhood B, Ashrafizadeh M, Khodamoradi E, Hoseini-Ghahfarokhi M, Afrashi S, Musa AE, Najafi M. Targeting of cellular redox metabolism for mitigation of radiation injury. Life Sci 2020; 250:117570. [PMID: 32205088 DOI: 10.1016/j.lfs.2020.117570] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/17/2020] [Accepted: 03/17/2020] [Indexed: 12/14/2022]
Abstract
Accidental exposure to ionizing radiation is a serious concern to human life. Studies on the mitigation of side effects following exposure to accidental radiation events are ongoing. Recent studies have shown that radiation can activate several signaling pathways, leading to changes in the metabolism of free radicals including reactive oxygen species (ROS) and nitric oxide (NO). Cellular and molecular mechanisms show that radiation can cause disruption of normal reduction/oxidation (redox) system. Mitochondria malfunction following exposure to radiation and mutations in mitochondria DNA (mtDNA) have a key role in chronic oxidative stress. Furthermore, exposure to radiation leads to infiltration of inflammatory cells such as macrophages, lymphocytes and mast cells, which are important sources of ROS and NO. These cells generate free radicals via upregulation of some pro-oxidant enzymes such as NADPH oxidases, inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Epigenetic changes also have a key role in a similar way. Other mediators such as mammalian target of rapamycin (mTOR) and peroxisome proliferator-activated receptor (PPAR), which are involved in the normal metabolism of cells have also been shown to regulate cell death following exposure to radiation. These mechanisms are tissue specific. Inhibition or activation of each of these targets can be suggested for mitigation of radiation injury in a specific tissue. In the current paper, we review the cellular and molecular changes in the metabolism of cells and ROS/NO following exposure to radiation. Furthermore, the possible strategies for mitigation of radiation injury through modulation of cellular metabolism in irradiated organs will be discussed.
Collapse
Affiliation(s)
- Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Milad Ashrafizadeh
- Department of Basic Science, Veterinary Medicine Faculty, Tabriz University, Tabriz, Iran
| | - Ehsan Khodamoradi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mojtaba Hoseini-Ghahfarokhi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Shima Afrashi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences (International Campus), Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
8
|
Yan XX, Li HL, Zhang YT, Wu SY, Lu HL, Yu XL, Meng FG, Sun JH, Gong LK. A new recombinant MS-superoxide dismutase alleviates 5-fluorouracil-induced intestinal mucositis in mice. Acta Pharmacol Sin 2020; 41:348-357. [PMID: 31506573 PMCID: PMC7468365 DOI: 10.1038/s41401-019-0295-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 08/06/2019] [Indexed: 12/21/2022]
Abstract
Intestinal mucositis is a common side effect of anticancer regimens that exerts a negative impact on chemotherapy. Superoxide dismutase (SOD) is a potential therapy for mucositis but efficient product is not available because the enzyme is degraded following oral administration or induces an immune reaction after intravascular infusion. Multi-modified Stable Anti-Oxidant Enzymes® (MS-AOE®) is a new recombinant SOD with better resistance to pepsin and trypsin. We referred it as MS-SOD to distinguish from other SODs. In this study we investigated its potential to alleviate 5-FU-induced intestinal injury and the mechanisms. An intestinal mucositis model was established in C57/BL6 mice by 5-day administration of 5-FU (50 mg/kg every day, ip). MS-SOD (800 IU/10 g, ig) was given once daily for 9 days. 5-FU caused severe mucositis with intestinal morphological damage, bodyweight loss and diarrhea; MS-SOD significantly decreased the severity. 5-FU markedly increased reactive oxygen species (ROS) and inflammatory cytokines in the intestine which were ameliorated by MS-SOD. Furthermore, MS-SOD modified intestinal microbes, particularly reduced Verrucomicrobia, compared with the 5-FU group. In Caco2 cells, MS-SOD (250–1000 U/mL) dose-dependently decreased tBHP-induced ROS generation. In RAW264.7 cells, MS-SOD (500 U/mL) had no effect on LPS-induced inflammatory cytokines, but inhibited iNOS expression. These results demonstrate that MS-SOD can scavenge ROS at the initial stage of injury, thus play an indirect role in anti-inflammatory and barrier protein protection. In conclusion, MS-SOD attenuates 5-FU-induced intestinal mucositis by suppressing oxidative stress and inflammation, and influencing microbes. MS-SOD may exert beneficial effect in prevention of intestinal mucositis during chemotherapy in clinic.
Collapse
|
9
|
Caputo F, Giovanetti A, Corsi F, Maresca V, Briganti S, Licoccia S, Traversa E, Ghibelli L. Cerium Oxide Nanoparticles Re-establish Cell Integrity Checkpoints and Apoptosis Competence in Irradiated HaCat Cells via Novel Redox-Independent Activity. Front Pharmacol 2018; 9:1183. [PMID: 30459604 PMCID: PMC6232693 DOI: 10.3389/fphar.2018.01183] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 09/28/2018] [Indexed: 12/11/2022] Open
Abstract
Cerium oxide nanoparticles (CNPs) are potent radical scavengers protecting cells from oxidative insults, including ionizing radiation. Here we show that CNPs prevent X-ray-induced oxidative imbalance reducing DNA breaks on HaCat keratinocytes, nearly abating mutagenesis. At the same time, and in spite of the reduced damage, CNPs strengthen radiation-induced cell cycle arrest and apoptosis outcome, dropping colony formation; notably, CNPs do not possess any intrinsic toxicity toward non-irradiated HaCat, indicating that they act on damaged cells. Thus CNPs, while exerting their antioxidant action, also reinforce the stringency of damage-induced cell integrity checkpoints, promoting elimination of the “tolerant” cells, being in fact radio-sensitizers. These two contrasting pathways are mediated by different activities of CNPs: indeed Sm-doped CNPs, which lack the Ce3+/Ce4+ redox switch and the correlated antioxidant action, fail to decrease radiation-induced superoxide formation, as expected, but surprisingly maintain the radio-sensitizing ability and the dramatic decrease of mutagenesis. The latter is thus attributable to elimination of damaged cells rather than decreased oxidative damage. This highlights a novel redox-independent activity of CNPs, allowing selectively eliminating heavily damaged cells through non-toxic mechanisms, rather reactivating endogenous anticancer pathways in transformed cells.
Collapse
Affiliation(s)
- Fanny Caputo
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Rome, Italy.,Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | - Francesca Corsi
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Rome, Italy
| | | | | | - Silvia Licoccia
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Rome, Italy
| | - Enrico Traversa
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu, China
| | - Lina Ghibelli
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
10
|
Cortese F, Klokov D, Osipov A, Stefaniak J, Moskalev A, Schastnaya J, Cantor C, Aliper A, Mamoshina P, Ushakov I, Sapetsky A, Vanhaelen Q, Alchinova I, Karganov M, Kovalchuk O, Wilkins R, Shtemberg A, Moreels M, Baatout S, Izumchenko E, de Magalhães JP, Artemov AV, Costes SV, Beheshti A, Mao XW, Pecaut MJ, Kaminskiy D, Ozerov IV, Scheibye-Knudsen M, Zhavoronkov A. Vive la radiorésistance!: converging research in radiobiology and biogerontology to enhance human radioresistance for deep space exploration and colonization. Oncotarget 2018; 9:14692-14722. [PMID: 29581875 PMCID: PMC5865701 DOI: 10.18632/oncotarget.24461] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/31/2018] [Indexed: 12/12/2022] Open
Abstract
While many efforts have been made to pave the way toward human space colonization, little consideration has been given to the methods of protecting spacefarers against harsh cosmic and local radioactive environments and the high costs associated with protection from the deleterious physiological effects of exposure to high-Linear energy transfer (high-LET) radiation. Herein, we lay the foundations of a roadmap toward enhancing human radioresistance for the purposes of deep space colonization and exploration. We outline future research directions toward the goal of enhancing human radioresistance, including upregulation of endogenous repair and radioprotective mechanisms, possible leeways into gene therapy in order to enhance radioresistance via the translation of exogenous and engineered DNA repair and radioprotective mechanisms, the substitution of organic molecules with fortified isoforms, and methods of slowing metabolic activity while preserving cognitive function. We conclude by presenting the known associations between radioresistance and longevity, and articulating the position that enhancing human radioresistance is likely to extend the healthspan of human spacefarers as well.
