Figure 1 - uploaded by Irina Zabbarova
Content may be subject to copyright.
Schematic depicting the inner mitochondrial membrane and the five subunits of the mitochondrial electron transport chain 

Schematic depicting the inner mitochondrial membrane and the five subunits of the mitochondrial electron transport chain 

Source publication
Article
Full-text available
Adverse effects of ionizing radiation are mediated through reactive oxygen and nitrogen species. Mitochondria are the principal source of these species in the cell and play an important role in irradiation-induced apoptosis. The use of free radical scavengers and nitric oxide synthase inhibitors has proven to protect normal tissues and, in some cas...

Contexts in source publication

Context 1
... development of agents that protect tissues against radiation damage (i.e., radioprotective agents) is currently the subject of intense research on at least two broad accounts. First, radiotherapy remains one of the most widely used treatments for cancer. Irradiation-induced DNA damage can halt tumor cell proliferation, but collateral radiation damage to surrounding tissues is always a concern. Accordingly, there is a need to develop drugs that will protect healthy cells while leaving malignant cells susceptible—and ideally, even sensitized—to radiation therapy. An additional impetus for research is the need to counteract occupational risks and terrorist threats of radiation exposure. Damage to DNA, the primary target of radiation treatment, can occur directly, but most genetic damage is mediated by reactive oxygen and nitrogen species (ROS and RNS). Hence, scavengers of free radicals form the principal group of radioprotective agents. It has been hypothesized that irradiation produces bursts of ROS [e.g., superoxide (O 2 − ) and hydroxyl (OH) radicals] by reacting with the aqueous environment of the cell. However, recent findings suggest that the principal origin of irradiation-induced free radicals is the mitochondria (1,2). Accordingly, antagonists of nitric oxide synthase (NOS) are also of interest for potential radioprotective measures. A significant challenge to the use of radio-protective agents will be to deliver them through biological membranes and accumulate them at effective concentrations within mitochondrial domains where ROS and NOS are generated. The cellular response to irradiation is complex and includes genomic instability. Bystander effects have been frequently observed at low doses and show a non-linear response (3). The genotype and phenotype of the irradiated cell or animal as well as the nature of irradiation determine cellular response to irradiation (4). Cell types that are especially sensitive to irradiation in different organs are listed in Table 1. Damage to DNA (e.g., double-strand breaks) triggers multiple signaling events, the variety of which extends to ataxia telangiectasia mutated (ATM) and Rad3-related protein; ERBB family and other tyrosine kinases (5,6); protein kinase C; extracellular signal-regulated kinase 1/2 (ERK1/2) (7,8); and increased production of ceramide (9). The initial cytotoxicity of irradiation, however, is believed to result from ROS and RNS generation (10). Ionizing radiation produces a burst of O 2 − in the cell within 10 − 13 seconds of radiation exposure. This free radical burst can damage DNA and proteins including mitochondrial respiratory complexes. Theoretical calculations suggest, however, that the initial ROS [i.e., O 2 − and its dismutation product hydrogen peroxide (H 2 O 2 )] formed during a clinical radiation dose [1–2 Gray (1 Gy = 1 J/kg = 100 rad)] may be etiologically insignificant. Indeed, much evidence suggests that ionizing radiation directly alters mitochondrial metabolism (11, 12) and transiently opens the unregulated mitochondrial permeability transition pore (MPTP). In particular, the opening of the MPTP causes an influx of Ca 2+ into the mitochondrial matrix, thereby activating mitochondrial nitric oxide synthase (mtNOS). The resulting nitric oxide (NO) inhibits the respiratory chain (Figure 1), thereby generating large amounts O 2 − that react with NO to form highly reactive peroxynitrite (ONO − ). Radioprotective agents must manifest one or more of the following properties: antioxidant avtivity, NOS inhibition, anti-inflammatory or immunomodulatory activity (Table 2). Most of these existing drugs can neutralize ROS by a number of different mechanisms. As introduced above, the utility of radioprotective agents may rest on strategies for delivering them to the mitochondrion; features related to these strategies are presented in Table 3. Aminofostine is the most prominent compound of this class of drugs. The prodrug (WR2721) accumulates preferentially in the kidneys and salivary glands, where it is metabolized to the active WR1065 (13). It scavenges O 2 − as well as OH and lipoperoxyl (LOO) radicals (14). Aminofostine is protective in normal but not tumor cells, reportedly due to higher alkaline phosphatase activity, pH, and oxygenation in normal tissue. Aminofostine was approved by the Food and Drug Administration for patients undergoing radiotherapy for ovarian and head and neck cancers, but its intravenous administration can cause nausea, vomiting, and hypotension, which has limited its use. Studies are in progress to evaluate the effects of alternative routes of delivery (15). Superoxide dismutases (SODs) are important components of the cellular antioxidant system, catalyzing the dismutation of O 2 − to O 2 and H 2 O 2 (16). There are three isoforms in mammalian cells that may vary with respect to the specific redox-active metal ion necessary for catalysis. The mitochondrial isoform of SOD (MnSOD or SOD2) is manganese-dependent and is called mitochondrial SOD. A specific N-terminal leader sequence directs the polypeptide to mitochondria and is then cleaved; MnSOD devoid of this leader sequence localizes to the cytosol and is not radioprotective (1,2). Mitochondrial localization of MnSOD has been shown to be associated with decreased irradiation-induced apoptosis (17). Nitroxides are low molecular-weight compounds containing tertiary amine functional groups that are oxidized (R 3 N + -O − ) to form relatively stable free radicals. They were initially used as probes for ESR spectroscopy and more recently were shown to have antioxidant activity. Many of them are water-soluble and act, as superoxide dismutase mimetics, to neutralize O 2 − and OH and organic hydroperoxides. Although nitroxides are cell membrane–permeable, they may not get into mitochondria. Using mass spectroscopy, we have shown that the nitroxide 4-amino- TEMPO is unable to penetrate mitochondrial membranes and requires targeting for effective antioxidant activity (18). Despite these findings, some nitroxides, including TEMPO, show radioprotective properties in vitro and in vivo in mice (when administered ...
Context 2
... is experimental and clinical evidence that the level of vitamins is reduced in irradiated tissue. Supplemental diets including high doses of vitamins A, C, and E are protective in normal tissues during radiation therapy [reviewed in (14), (20)]. Moreover, when present in high doses throughout the treatment period, these vitamins have been shown to be radiosensitizing in some types of cancer cells. In this regard, it is thought that these vitamins each have a distinct mechanism of action, and thus a mixture of them is more effective (20). Although high doses of vitamins are necessary for beneficial effects, toxicity limits their use to particular organs. High dose–vitamin C can cause diarrhea; vitamin E is associated with defective blood clotting; and vitamin A is linked to skin bronzing. Melatonin ( N -acetyl-5-methoxytryptamine) is an endogenous compound synthesized by the pineal gland and released into the bloodstream (21). It is a potent antioxidant and can directly scavenge OH radicals as well as increase the activity of enzymes such as SOD and glutathione peroxidase (GPX). Melatonin decreases the activity of NOS and is radioprotective and nontoxic in vitro and in vivo [reviewed in (21)]. Melatonin is also reported to reduce tumor cell growth by inducing apoptosis or reducing invasiveness, underscoring its clinical potential. Although they may not be generally considered first and foremost as radioprotective agents, NOS antagonists (e.g., L -NMMA, AMT) have been shown to prevent radiation cystitis more effectively than MnSOD transgene therapy (22). Their intravesical administration protects the umbrella cell layer of the urinary bladder from superficial ulcerations, thereby preventing changes in trans-epithelial resistance and permeability to water or urine. NOS antagonists may prevent both NO and O 2 − production, whereas the scavenging and dismutation of O 2 − produces H 2 O 2 , which must be neutralized by other antioxidant enzymes, such as GPX and catalase (Figure 1). Since systemic administration of NOS inhibitors can cause hypertension, targeting them to mitochondria is therapeutically relevant. Inflammation can be a direct effect of irradiation as well as a secondary effect caused by oxidative stress. An acute inflammatory response involves activation of stress-sensitive kinases and transcription