Collapse
Affiliation(s)
- Franco Cortese
- Biogerontology Research Foundation, London, UK
- Department of Biomedical and Molecular Sciences, Queen's University School of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Dmitry Klokov
- Canadian Nuclear Laboratories, Chalk River, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Andreyan Osipov
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow, Russia
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Jakub Stefaniak
- Biogerontology Research Foundation, London, UK
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, UK
| | - Alexey Moskalev
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Laboratory of Molecular Radiobiology and Gerontology, Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia
- Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, Russia
| | - Jane Schastnaya
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
| | - Charles Cantor
- Boston University, Department of Biomedical Engineering, Boston, MA, USA
| | - Alexander Aliper
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
- Laboratory of Bioinformatics, D. Rogachev Federal Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Polina Mamoshina
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
- Computer Science Department, University of Oxford, Oxford, UK
| | - Igor Ushakov
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow, Russia
| | - Alex Sapetsky
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow, Russia
| | - Quentin Vanhaelen
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
| | - Irina Alchinova
- Laboratory of Physicochemical and Ecological Pathophysiology, Institute of General Pathology and Pathophysiology, Moscow, Russia
- Research Institute for Space Medicine, Federal Medical Biological Agency, Moscow, Russia
| | - Mikhail Karganov
- Laboratory of Physicochemical and Ecological Pathophysiology, Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Olga Kovalchuk
- Canada Cancer and Aging Research Laboratories, Ltd., Lethbridge, Alberta, Canada
- University of Lethbridge, Lethbridge, Alberta, Canada
| | - Ruth Wilkins
- Environmental and Radiation and Health Sciences Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Andrey Shtemberg
- Laboratory of Extreme Physiology, Institute of Medical and Biological Problems RAS, Moscow, Russia
| | - Marjan Moreels
- Radiobiology Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre, (SCK·CEN), Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Interdisciplinary Biosciences, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre, (SCK·CEN), Mol, Belgium
- Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| | - Evgeny Izumchenko
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
- The Johns Hopkins University, School of Medicine, Department of Otolaryngology, Head and Neck Cancer Research, Baltimore, MD, USA
| | - João Pedro de Magalhães
- Biogerontology Research Foundation, London, UK
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Artem V. Artemov
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
| | | | - Afshin Beheshti
- Wyle Laboratories, Space Biosciences Division, NASA Ames Research Center, Mountain View, CA, USA
- Division of Hematology/Oncology, Molecular Oncology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Xiao Wen Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University, Loma Linda, CA, USA
| | - Michael J. Pecaut
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University, Loma Linda, CA, USA
| | - Dmitry Kaminskiy
- Biogerontology Research Foundation, London, UK
- Deep Knowledge Life Sciences, London, UK
| | - Ivan V. Ozerov
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow, Russia
| | | | - Alex Zhavoronkov
- Biogerontology Research Foundation, London, UK
- Insilico Medicine, Inc., Emerging Technology Centers, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
11
|
Khan AA, Paget JT, McLaughlin M, Kyula JN, Wilkinson MJ, Pencavel T, Mansfield D, Roulstone V, Seth R, Halle M, Somaiah N, Boult JKR, Robinson SP, Pandha HS, Vile RG, Melcher AA, Harris PA, Harrington KJ. Genetically modified lentiviruses that preserve microvascular function protect against late radiation damage in normal tissues. Sci Transl Med 2018; 10:eaar2041. [PMID: 29367346 PMCID: PMC6020074 DOI: 10.1126/scitranslmed.aar2041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 10/15/2017] [Accepted: 11/08/2017] [Indexed: 12/25/2022]
Abstract
Improvements in cancer survival mean that long-term toxicities, which contribute to the morbidity of cancer survivorship, are being increasingly recognized. Late adverse effects (LAEs) in normal tissues after radiotherapy (RT) are characterized by vascular dysfunction and fibrosis causing volume loss and tissue contracture, for example, in the free flaps used for immediate breast reconstruction after mastectomy. We evaluated the efficacy of lentivirally delivered superoxide dismutase 2 (SOD2) overexpression and connective tissue growth factor (CTGF) knockdown by short hairpin RNA in reducing the severity of LAEs in an animal model of free flap LAEs. Vectors were delivered by intra-arterial injection, ex vivo, to target the vascular compartment. LVSOD2 and LVshCTGF monotherapy before irradiation resulted in preservation of flap volume or reduction in skin contracture, respectively. Flaps transduced with combination therapy experienced improvements in both volume loss and skin contracture. Both therapies reduced the fibrotic burden after irradiation. LAEs were associated with impaired vascular perfusion, loss of endothelial permeability, and stromal hypoxia, which were all reversed in the treatment model. Using a tumor recurrence model, we showed that SOD2 overexpression in normal tissues did not compromise the efficacy of RT against tumor cells but appeared to enhance it. LVSOD2 and LVshCTGF combination therapy by targeted, intravascular delivery reduced LAE severities in normal tissues without compromising the efficacy of RT and warrants translational evaluation as a free flap-targeted gene therapy.
Collapse
Affiliation(s)
- Aadil A Khan
- Targeted Therapy Team, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW3 6JB, UK
- Department of Plastic Surgery, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - James T Paget
- Targeted Therapy Team, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW3 6JB, UK
- Department of Plastic Surgery, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Martin McLaughlin
- Targeted Therapy Team, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW3 6JB, UK
| | - Joan N Kyula
- Targeted Therapy Team, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW3 6JB, UK
| | - Michelle J Wilkinson
- Targeted Therapy Team, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW3 6JB, UK
| | - Timothy Pencavel
- Targeted Therapy Team, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW3 6JB, UK
| | - David Mansfield
- Targeted Therapy Team, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW3 6JB, UK
| | - Victoria Roulstone
- Targeted Therapy Team, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW3 6JB, UK
| | - Rohit Seth
- Targeted Therapy Team, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW3 6JB, UK
| | - Martin Halle
- Department of Molecular Medicine and Surgery, Section of Plastic Surgery, Karolinska Institute, Stockholm 17176, Sweden
- Department of Reconstructive Plastic Surgery, Karolinska University Hospital, Stockholm 17176, Sweden
| | - Navita Somaiah
- Targeted Therapy Team, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW3 6JB, UK
| | - Jessica K R Boult
- Magnetic Resonance Team, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SM2 5NG, UK
| | - Simon P Robinson
- Magnetic Resonance Team, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SM2 5NG, UK
| | - Hardev S Pandha
- Postgraduate Medical School, University of Surrey, Guildford GU2 7XH, UK
| | - Richard G Vile
- Molecular Medicine Program, Mayo Clinic, Rochester, MN 55905, USA
| | - Alan A Melcher
- Translational Immunotherapy Team, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Paul A Harris
- Department of Plastic Surgery, The Royal Marsden Hospital, London SW3 6JJ, UK
| | - Kevin J Harrington
- Targeted Therapy Team, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW3 6JB, UK.
| |
Collapse
|
12
|
Prévention médicale et traitement des complications pulmonaires secondaires à la radiothérapie. Cancer Radiother 2017; 21:411-423. [DOI: 10.1016/j.canrad.2017.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/13/2017] [Accepted: 03/24/2017] [Indexed: 12/12/2022]
|
13
|
Šimčíková M, Prather KLJ, Prazeres DMF, Monteiro GA. Towards effective non-viral gene delivery vector. Biotechnol Genet Eng Rev 2017; 31:82-107. [PMID: 27160661 DOI: 10.1080/02648725.2016.1178011] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Despite very good safety records, clinical trials using plasmid DNA failed due to low transfection efficiency and brief transgene expression. Although this failure is both due to poor plasmid design and to inefficient delivery methods, here we will focus on the former. The DNA elements like CpG motifs, selection markers, origins of replication, cryptic eukaryotic signals or nuclease-susceptible regions and inverted repeats showed detrimental effects on plasmids' performance as biopharmaceuticals. On the other hand, careful selection of promoter, polyadenylation signal, codon optimization and/or insertion of introns or nuclear-targeting sequences for therapeutic protein expression can enhance the clinical efficacy. Minimal vectors, which are devoid of the bacterial backbone and consist exclusively of the eukaryotic expression cassette, demonstrate better performance in terms of expression levels, bioavailability, transfection rates and increased therapeutic effects. Although the results are promising, minimal vectors have not taken over the conventional plasmids in clinical trials due to challenging manufacturing issues.