factors and the production of inflammatory cytokines (23). Because the chronic overexpression of cytokines and growth factors can result in fibrosis or necrosis, anti- inflammatory agents are used for radioprotection. Among these is palifermin, a recombinant human keratinocyte growth factor. This agent can stimulate cellular proliferation and differentiation in epithelial cells and enhance intrinsic glutathione peroxidase activity (13). Another radioprotective agent in this category is resveratrol (polyphenolic phytoalexin), which prosesses potent anti-oxidant, anti-inflammatory and anti-proliferative properties. Resveratrol has been shown to protect keratinocytes from UVB radiation–induced cell death by inhibiting caspases (24). Hematopoietic cells have a high turnover rate and are thus extremely sensitive to irradiation. The immune system is affected and suppressed at relatively low doses of acute irradiation, reducing bone marrow cell production as well as inducing apoptosis in mature cells (25). Accordingly, compounds that regulate cytokine production can protect against irradiation damage and bring about repair through enhanced production of bone marrow cells, lymphocytes, platelets, and circulating granulocytes. Finally, radioprotective properties have been attributed to propolis (26), PR-1 (an extract of Podophylium hexandrum rhizomes) (27), and a number of other immunomodulatory agents. To be properly recognized and imported into mitochondria, translated proteins require an N- terminal specific amino acid sequence (28,29). The mitochondrial signal peptide can be experimentally linked to non-mitochondrial proteins to promote their uptake into the mitochondrial matrix; the mitochondrial protein import machinery includes the translocase of the outer membrane (TOM) complex and the translocase of the inner mitochondrial membrane (TIM) complexes (30). Proteins interacting with the TIM complexes are either integrated into the inner mitochondrial membrane or transported into the mitochondrial matrix and processed by the mitochondrial processing peptidase (MPP). Less frequently, proteins may be recognized by a C-terminal sequence consisting of twenty to thirty residues. Certain proteins encode the necessary recognition elements as parts of their primary sequence, in which case import occurs with minimal processing (31). MnSOD gene therapy strategies for irradiation protection may be based on administration of a plasmidal MnSOD-encoding transgene carried within liposomes or adenoviruses. Intratracheal injections of either MnSOD-endocing plasmids or adenoviruses were shown to be protective against total lung irradiation in a mouse model (32–34); oral administration protected the mouse esophagus from irradiation-induced esophagitis (35) (Figure 2A) and prevented oral cavity mucositis (36). Finally, intravesical instillation of MnSOD-encoding plasmid DNA twenty-four hours prior to irradiation protected bladders from radiation cystitis (Figure 2B) (37). Several classes of cell-permeable antioxidant peptides that permeate into the inner mitochondria membrane have recently been used as targeting systems. One of the classes, known as Szeto-Schiller (SS) peptides [reviewed in (38,39)], comprises tetrapeptides of alternating aromatic and basic residues. Although these peptides are cationic, they localize to the inner membrane rather than the matrix. Accordingly, their uptake is independent of the mitochondrial membrane potential and they concentrate rapidly (1000–5000-fold after two minutes) within mitochondria (40). The antioxidant properties of SS peptides do not appear to be derived from the specific tetrapeptide sequence, although scavenging activity is highly dependent upon the inclusion of a dimethyltyrosine (DMT) residue. For example, DMT- D -Arg-Phe-Lys-NH 2 scavenges as well as D -Arg-DMT-Lys-Phe-NH 2 ; substitution of DMT with phenylalanine, however, abolishes scavenging activity (40). SS peptides have been shown to inhibit lipid peroxidation by scavenging OH radicals (40). They can reduce both spontaneous and Antimycin A–induced mitochondrial H 2 O 2 production without uncoupling mitochondria (39). They also inhibit mitochondrial permeability transition and swelling (induced by calcium overload or inhibitors of the mitochondrial electron transport chain) (38). SS peptides are not cytotoxic and are effective at concentrations ranging from 0.1–100 nM (40,41). Another class of potential-independent peptides have been prepared through derivatization of the antibiotic gramicidin S (31). These gramicidin S analogs were used to target mitochondria with TEMPO (the SOD ...
Context 3
... particular, the opening of the MPTP causes an influx of Ca 2+ into the mitochondrial matrix, thereby activating mitochondrial nitric oxide synthase (mtNOS). The resulting nitric oxide (NO) inhibits the respiratory chain (Figure 1), thereby generating large amounts O 2 − that react with NO to form highly reactive peroxynitrite (ONO 2 − ). ...
Context 4
... intravesical administration protects the umbrella cell layer of the urinary bladder from superficial ulcerations, thereby preventing changes in trans-epithelial resistance and permeability to water or urine. NOS antagonists may prevent both NO and O 2 − production, whereas the scavenging and dismutation of O 2 − produces H 2 O 2 , which must be neutralized by other antioxidant enzymes, such as GPX and catalase ( Figure 1). Since systemic administration of NOS inhibitors can cause hypertension, targeting them to mitochondria is therapeutically relevant. ...

Similar publications

Article
Full-text available
The effect of radioprotector indralin on the graft-versus-host reaction was studied on the model of acute GVH disease induced in mice by intraperitoneal transplantation of 40x10(6)semiallogenic splenocytes. The effect was evaluated by animal mortality from GVH disease. Recipients were male F1(CBAxC57Bl/6) mice exposed to 7 Gy 24 h before transplant...
Article
Full-text available
Oxidative stress has been the object of considerable biological and biochemical investigation. Quantification has been difficult although the quantitative level of products of biological oxidations in tissues and tissue products has emerged as a widely used technique. The relationship between these products and the amount of oxidative stress is les...
Article
Full-text available
Gold nanoparticles (AuNPs) could serve as potential radiotherapy sensitizers because of their exceptional biocompatibility and high-Z material nature; however, since in vitro and in vivo behaviors of AuNPs are determined not only by their particle size but also by their surface chemistries, whether surface ligands can affect their radiosensitizatio...
Article
Full-text available
Snow interception in a coniferous forest canopy is an important hydrological feature, producing complex mass and energy exchanges with the surrounding atmosphere and the snowpack below. Subcanopy snowpack accumulation and ablation depends on the effects of canopy architecture on meteorological conditions and on interception storage by stems, branch...
Conference Paper
Full-text available
Soft-tissue sarcoma is a disease having profound social impacts on patients of all ages. Conventional in vitro 2D models are insufficient to study the effect of radiotherapy as well as chemotherapy. Here, we develop a microfluidic device to form and culture soft-tissue sarcoma spheroids to study the combined effect of radiotherapy and chemotherapy....

Citations

... Принципиально новым интересом в поиске средств модификации стохастических последствий может быть система трансдукции сигнала в ответ на повреждение ДНК в интересах стимуляции апоптотического очищения от клеток с нестабильным потенциально канцерогенным геномом [139,[206][207][208], в отличие от ингибирования апоптоза при защите от детерминированных эффектов в больших дозах [101,116,174]. При этом в последние годы исследования радиопротекторов как раз и проводят в плане ингибирования механизма апоптоза [29,50,51,174]. ...