Collapse
Affiliation(s)
- Michaela Šimčíková
- a MIT-Portugal Program.,b iBB-Institute for Bioengineering and Biosciences , Lisbon , Portugal
| | - Kristala L J Prather
- a MIT-Portugal Program.,c Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Duarte M F Prazeres
- a MIT-Portugal Program.,c Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , MA , USA.,d Department of Bioengineering , Instituto Superior Técnico , Lisbon , Portugal
| | - Gabriel A Monteiro
- a MIT-Portugal Program.,c Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , MA , USA.,d Department of Bioengineering , Instituto Superior Técnico , Lisbon , Portugal
| |
Collapse
|
14
|
Mignon C, Sodoyer R, Werle B. Antibiotic-free selection in biotherapeutics: now and forever. Pathogens 2015; 4:157-81. [PMID: 25854922 PMCID: PMC4493468 DOI: 10.3390/pathogens4020157] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/09/2015] [Accepted: 03/23/2015] [Indexed: 11/16/2022] Open
Abstract
The continuously improving sophistication of molecular engineering techniques gives access to novel classes of bio-therapeutics and new challenges for their production in full respect of the strengthening regulations. Among these biologic agents are DNA based vaccines or gene therapy products and to a lesser extent genetically engineered live vaccines or delivery vehicles. The use of antibiotic-based selection, frequently associated with genetic manipulation of microorganism is currently undergoing a profound metamorphosis with the implementation and diversification of alternative selection means. This short review will present examples of alternatives to antibiotic selection and their context of application to highlight their ineluctable invasion of the bio-therapeutic world.
Collapse
Affiliation(s)
- Charlotte Mignon
- Technology Research Institute Bioaster, 317 avenue Jean-Jaurés, 69007 Lyon, France.
| | - Régis Sodoyer
- Technology Research Institute Bioaster, 317 avenue Jean-Jaurés, 69007 Lyon, France.
| | - Bettina Werle
- Technology Research Institute Bioaster, 317 avenue Jean-Jaurés, 69007 Lyon, France.
| |
Collapse
|
15
|
Kobashigawa S, Kashino G, Suzuki K, Yamashita S, Mori H. Ionizing radiation-induced cell death is partly caused by increase of mitochondrial reactive oxygen species in normal human fibroblast cells. Radiat Res 2015; 183:455-64. [PMID: 25807320 DOI: 10.1667/rr13772.1] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiation-induced cell death is thought to be caused by nuclear DNA damage that cannot be repaired. However, in this study we found that a delayed increase of mitochondrial reactive oxygen species (ROS) is responsible for some of the radiation-induced cell death in normal human fibroblast cells. We have previously reported that there is a delayed increase of mitochondrial (·)O2(-), measured using MitoSOX™ Red reagent, due to gamma irradiation. This is dependent on Drp1 localization to mitochondria. Here, we show that knockdown of Drp1 expression reduces the level of DNA double-strand breaks (DSBs) remaining 3 days after 6 Gy irradiation. Furthermore, cells with knockdown of Drp1 expression are more resistant to gamma radiation. We then tested whether the delayed increase of ROS causes DNA damage. The antioxidant, 2-glucopyranoside ascorbic acid (AA-2G), was applied before or after irradiation to inhibit ROS production during irradiation or to inhibit delayed ROS production from mitochondria. Interestingly, 1 h after exposure, the AA-2G treatment reduced the level of DSBs remaining 3 days after 6 Gy irradiation. In addition, irradiated AA-2G-treated cells were more resistant to radiation than the untreated cells. These results indicate that delayed mitochondrial ROS production may cause some of the cell death after irradiation.
Collapse
Affiliation(s)
- Shinko Kobashigawa
- a Department of Radiology, School of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu City, Oita, Japan
| | | | | | | | | |
Collapse
|
16
|
Gene therapy for radioprotection. Cancer Gene Ther 2015; 22:172-80. [PMID: 25721205 DOI: 10.1038/cgt.2015.8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 08/01/2014] [Accepted: 01/22/2015] [Indexed: 11/08/2022]
Abstract
Radiation therapy is a critical component of cancer treatment with over half of patients receiving radiation during their treatment. Despite advances in image-guided therapy and dose fractionation, patients receiving radiation therapy are still at risk for side effects due to off-target radiation damage of normal tissues. To reduce normal tissue damage, researchers have sought radioprotectors, which are agents capable of protecting tissue against radiation by preventing radiation damage from occurring or by decreasing cell death in the presence of radiation damage. Although much early research focused on small-molecule radioprotectors, there has been a growing interest in gene therapy for radioprotection. The amenability of gene therapy vectors to targeting, as well as the flexibility of gene therapy to accomplish ablation or augmentation of biologically relevant genes, makes gene therapy an excellent strategy for radioprotection. Future improvements to vector targeting and delivery should greatly enhance radioprotection through gene therapy.
Collapse
|
17
|
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
|
18
|
Mikkelsen JG. Nonviral Gene Therapy—The Challenge of Mobilizing DNA. SOMATIC GENOME MANIPULATION 2015:69-104. [DOI: 10.1007/978-1-4939-2389-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
19
|
Wong SP, Argyros O, Harbottle RP. Sustained expression from DNA vectors. ADVANCES IN GENETICS 2014; 89:113-152. [PMID: 25620010 DOI: 10.1016/bs.adgen.2014.11.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
DNA vectors have the potential to become powerful medical tools for treatment of human disease. The human body has, however, developed a range of defensive strategies to detect and silence foreign or misplaced DNA, which is more typically encountered during infection or chromosomal damage. A clinically relevant human gene therapy vector must overcome or avoid these protections whilst delivering sustained levels of therapeutic gene product without compromising the vitality of the recipient host. Many non-viral DNA vectors trigger these defense mechanisms and are subsequently destroyed or rendered silent. Thus, without modification or considered design, the clinical utility of a typical DNA vector is fundamentally limited due to the transient nature of its transgene expression. The development of safe and persistently expressing DNA vectors is a crucial prerequisite for its successful clinical application and subsequently remains, therefore, one of the main strategic tasks of non-viral gene therapy research. In this chapter we will describe our current understanding of the mechanisms that can destroy or silence DNA vectors and discuss strategies, which have been utilized to improve their sustenance and the level and duration of their transgene expression.
Collapse
Affiliation(s)
- Suet Ping Wong
- Leukocyte Biology Section, National Heart & Lung Institute, Imperial College London, London, UK
| | - Orestis Argyros
- Division of Pharmacology-Pharmacotechnology, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Richard P Harbottle
- DNA Vector Research, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| |
Collapse
|
20
|
Kam WWY, Banati RB. Effects of ionizing radiation on mitochondria. Free Radic Biol Med 2013; 65:607-619. [PMID: 23892359 DOI: 10.1016/j.freeradbiomed.2013.07.024] [Citation(s) in RCA: 271] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 07/16/2013] [Accepted: 07/16/2013] [Indexed: 01/08/2023]
Abstract
The current concept of radiobiology posits that damage to the DNA in the cell nucleus is the primary cause for the detrimental effects of radiation. However, emerging experimental evidence suggests that this theoretical framework is insufficient for describing extranuclear radiation effects, particularly the response of the mitochondria, an important site of extranuclear, coding DNA. Here, we discuss experimental observations of the effects of ionizing radiation on the mitochondria at (1) the DNA and (2) functional levels. The roles of mitochondria in (3) oxidative stress and (4) late radiation effects are discussed. In this review, we summarize the current understanding of targets for ionizing radiation outside the cell nucleus. Available experimental data suggest that an increase in the tumoricidal efficacy of radiation therapy might be achievable by targeting mitochondria. Likewise, more specific protection of mitochondria and its coding DNA should reduce damage to healthy cells exposed to ionizing radiation.
Collapse
Affiliation(s)
- Winnie Wai-Ying Kam
- Australian Nuclear Science and Technology Organisation, Lucas Heights, Sydney, New South Wales 2234, Australia; Medical Radiation Sciences, Faculty of Health Sciences, University of Sydney, Cumberland, Sydney, New South Wales 2141, Australia.
| | - Richard B Banati
- Australian Nuclear Science and Technology Organisation, Lucas Heights, Sydney, New South Wales 2234, Australia; Medical Radiation Sciences, Faculty of Health Sciences, University of Sydney, Cumberland, Sydney, New South Wales 2141, Australia; National Imaging Facility at Brain and Mind Research Institute (BMRI), University of Sydney, Camperdown, Sydney, New South Wales 2050, Australia
| |
Collapse
|
21
|
In vivo electroporation of minicircle DNA as a novel method of vaccine delivery to enhance HIV-1-specific immune responses. J Virol 2013; 88:1924-34. [PMID: 24284319 DOI: 10.1128/jvi.02757-13] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
DNA vaccines offer advantage over conventional vaccines, as they are safer to use, easier to produce, and able to induce humoral as well cellular immune responses. Unfortunately, no DNA vaccines have been licensed for human use for the difficulties in developing an efficient and safe in vivo gene delivery system. In vivo electroporation (EP)-based DNA delivery has attracted great attention for its potency to enhance cellular uptake of DNA vaccines and function as an adjuvant. Minicircle DNA (a new form of DNA containing only a gene expression cassette and lacking a backbone of bacterial plasmid DNA) is a powerful candidate of gene delivery in terms of improving the levels and the duration of transgene expression in vivo. In this study, as a novel vaccine delivery system, we combined in vivo EP and the minicircle DNA carrying a codon-optimized HIV-1 gag gene (minicircle-gag) to evaluate the immunogenicity of this system. We found that minicircle-gag conferred persistent and high levels of gag expression in vitro and in vivo. The use of EP delivery further increased minicircle-based gene expression. Moreover, when delivered by EP, minicircle-gag vaccination elicited a 2- to 3-fold increase in cellular immune response and a 1.5- to 3-fold augmentation of humoral immune responses compared with those elicited by a pVAX1-gag positive control. Increased immunogenicity of EP-assisted minicircle-gag may benefit from increasing local antigen expression, upregulating inflammatory genes, and recruiting immune cells. Collectively, in vivo EP of minicircle DNA functions as a novel vaccine platform that can enhance efficacy and immunogenicity of DNA vaccines.
Collapse
|
22
|
Sun GG, Hu WN, Wang YD, Yang CR, Lu YF. Bidirectional regulation of manganese superoxide dismutase (MnSOD) on the radiosensitivity of esophageal cancer cells. Asian Pac J Cancer Prev 2013; 13:3015-23. [PMID: 22994704 DOI: 10.7314/apjcp.2012.13.7.3015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The mitochondrial antioxidant protein manganese superoxide dismutase (MnSOD) may represent a new type of tumor suppressor protein. Overexpression of the cDNA of this gene by plasmid or recombinant lentiviral transfection in various types of cancer leads to growth suppression both in vitro and in vivo. We previously determined that changes in MnSOD expression had bidirectional effects on adriamycin (ADR) when combined with nitric oxide (NO). Radiation induces free radicals in a manner similar to ADR, so we speculated that MnSOD combined with NO would also have a bidirectional effect on cellular radiosensitivity. To examine this hypothesis, TE-1 human esophageal squamous carcinoma cells were stably transfected using lipofectamine with a pLenti6-DEST plasmid containing human MnSOD cDNA at moderate to high overexpression levels or with no MnSOD insert. Blastidicin-resistant colonies were isolated, grown, and maintained in culture. We found that moderate overexpression of MnSOD decreased growth rates, plating efficiency, and increased apoptosis. However, high overexpression increased growth rates, plating efficiency, and decreased apoptosis. When combined with NO, moderate overexpression of MnSOD increased the radiosensitivity of esophageal cancer cells, whereas high MnSOD overexpression had the opposite effect. This finding suggests a potential new method to kill certain radioresistant tumors and to provide radioresistance to normal cells.
Collapse
Affiliation(s)
- Guo-Gui Sun
- Department of Chemoradiotherapy, Tangshan People's Hospital, Tangshan, China
| | | | | | | | | |
Collapse
|
23
|
Dietz WM, Skinner NEB, Hamilton SE, Jund MD, Heitfeld SM, Litterman AJ, Hwu P, Chen ZY, Salazar AM, Ohlfest JR, Blazar BR, Pennell CA, Osborn MJ. Minicircle DNA is superior to plasmid DNA in eliciting antigen-specific CD8+ T-cell responses. Mol Ther 2013; 21:1526-35. [PMID: 23689601 DOI: 10.1038/mt.2013.85] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 04/10/2013] [Indexed: 01/19/2023] Open
Abstract
Clinical trials reveal that plasmid DNA (pDNA)-based gene delivery must be improved to realize its potential to treat human disease. Current pDNA platforms suffer from brief transgene expression, primarily due to the spread of transcriptionally repressive chromatin initially deposited on plasmid bacterial backbone sequences. Minicircle (MC) DNA lacks plasmid backbone sequences and correspondingly confers higher levels of sustained transgene expression upon delivery, accounting for its success in preclinical gene therapy models. In this study, we show for the first time that MC DNA also functions as a vaccine platform. We used a luciferase reporter transgene to demonstrate that intradermal delivery of MC DNA, relative to pDNA, resulted in significantly higher and persistent levels of luciferase expression in mouse skin. Next, we immunized mice intradermally with DNA encoding a peptide that, when presented by the appropriate major histocompatibility complex class I molecule, was recognized by endogenous CD8(+) T cells. Finally, immunization with peptide-encoding MC DNA, but not the corresponding full-length (FL) pDNA, conferred significant protection in mice challenged with Listeria monocytogenes expressing the model peptide. Together, our results suggest intradermal delivery of MC DNA may prove more efficacious for prophylaxis than traditional pDNA vaccines.
Collapse
Affiliation(s)
- Wynette M Dietz
- Department of Laboratory Medicine and Pathology, Center for Immunology and Masonic Cancer Center, University of Minnesota School of Medicine, Minneapolis, Minnesota, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Hematopoietic stem cell regeneration enhanced by ectopic expression of ROS-detoxifying enzymes in transplant mice. Mol Ther 2013; 21:423-32. [PMID: 23295952 DOI: 10.1038/mt.2012.232] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
High levels of reactive oxygen species (ROS) can exhaust hematopoietic stem cells (HSCs). Thus, maintaining a low state of redox in HSCs by modulating ROS-detoxifying enzymes may augment the regeneration potential of HSCs. Our results show that basal expression of manganese superoxide dismutase (MnSOD) and catalase were at low levels in long-term and short-term repopulating HSCs, and administration of a MnSOD plasmid and lipofectin complex (MnSOD-PL) conferred radiation protection on irradiated recipient mice. To assess the intrinsic role of elevated MnSOD or catalase in HSCs and hematopoietic progenitor cells, the MnSOD or catalase gene was overexpressed in mouse hematopoietic cells via retroviral transduction. The impact of MnSOD and catalase on hematopoietic progenitor cells was mild, as measured by colony-forming units (CFUs). However, overexpressed catalase had a significant beneficial effect on long-term engraftment of transplanted HSCs, and this effect was further enhanced after an insult of low-dose γ-irradiation in the transplant mice. In contrast, overexpressed MnSOD exhibited an insignificant effect on long-term engraftment of transplanted HSCs, but had a significant beneficial effect after an insult of sublethal irradiation. Taken together, these results demonstrate that HSC function can be enhanced by ectopic expression of ROS-detoxifying enzymes, especially after radiation exposure in vivo.
Collapse
|
25
|
Wang HS, Chen ZJ, Zhang G, Ou XL, Yang XL, Wong CKC, Giesy JP, Du J, Chen SY. A novel micro-linear vector for in vitro and in vivo gene delivery and its application for EBV positive tumors. PLoS One 2012; 7:e47159. [PMID: 23077563 PMCID: PMC3471901 DOI: 10.1371/journal.pone.0047159] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 09/10/2012] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND The gene delivery vector for DNA-based therapy should ensure its transfection efficiency and safety for clinical application. The Micro-Linear vector (MiLV) was developed to improve the limitations of traditional vectors such as viral vectors and plasmids. METHODS The MiLV which contained only the gene expression cassette was amplified by polymerase chain reaction (PCR). Its cytotoxicity, transfection efficiency in vitro and in vivo, duration of expression, pro-inflammatory responses and potential application for Epstein-Barr virus (EBV) positive tumors were evaluated. RESULTS Transfection efficiency for exogenous genes transferred by MiLV was at least comparable with or even greater than their corresponding plasmids in eukaryotic cell lines. MiLV elevated the expression and prolonged the duration of genes in vitro and in vivo when compared with that of the plasmid. The in vivo pro-inflammatory response of MiLV group was lower than that of the plasmid group. The MEKK1 gene transferred by MiLV significantly elevated the sensitivity of B95-8 cells and transplanted tumor to the treatment of Ganciclovir (GCV) and sodium butyrate (NaB). CONCLUSIONS The present study provides a safer, more efficient and stable MiLV gene delivery vector than plasmid. These advantages encourage further development and the preferential use of this novel vector type for clinical gene therapy studies.