Article
Purpose: Review and systematization of data on the development of studies of drugs intended for protection against radiation in medium doses, assessment of the latest proposals and directions of pharmacological influence on radiation effects, incitement to discussion on the issue under consideration. Results: An analysis of domestic and foreign literature for the period of the 70–90s of the twentieth century and the latest period in relation to the prevention of tissue reactions and long-term effects of low-dose low-power irradiation was carried out, classes (groups) and samples of pharmaceuticals used were considered. Conclusions: 1. Very diverse previously proposed pharmaceuticals are still suitable, acting on various mechanisms of the genesis of the consequences of exposure to medium doses. 2. In recent years, substances (drugs) with receptor action, including genetically engineered products, as well as gene therapy agents, have been of primary interest, but they have been little studied as the means of choice due to their status as demonstration samples for use at medium doses. 3. Drugs of choice, made on the basis of many considerations over 30 years ago, until a replacement not be found.
... Besides the above-mentioned radioprotectors, and as briefly described in the Introduction, TEMPO and its derivatives ( Figure 1) have also been investigated as powerful ROS scavengers [11,[30][31][32]. However, as all low molecular weight (MWM) molecules, the TEMPO derivatives are rapidly eliminated and/or degraded upon administration whatever the route used. ...
... The use of nanotechnology in delivering radioprotective drugs significantly improved their blood circulation time and biodistribution/tissue residence time. In most of the studies conducted, the nanoparticles were administered several hours (1 to 24 h) before exposure to ionizing radiation [20][21][22][23][24][25][26][27][28][29][30][31][32][33][34]. Intravenous, intraperitoneal, and subcutaneous routes are preferred, mainly to escape the harsh environment of the gastrointestinal system and to deliver a significant amount of nanoparticles into the blood. ...
Article
Full-text available
The search for potent radiation-protective drugs for clinical use continues. Studies have, so far, focused on targeting the neutralization of radiation-generated reactive oxygen species (ROS) to protect the cells against the deleterious effects of exposure to ionizing radiation. However, the development of efficacious radioprotective drugs, which are mostly low molecular weight (LMW) compounds, is mainly limited due to their inherent toxicity and rapid excretion from the body. Thus, researchers reformulated these LMW compounds into nano-based formulations. This review discusses recent advances in the use of self-assembling redox nanoparticles as a new group of radioprotective agents. The copolymer micelle (herein referred to as redox nanoparticles; RNP) contains an active part, amino-TEMPO, that effectively scavenges radiation-induced ROS in the body, as demonstrated in vivo. With the use of nanoparticle-based technologies, optimized formulations of these LMW ROS-neutralizers lead to the significant reduction of its toxicity, high bioavailability and longer blood circulation, which consequently resulted in its notable enhanced efficacy (for example, increased survival rate, reduced radiation-induced syndromes and organ damage) against the damaging effects of ionizing radiation. Consistent with the available data on the use of RNP and other nano-based radioprotective agents, it can be concluded that the inherent ROS-targeting activity of a drug intended for radiation protection is as vital as its bioavailability in the specific tissues and organs, where the short-lived ROS are produced during radiation exposure. In this review article, we summarized the current status of the development of radioprotective agents, including our self-assembling radioprotective agents.
... In addition, downregulation of BCS1L was associated with a decrease of complex III activity and oxidation in the mitochondrial respiratory chain in PBMC of patients with RT (Hsiao et al., 2018). RT targets rapidly dividing mitotic cells, inducing molecular-genetic damage to mitochondria (Karim et al., 2016;Zabbarova & Kanai, 2008), which might contribute to acute and chronic CRF associated with RT. Several limitations are recognized, including the lack of measurement of other variables that might affect CRF, such as cognitive function, sleep disturbance, physical activity, and diet. ...