Collapse
Affiliation(s)
- Hong-Sheng Wang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- * E-mail: (JD); (HSW)
| | - Zhuo-Jia Chen
- Institute of Clinical Pharmacology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Ge Zhang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Xue-Ling Ou
- Department of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangzhou, People’s Republic of China
| | - Xiang-Ling Yang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Chris K. C. Wong
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong SAR, People’s Republic of China
| | - John P. Giesy
- Department of Veterinary Biomedical Sciences & Toxicological Center, University of Saskatchewan, Saskatchewan, Canada
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, People’s Republic of China
- * E-mail: (JD); (HSW)
| | - Shou-Yi Chen
- Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| |
Collapse
|
26
|
Caputo F, Vegliante R, Ghibelli L. Redox modulation of the DNA damage response. Biochem Pharmacol 2012; 84:1292-306. [PMID: 22846600 DOI: 10.1016/j.bcp.2012.07.022] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 07/18/2012] [Accepted: 07/19/2012] [Indexed: 01/09/2023]
Abstract
Lesions to DNA trigger the DNA-damage response (DDR), a complex, multi-branched cell-intrinsic process targeted to DNA repair, or elimination of the damaged cells by apoptosis. DDR aims at reducing permanence of mutated cells, decreasing the risk of tumor development: the more stringent the response, the lower the likelihood that sub-lethally damaged, unrepaired cells survive and proliferate. Accordingly, leakage often occurs in tumor cells with compromised DDR, accumulating mutations and accelerating tumor progression. Oxidations mediate DNA damage upon different insults such as UV, X and γ radiation, pollutants, poisons, or endogenous disequilibria, producing different types of lesions that trigger DDR, which can be alleviated by antioxidants. But reactive oxygen species (ROS), and the enzymes involved in their production or scavenging, also participate in DDR signaling, modulating the activity of key enzymes, and regulating the stringency of DDR. Accordingly, antioxidant enzymes such as superoxide dismutase play intimate and complex roles in tumor development, exceeding the basal roles of preventing the initial DNA damage. Likewise, it is emerging that dietary antioxidants help controlling tumor onset and progression by preventing DNA damage and by acting on cell cycle checkpoints, opening a novel and promising frontier to anticancer therapy.
Collapse
Affiliation(s)
- Fanny Caputo
- Dipartimento di Scienze e Tecnologie Chimiche, Universita' di Roma Tor Vergata, Roma, Italy
| | | | | |
Collapse
|
27
|
Chabot S, Orio J, Schmeer M, Schleef M, Golzio M, Teissié J. Minicircle DNA electrotransfer for efficient tissue-targeted gene delivery. Gene Ther 2012; 20:62-8. [DOI: 10.1038/gt.2011.215] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
28
|
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
|
29
|
Wong SP, Argyros O, Harbottle RP. Vector systems for prenatal gene therapy: principles of non-viral vector design and production. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2012; 891:133-67. [PMID: 22648771 DOI: 10.1007/978-1-61779-873-3_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gene therapy vectors based on viruses are the most effective gene delivery systems in use today and although efficient at gene transfer their potential toxicity (Hacein-Bey-Abina et al., Science 302:415-419, 2003) provides impetus for the development of safer non-viral alternatives. An ideal vector for human gene therapy should deliver sustainable therapeutic levels of gene expression without affecting the viability of the host at either the cellular or somatic level. Vectors, which comprise entirely human elements, may provide the most suitable method of achieving this. Non-viral vectors are attractive alternatives to viral gene delivery systems because of their low toxicity, relatively easy production, and great versatility. The development of more efficient, economically prepared, and safer gene delivery vectors is a crucial prerequisite for their successful clinical application and remains a primary strategic task of gene therapy research.
Collapse
Affiliation(s)
- Suet Ping Wong
- Faculty of Medicine, Molecular and Cellular Medicine Section, National Heart and Lung Institute, Imperial College London, London, UK
| | | | | |
Collapse
|
30
|
Abstract
Plasmid DNA (pDNA) is the base for promising DNA vaccines and gene therapies against many infectious, acquired, and genetic diseases, including HIV-AIDS, Ebola, Malaria, and different types of cancer, enteric pathogens, and influenza. Compared to conventional vaccines, DNA vaccines have many advantages such as high stability, not being infectious, focusing the immune response to only those antigens desired for immunization and long-term persistence of the vaccine protection. Especially in developing countries, where conventional effective vaccines are often unavailable or too expensive, there is a need for both new and improved vaccines. Therefore the demand of pDNA is expected to rise significantly in the near future. Since the injection of pDNA usually only leads to a weak immune response, several milligrams of DNA vaccine are necessary for immunization protection. Hence, there is a special interest to raise the product yield in order to reduce manufacturing costs. In this chapter, the different stages of plasmid DNA production are reviewed, from the vector design to downstream operation options. In particular, recent advances on cell engineering for improving plasmid DNA production are discussed.
Collapse
Affiliation(s)
- Alvaro R Lara
- Departamento de Procesos y Tecnología, Universidad Autónoma Metropolitana-Cuajimalpa, Mexico City, Mexico.
| | | | | |
Collapse
|
31
|
Vandermeulen G, Marie C, Scherman D, Préat V. New generation of plasmid backbones devoid of antibiotic resistance marker for gene therapy trials. Mol Ther 2011; 19:1942-9. [PMID: 21878901 PMCID: PMC3222533 DOI: 10.1038/mt.2011.182] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 08/03/2011] [Indexed: 12/29/2022] Open
Abstract
Since it has been established that the injection of plasmid DNA can lead to an efficient expression of a specific protein in vivo, nonviral gene therapy approaches have been considerably improved, allowing clinical trials. However, the use of antibiotic resistance genes as selection markers for plasmid production raises safety concerns which are often pointed out by the regulatory authorities. Indeed, a horizontal gene transfer to patient's bacteria cannot be excluded, and residual antibiotic in the final product could provoke allergic reactions in sensitive individuals. A new generation of plasmid backbones devoid of antibiotic resistance marker has emerged to increase the safety profile of nonviral gene therapy trials. This article reviews the existing strategies for plasmid maintenance and, in particular, those that do not require the use of antibiotic resistance genes. They are based either on the complementation of auxotrophic strain, toxin-antitoxin systems, operator-repressor titration, RNA markers, or on the overexpression of a growth essential gene. Minicircles that allow removing of the antibiotic resistance gene from the initial vector will also be discussed. Furthermore, reported use of antibiotic-free plasmids in preclinical or clinical studies will be listed to provide a comprehensive view of these innovative technologies.
Collapse
Affiliation(s)
- Gaëlle Vandermeulen
- Université catholique de Louvain, Louvain Drug Research Institute, Pharmaceutics and Drug Delivery, Brussels, Belgium
| | | | | | | |
Collapse
|
32
|
Zuo Y, Wu J, Xu Z, Yang S, Yan H, Tan L, Meng X, Ying X, Liu R, Kang T, Huang W. Minicircle-oriP-IFNγ: a novel targeted gene therapeutic system for EBV positive human nasopharyngeal carcinoma. PLoS One 2011; 6:e19407. [PMID: 21573215 PMCID: PMC3088667 DOI: 10.1371/journal.pone.0019407] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Accepted: 03/31/2011] [Indexed: 12/15/2022] Open
Abstract
Background Nonviral vectors are attractively used for gene therapy owing to their distinctive advantages. Our previous study has demonstrated that transfer of human IFNγ gene into nasopharyngeal carcinoma (NPC) by using a novel nonviral vector, minicircle (mc), under the control of cytomegalovirus (CMV) promoter was effective to inhibit tumor growth. However, therapies based on CMV promoter cannot express the targeted genes in cancer tissues. Previous studies indicated that the development of human NPC was closely associated with Epstein-Barr virus (EBV) and demonstrated the transcriptional enhancer function of oriP when bound by EBV protein. Therefore, the present study is to explore the targeted gene expression and the anti-tumor effect of a novel tumor-specific gene therapeutic system (mc-oriP-IFNγ) in which the transgene expression was under the transcriptional regulation of oriP promoter. Methodology/Principal Findings Dual-luciferase reporter assay and ELISA were used to assess the expression of luciferase and IFNγ. WST assay was used to assess the cell proliferation. RT-PCR was used to detect the mRNA level of EBNA1. RNAi was used to knockdown the expression of EBNA1. NPC xenograft models in nude mice were used to investigate the targeted antitumor efficacy of mc-oriP-IFNγ. Immunohistochemistry was used to detect the expression and the activity of the IFNγ in tumor sections. Our results demonstrated that mc-oriP vectors mediated comparable gene expression and anti-proliferative effect in the EBV-positive NPC cell line C666-1 compared to mc-CMV vectors. Furthermore, mc-oriP vectors exhibited much lower killing effects on EBV-negative cell lines compared to mc-CMV vectors. The targeted expression of mc-oriP vectors was inhibited by EBNA1-siRNA in C666-1. This selective expression was corroborated in EBV-positive and -negative tumor models. Conclusions/Significance This study demonstrates the feasibility of mc-oriP-IFNγ as a safe and highly effective targeted gene therapeutic system for the treatment of EBV positive NPC.
Collapse
Affiliation(s)
- Yufang Zuo
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Jiangxue Wu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Zumin Xu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Department of Radiation Oncology, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Shiping Yang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Haijiao Yan
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Li Tan
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xiangqi Meng
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xiaofang Ying
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Ranyi Liu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Tiebang Kang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Wenlin Huang
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-Sen University, Guangzhou, People's Republic of China
- Institute of Microbiology, Chinese Academy of Science, Beijing, People's Republic of China
- * E-mail:
| |
Collapse
|
33
|
Wang Y, Osatomi K, Nagatomo Y, Yoshida A, Hara K. Purification, molecular cloning, and some properties of a manganese-containing superoxide dismutase from Japanese flounder (Paralichthys olivaceus). Comp Biochem Physiol B Biochem Mol Biol 2011; 158:289-96. [DOI: 10.1016/j.cbpb.2010.12.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 12/28/2010] [Accepted: 12/29/2010] [Indexed: 11/30/2022]
|
34
|
Abstract
Radiotherapy is used to treat approximately 50% of all cancer patients, with varying success. The dose of ionizing radiation that can be given to the tumour is determined by the sensitivity of the surrounding normal tissues. Strategies to improve radiotherapy therefore aim to increase the effect on the tumour or to decrease the effects on normal tissues. These aims must be achieved without sensitizing the normal tissues in the first approach and without protecting the tumour in the second approach. Two factors have made such approaches feasible: namely, an improved understanding of the molecular response of cells and tissues to ionizing radiation and a new appreciation of the exploitable genetic alterations in tumours. These have led to the development of treatments combining pharmacological interventions with ionizing radiation that more specifically target either tumour or normal tissue, leading to improvements in efficacy.
Collapse
Affiliation(s)
- Adrian C Begg
- Division of Experimental Therapy, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands.
| | | | | |
Collapse
|
35
|
Argyros O, Wong SP, Fedonidis C, Tolmachov O, Waddington SN, Howe SJ, Niceta M, Coutelle C, Harbottle RP. Development of S/MAR minicircles for enhanced and persistent transgene expression in the mouse liver. J Mol Med (Berl) 2011; 89:515-29. [PMID: 21301798 DOI: 10.1007/s00109-010-0713-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 10/29/2010] [Accepted: 12/03/2010] [Indexed: 12/29/2022]
Abstract
We have previously described the development of a scaffold/matrix attachment region (S/MAR) episomal vector system for in vivo application and demonstrated its utility to sustain transgene expression in the mouse liver for at least 6 months following a single administration. Subsequently, we observed that transgene expression is sustained for the lifetime of the animal. The level of expression, however, does drop appreciably over time. We hypothesised that by eliminating the bacterial components in our vectors, we could improve their performance since bacterial sequences have been shown to be responsible for the immunotoxicity of the vector and the silencing of its expression when applied in vivo. We describe here the development of a minimally sized S/MAR vector, which is devoid of extraneous bacterial sequences. This minicircle vector comprises an expression cassette and an S/MAR moiety, providing higher and more sustained transgene expression for several months in the absence of selection, both in vitro and in vivo. In contrast to the expression of our original S/MAR plasmid vector, the novel S/MAR minicircle vectors mediate increased transgene expression, which becomes sustained at about twice the levels observed immediately after administration. These promising results demonstrate the utility of minimally sized S/MAR vectors for persistent, atoxic gene expression.
Collapse
Affiliation(s)
- Orestis Argyros
- Gene Therapy Research Group, Section of Molecular Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Stitt-Fischer MS, Ungerman RK, Wilen DS, Wasserloos K, Renz LM, Raub SE, Peterson J, Pearce LL. Manganese superoxide dismutase is not protective in bovine pulmonary artery endothelial cells at systemic oxygen levels. Radiat Res 2010; 174:679-90. [PMID: 21128791 DOI: 10.1667/rr2062.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Bovine pulmonary artery endothelial cells (BPAEC) are extremely sensitive to oxygen, mediated by superoxide production. Ionizing radiation is known to generate superoxide in oxygenated aqueous media; however, at systemic oxygen levels (3%), no oxygen enhancement is observed after irradiation. A number of markers (cell growth, alamarBlue, mitochondrial membrane polarization) for metabolic activity indicate that BPAEC maintained under 20% oxygen grow and metabolize more slowly than cells maintained under 3% oxygen. BPAEC cultured in 20% oxygen grow better when they are transiently transfected with either manganese superoxide dismutase (MnSOD) or copper zinc superoxide dismutase (CuZnSOD) and exhibit improved survival after irradiation (0.5-10 Gy). Furthermore, X irradiation of BPAEC grown in 20% oxygen results in very diffuse colony formation, which is completely ameliorated by either growth in 3% oxygen or overexpression of MnSOD. However, MnSOD overexpression in BPAEC grown in 3% oxygen provides no further radioprotection, as judged by clonogenic survival curves. Radiation does not increase apoptosis in BPAEC but inhibits cell growth and up-regulates p53 and p21 at either 3% or 20% oxygen.
Collapse
Affiliation(s)
- Molly S Stitt-Fischer
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15219-3138, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Rajagopalan MS, Stone B, Rwigema JC, Salimi U, Epperly MW, Goff J, Franicola D, Dixon T, Cao S, Zhang X, Buchholz BM, Bauer AJ, Choi S, Bakkenist C, Wang H, Greenberger JS. Intraesophageal manganese superoxide dismutase-plasmid liposomes ameliorates novel total-body and thoracic radiation sensitivity of NOS1-/- mice. Radiat Res 2010; 174:297-312. [PMID: 20726721 DOI: 10.1667/rr2019.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The effect of deletion of the nitric oxide synthase 1 gene (NOS1(-/-)) on radiosensitivity was determined. In vitro, long-term cultures of bone marrow stromal cells derived from NOS1(-/-) were more radioresistant than cells from C57BL/6NHsd (wild-type), NOS2(-/-) or NOS3(-/-) mice. Mice from each strain received 20 Gy thoracic irradiation or 9.5 Gy total-body irradiation (TBI), and NOS1(-/-) mice were more sensitive to both. To determine the etiology of radiosensitivity, studies of histopathology, lower esophageal contractility, gastrointestinal transit, blood counts, electrolytes and inflammatory markers were performed; no significant differences between irradiated NOS1(-/-) and control mice were found. Video camera surveillance revealed the cause of death in NOS1(-/-) mice to be grand mal seizures; control mice died with fatigue and listlessness associated with low blood counts after TBI. NOS1(-/-) mice were not sensitive to brain-only irradiation. MnSOD-PL therapy delivered to the esophagus of wild-type and NOS1(-/-) mice resulted in equivalent biochemical levels in both; however, in NOS1(-/-) mice, MnSOD-PL significantly increased survival after both thoracic and total-body irradiation. The mechanism of radiosensitivity of NOS1(-/-) mice and its reversal by MnSOD-PL may be related to the developmental esophageal enteric neuronal innervation abnormalities described in these mice.
Collapse
Affiliation(s)
- Malolan S Rajagopalan
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15232, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Shuvaev VV, Han J, Yu KJ, Huang S, Hawkins BJ, Madesh M, Nakada M, Muzykantov VR. PECAM-targeted delivery of SOD inhibits endothelial inflammatory response. FASEB J 2010; 25:348-57. [PMID: 20876216 DOI: 10.1096/fj.10-169789] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Elevated generation of reactive oxygen species (ROS) by endothelial enzymes, including NADPH-oxidase, is implicated in vascular oxidative stress and endothelial proinflammatory activation involving exposure of vascular cell adhesion molecule-1 (VCAM-1). Catalase and superoxide dismutase (SOD) conjugated with antibodies to platelet/endothelial cell adhesion molecule 1 (PECAM-1) bind specifically to endothelium and inhibit effects of corresponding ROS, H(2)O(2), and superoxide anion. In this study, anti-PECAM/SOD, but not anti-PECAM/catalase or nontargeted enzymes, including polyethylene glycol (PEG)-SOD, inhibited 2- to 3-fold VCAM expression caused by tumor necrosis factor (TNF), interleukin-1β, and lipopolysaccharide. Anti- PECAM/SOD, but not nontargeted counterparts, accumulated in vascular endothelium after intravenous injection, localized in endothelial endosomes, and inhibited by 70% lipopolysaccharide-caused VCAM-1 expression in mice. Anti-PECAM/SOD colocalized with EEA-1-positive endothelial vesicles and quenched ROS produced in response to TNF. Inhibitors of NADPH oxidase and anion channel ClC3 blocked TNF-induced VCAM expression, affirming that superoxide produced and transported by these proteins, respectively, mediates inflammatory signaling. Anti-PECAM/SOD abolished VCAM expression caused by poly(I:C)-induced activation of toll-like receptor 3 localized in intracellular vesicles. These results directly implicate endosomal influx of superoxide in endothelial inflammatory response and suggest that site-specific interception of this signal attained by targeted delivery of anti-PECAM/SOD into endothelial endosomes may have anti-inflammatory effects.
Collapse
Affiliation(s)
- Vladimir V Shuvaev
- Institute for Environmental Medicine, Department of Pharmacology, University of Pennsylvania School of Medicine, 1 John Morgan Bldg., 3620 Hamilton Walk, Philadelphia, PA 19104-6068, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Holley AK, Xu Y, St Clair DK, St Clair WH. RelB regulates manganese superoxide dismutase gene and resistance to ionizing radiation of prostate cancer cells. Ann N Y Acad Sci 2010; 1201:129-36. [PMID: 20649549 PMCID: PMC3107504 DOI: 10.1111/j.1749-6632.2010.05613.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Radiation therapy is in the front line for treatment of localized prostate cancer. However, a significant percentage of patients have radiation-resistant disease. The NF-kappaB pathway is an important factor for radiation resistance, and the classical (canonical) pathway is thought to confer protection of prostate cancer cells from ionizing radiation. Recently, the alternative (non-canonical) pathway, which is involved in prostate cancer aggressiveness, has also been shown to be important for radiation resistance in prostate cancer. The alternative NF-kappaB pathway component RelB protects prostate cancer cells from the detrimental effects of ionizing radiation, in part, by stimulating expression of the mitochondria-localized antioxidant enzyme manganese superoxide dismutase (MnSOD). Blocking RelB activation suppresses MnSOD expression and sensitizes prostate cancer cells to radiation. These results suggest that RelB-mediated modulation of the antioxidant capacity of prostate cancer cells is an important mechanism of radiation resistance. Therefore, targeting RelB activation may prove to be a valuable weapon in the oncologist's arsenal to defeat aggressive and radiation-resistant prostate cancer.
Collapse
Affiliation(s)
- Aaron K Holley
- Graduate Center for Toxicology, University of Kentucky, Lexington, Kentucky 40536, USA
| | | | | | | |
Collapse
|
40
|
Kim A, Joseph S, Khan A, Epstein CJ, Sobel R, Huang TT. Enhanced expression of mitochondrial superoxide dismutase leads to prolonged in vivo cell cycle progression and up-regulation of mitochondrial thioredoxin. Free Radic Biol Med 2010; 48:1501-12. [PMID: 20188820 PMCID: PMC2945707 DOI: 10.1016/j.freeradbiomed.2010.02.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 02/06/2010] [Accepted: 02/19/2010] [Indexed: 02/09/2023]
Abstract
Mn superoxide dismutase (MnSOD) is an important mitochondrial antioxidant enzyme, and elevated MnSOD levels have been shown to reduce tumor growth in part by suppressing cell proliferation. Studies with fibroblasts have shown that increased MnSOD expression prolongs cell cycle transition time in G1/S and favors entrance into the quiescent state. To determine if the same effect occurs during tissue regeneration in vivo, we used a transgenic mouse system with liver-specific MnSOD expression and a partial hepatectomy paradigm to induce synchronized in vivo cell proliferation during liver regeneration. We show in this experimental system that a 2.6-fold increase in MnSOD activity leads to delayed entry into S phase, as measured by reduction in bromodeoxyuridine (BrdU) incorporation and decreased expression of proliferative cell nuclear antigen (PCNA). Thus, compared to control mice with baseline MnSOD levels, transgenic mice with increased MnSOD expression in the liver have 23% fewer BrdU-positive cells and a marked attenuation of PCNA expression. The increase in MnSOD activity also leads to an increase in the mitochondrial form of thioredoxin (thioredoxin 2), but not in several other peroxidases examined, suggesting the importance of thioredoxin 2 in maintaining redox balance in mitochondria with elevated levels of MnSOD.
Collapse
Affiliation(s)
- Aekyong Kim
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Suman Joseph
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Aslam Khan
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Charles J Epstein
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Raymond Sobel
- Department of Pathology, Stanford University, Stanford, CA, USA
- Laboratory Service, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Ting-Ting Huang
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
- GRECC, VA Palo Alto Health Care System, Palo Alto, CA, USA
- Corresponding author: Ting-Ting Huang, Department of Neurology and Neurological Sciences, Stanford University School of Medicine, and GRECC, VA Palo Alto Health Care System, 3801 Miranda Ave. Building 100, D3-101, Palo Alto, CA 94304, USA, Phone 650-496-2581, Fax 650-849-0457
| |
Collapse
|
41
|
Lee JC, Kinniry PA, Arguiri E, Serota M, Kanterakis S, Chatterjee S, Solomides CC, Javvadi P, Koumenis C, Cengel KA, Christofidou-Solomidou M. Dietary curcumin increases antioxidant defenses in lung, ameliorates radiation-induced pulmonary fibrosis, and improves survival in mice. Radiat Res 2010; 173:590-601. [PMID: 20426658 DOI: 10.1667/rr1522.1] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The effectiveness of lung radiotherapy is limited by radiation tolerance of normal tissues and by the intrinsic radiosensitivity of lung cancer cells. The chemopreventive agent curcumin has known antioxidant and tumor cell radiosensitizing properties. Its usefulness in preventing radiation-induced pneumonopathy has not been tested previously. We evaluated dietary curcumin in radiation-induced pneumonopathy and lung tumor regression in a murine model. Mice were given 1% or 5% (w/w) dietary curcumin or control diet prior to irradiation and for the duration of the experiment. Lungs were evaluated at 3 weeks after irradiation for acute lung injury and inflammation by evaluating bronchoalveolar lavage (BAL) fluid content for proteins, neutrophils and at 4 months for pulmonary fibrosis. In a separate series of experiments, an orthotopic model of lung cancer using intravenously injected Lewis lung carcinoma (LLC) cells was used to exclude possible tumor radioprotection by dietary curcumin. In vitro, curcumin boosted antioxidant defenses by increasing heme oxygenase 1 (HO-1) levels in primary lung endothelial and fibroblast cells and blocked radiation-induced generation of reactive oxygen species (ROS). Dietary curcumin significantly increased HO-1 in lungs as early as after 1 week of feeding, coinciding with a steady-state level of curcumin in plasma. Although both 1% and 5% w/w dietary curcumin exerted physiological changes in lung tissues by significantly decreasing LPS-induced TNF-alpha production in lungs, only 5% dietary curcumin significantly improved survival of mice after irradiation and decreased radiation-induced lung fibrosis. Importantly, dietary curcumin did not protect LLC pulmonary metastases from radiation killing. Thus dietary curcumin ameliorates radiation-induced pulmonary fibrosis and increases mouse survival while not impairing tumor cell killing by radiation.
Collapse
Affiliation(s)
- James C Lee
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, University of Pennsylvania Medical Center, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Faurez F, Dory D, Le Moigne V, Gravier R, Jestin A. Biosafety of DNA vaccines: New generation of DNA vectors and current knowledge on the fate of plasmids after injection. Vaccine 2010; 28:3888-95. [DOI: 10.1016/j.vaccine.2010.03.040] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Revised: 03/09/2010] [Accepted: 03/21/2010] [Indexed: 12/16/2022]
|
43
|
Epperly MW, Lai SY, Kanai AJ, Mason N, Lopresi B, Dixon T, Franicola D, Niu Y, Wilson WR, Greenberger JS. Effectiveness of combined modality radiotherapy of orthotopic human squamous cell carcinomas in Nu/Nu mice using cetuximab, tirapazamine and MnSOD-plasmid liposome gene therapy. In Vivo 2010; 24:1-8. [PMID: 20133969 PMCID: PMC2899489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Hypoxic regions limit the radiocontrollability of head and neck carcinomas. Whether or not combinations of plasmid/liposome mediated overexpression of normal tissue protective manganese superoxide dismutase (MnSOD), cetuximab (C225), and the hypoxic cytotoxin tirapazamine (TPZ) enhanced radiotherapeutic effects was tested in a CAL-33 orthotopic mouse cheek tumor model. The tumor volume continued to increase in the control (untreated) mice, with a ninefold increase by 10 days when the tumors exceeded 2 cm(3). The mice receiving 14 Gy only showed reduced tumor growth to 3.1+/-0.1 fold at day 10. The mice receiving MnSOD-PL, C225, TPZ plus 14 Gy had the best outcome with 0.7+/-0.1 fold increase in tumor volume by 10 days (p=0.015) compared to irradiation only. The addition of MnSOD-PL, TPZ, and C225 to irradiation optimized the therapeutic ratio for the local control of hypoxic region-containing CAL-33 orthotopic tumors.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal, Humanized
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/therapy
- Cell Line, Tumor
- Cetuximab
- Dose-Response Relationship, Radiation
- Genetic Therapy
- Humans
- Liposomes
- Mice
- Mice, Nude
- Mouth Neoplasms/metabolism
- Mouth Neoplasms/pathology
- Mouth Neoplasms/therapy
- Nitric Oxide/metabolism
- Radiation-Sensitizing Agents/administration & dosage
- Radiotherapy, Adjuvant
- Superoxide Dismutase/genetics
- Tirapazamine
- Triazines/administration & dosage
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Michael W Epperly
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kagan VE, Wipf P, Stoyanovsky D, Greenberger JS, Borisenko G, Belikova NA, Yanamala N, Samhan Arias AK, Tungekar MA, Jiang J, Tyurina YY, Ji J, Klein-Seetharaman J, Pitt BR, Shvedova AA, Bayır H. Hydrogen sulfide is a reversible inhibitor of the NADH oxidase activity of synaptic plasma membranes. Adv Drug Deliv Rev 2009; 388:718-722. [PMID: 19695225 PMCID: PMC2784017 DOI: 10.1016/j.addr.2009.06.008] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Accepted: 08/13/2009] [Indexed: 10/20/2022]
Abstract
Hydrogen sulfide is now accepted as a neuromodulator, which can be involved in neuronal defence against oxidative stress insults in the brain. In this work we show that concentrations of H(2)S within the physiological range reported in the brain produce a reversible inhibition of the NADH oxidase activity and coupled superoxide anion production by synaptic plasma membranes from rat brain. At physiological pH 7 the concentration of H(2)S needed for 50% inhibition of the NADH oxidase activity is 5+/-1 microM, which is within the low range of the reported physiological H(2)S concentrations. Thus, the NADH oxidase activity of the neuronal plasma membrane can act as a sensor of local H(2)S depletion in neurones. H(2)S inhibition of the NADH oxidase activity of the neuronal plasma membrane can be accounted for direct reduction by H(2)S of cytochrome b(5). However, H(2)S fails to afford a significant protection against the inhibition of this activity by peroxynitrite. In conclusion, our results point out that H(2)S is more potent as inhibitor of reactive oxygen species formation than as a sacrificial antioxidant.
Collapse
Affiliation(s)
- Valerian E. Kagan
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Detcho Stoyanovsky
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Joel S. Greenberger
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | | | - Natalia A. Belikova
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Naveena Yanamala
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Alejandro K. Samhan Arias
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Muhammad A. Tungekar
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Jianfei Jiang
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Yulia Y. Tyurina
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Jing Ji
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | | | - Bruce R. Pitt
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| | - Anna A Shvedova
- Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, and West Virginia University, Morgantown, WV, 26505, USA
| | - Hülya Bayır
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, 15219, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA
| |
Collapse
|
45
|
Pollard JM, Reboucas JS, Durazo A, Kos I, Fike F, Panni M, Gralla EB, Valentine JS, Batinic-Haberle I, Gatti RA. Radioprotective effects of manganese-containing superoxide dismutase mimics on ataxia-telangiectasia cells. Free Radic Biol Med 2009; 47:250-60. [PMID: 19389472 PMCID: PMC3592562 DOI: 10.1016/j.freeradbiomed.2009.04.018] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2008] [Revised: 03/30/2009] [Accepted: 04/13/2009] [Indexed: 01/01/2023]
Abstract
We tested several classes of antioxidant manganese compounds for radioprotective effects using human lymphoblastoid cells: six porphyrins, three salens, and two cyclic polyamines. Radioprotection was evaluated by seven assays: XTT, annexin V and propidium iodide flow cytometry analysis, gamma-H2AX immunofluorescence, the neutral comet assay, dichlorofluorescein and dihydroethidium staining, resazurin, and colony survival assay. Two compounds were most effective in protecting wild-type and A-T cells against radiation-induced damage: MnMx-2-PyP-Calbio (a mixture of differently N-methylated MnT-2-PyP+ from Calbiochem) and MnTnHex-2-PyP. MnTnHex-2-PyP protected WT cells against radiation-induced apoptosis by 58% (p = 0.04), using XTT, and A-T cells by 39% (p = 0.01), using annexin V and propidium iodide staining. MnTnHex-2-PyP protected WT cells against DNA damage by 57% (p = 0.005), using gamma-H2AX immunofluorescence, and by 30% (p < 0.01), using neutral comet assay. MnTnHex-2-PyP is more lipophilic than MnMx-2-PyP-Calbio and is also >10-fold more SOD-active; consequently it is >50-fold more potent as a radioprotectant, as supported by six of the tests employed in this study. Thus, lipophilicity and antioxidant potency correlated with the magnitude of the beneficial radioprotectant effects observed. Our results identify a new class of porphyrinic radioprotectants for the general and radiosensitive populations and may also provide a new option for treating A-T patients.
Collapse
Affiliation(s)
- Julianne M Pollard
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Weiss JF, Landauer MR. History and development of radiation-protective agents. Int J Radiat Biol 2009; 85:539-73. [DOI: 10.1080/09553000902985144] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
47
|
Grdina DJ, Murley JS, Kataoka Y, Baker KL, Kunnavakkam R, Coleman MC, Spitz DR. Amifostine induces antioxidant enzymatic activities in normal tissues and a transplantable tumor that can affect radiation response. Int J Radiat Oncol Biol Phys 2009; 73:886-96. [PMID: 19215822 DOI: 10.1016/j.ijrobp.2008.10.061] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Revised: 10/03/2008] [Accepted: 10/08/2008] [Indexed: 01/24/2023]
Abstract
PURPOSE To determine whether amifostine can induce elevated manganese superoxide dismutase (SOD2) in murine tissues and a transplantable SA-NH tumor, resulting in a delayed tumor cell radioprotective effect. METHODS AND MATERIALS SA-NH tumor-bearing C3H mice were treated with a single 400 mg/kg or three daily 50 mg/kg doses of amifostine administered intraperitoneally. At selected time intervals after the last injection, the heart, liver, lung, pancreas, small intestine, spleen, and SA-NH tumor were removed and analyzed for SOD2, catalase, and glutathione peroxidase (GPx) enzymatic activity. The effect of elevated SOD2 enzymatic activity on the radiation response of SA-NH cells was determined. RESULTS SOD2 activity was significantly elevated in selected tissues and a tumor 24 h after amifostine treatment. Catalase and GPx activities remained unchanged except for significant elevations in the spleen. GPx was also elevated in the pancreas. SA-NH tumor cells exhibited a twofold elevation in SOD2 activity and a 27% elevation in radiation resistance. Amifostine administered in three daily fractions of 50 mg/kg each also resulted in significant elevations of these antioxidant enzymes. CONCLUSIONS Amifostine can induce a delayed radioprotective effect that correlates with elevated levels of SOD2 activity in SA-NH tumor. If limited to normal tissues, this delayed radioprotective effect offers an additional potential for overall radiation protection. However, amifostine-induced elevation of SOD2 activity in tumors could have an unanticipated deleterious effect on tumor responses to fractionated radiation therapy, given that the radioprotector is administered daily just before each 2-Gy fractionated dose.
Collapse
Affiliation(s)
- David J Grdina
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Greenberger JS. Radioprotection. In Vivo 2009; 23:323-36. [PMID: 19414422 PMCID: PMC2981866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Over 40% of cancer patients will require radiation therapy during management of their disease. Although radiation therapy improves the survival of a significant number of cancer patients, both acute radiation toxicity (which manifests during a course of clinical radiotherapy or shortly thereafter), and late toxicity (developing months to years after completion of radiotherapy) compromise overall outcomes for successfully treated cancer patients.
Collapse
Affiliation(s)
- Joel S Greenberger
- Department of Radiation Oncology, University of Pittsburgh Medical Center, 200 Lothrop Street, Pittsburgh, PA 15213, USA.
| |
Collapse
|
49
|
Gill DR, Pringle IA, Hyde SC. Progress and prospects: the design and production of plasmid vectors. Gene Ther 2009; 16:165-71. [PMID: 19129858 DOI: 10.1038/gt.2008.183] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Plasmid DNA (pDNA) expression vectors are fundamental to all forms of non-viral gene transfer. In this review, we discuss principles of pDNA design and production including the impact of bacterially derived sequences on transgene expression and minicircle approaches to minimize their effects. The impact of inclusion of DNA elements such as scaffold matrix attachment regions (S/MARs), transcription factor (TF)-binding sites and tissue-specific promoters are described. The benefits of eliminating CG dinucleotides (CpGs) from the pDNA are also considered.
Collapse
Affiliation(s)
- D R Gill
- Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | | | | |
Collapse
|