Article
Background: Cancer-related fatigue (CRF) is a highly prevalent, debilitating, and persistent symptom experienced by patients receiving cancer treatments. Up to 71% of men with prostate cancer receiving radiation therapy experience acute and persistent CRF. There is neither an effective therapy nor a diagnostic biomarker for CRF. This pilot study aimed to discover potential biomarkers associated with chronic CRF in men with prostate cancer receiving radiation therapy. Methods: We used a longitudinal repeated-measures research design. Twenty men with prostate cancer undergoing radiation therapy completed all study visits. CRF was evaluated by a well-established and validated questionnaire, the Patient-Reported Outcomes Measurement Information System for Fatigue (PROMIS-F) Short Form. In addition, peripheral blood mononuclear cells were harvested to quantify ribonucleic acid (RNA) gene expression of mitochondria-related genes. Data were collected before, during, on completion, and 24 months postradiation therapy and analyzed using paired t-tests and repeated-measures analysis of variance. Results: The mean of the PROMIS-F T score was significantly increased over time in patients with prostate cancer, remaining elevated at 24 months postradiation therapy compared to baseline. A significant downregulated BC1 ubiquinol-cytochrome c reductase synthesis-like (BCS1L) was observed over time during radiation therapy and at 24 months postradiation therapy. An increased PROMIS-F score was trended with downregulated BCS1L in patients 24 months after completing radiation therapy. Discussion: This is the first evidence to describe altered messenger RNA for BCS1L in chronic CRF using the PROMIS-F measure with men receiving radiation therapy for prostate cancer. Conclusion: Our results suggest that peripheral blood mononuclear cell messenger RNA for BCS1L is a potential biomarker and therapeutic target for radiation therapy-induced chronic CRF in this clinical population.
... In recent years, it has been shown that cellular oxidative stress plays an important role in radiation-induced lung injury (RILI), and thus, oxidative stress can be used as a monitoring index for the development, progression and prognosis of RILI [5]. A key factor in determining the state of oxidative stress is the presence of reactive oxygen species (ROS), which can interact with bases, ribose and phosphodiester linkages and can cause deoxyribonucleic acid (DNA) damage and subsequent apoptosis [6]. A growing number of studies have demonstrated that mitochondria are important sites for ROS production in cells, and that the exact location of the production is the substrate end of the respiratory chain. ...
Article
Full-text available
The purpose of this study was to set up a beagle dog model, for radiation-induced lung injury, that would be able to supply fresh lung tissues in the different injury phases for research into oxidative stress levels and mitochondrial gene expression. Blood serum and tissues were collected via CT-guided core needle biopsies from dogs in the various phases of the radiation response over a 40-week period. Levels of reactive oxygen species (ROS) and manganese superoxide dismutase 2 (MnSOD) protein expression in radiation-induced lung injury were determined by in situ immunocytochemistry; malondialdehyde (MDA) content and reductase activity in the peripheral blood were also tested; in addition, the copy number of the mitochondrial DNA and the level of function of the respiratory chain in the lung tissues were assessed. ROS showed dynamic changes and peaked at 4 weeks; MnSOD was mainly expressed in the Type II alveolar epithelium at 8 weeks; the MDA content and reductase activity in the peripheral blood presented no changes; the copy numbers of most mitochondrial genes peaked at 8 weeks, similarly to the level of function of the corresponding respiratory chain complexes; the level of function of the respiratory chain complex III did not peak until 24 weeks, similarly to the level of function of the corresponding gene Cytb. Radiation-induced lung injury was found to be a dynamically changing process, mainly related to interactions between local ROS, and it was not associated with the levels of oxidative stress in the peripheral blood. Mitochondrial genes and their corresponding respiratory chain complexes were found to be involved in the overall process.
... According to Kuntic et al. (2013), after ionizing radiation, ROS by inducing lipid peroxidation produce cytotoxic aldehydes resulting in inflammatory reactions. This eventually leads to increased synthesis of cytokines, infiltration of mononuclear cells and cellular death (Jamalludin et al., 2007;Zabbarova and Kanai, 2008). In accordance with this, in our experiment the presence of mononuclear cells and fibroblasts was decreased in AMI-protected rats, while necrotic cells in the heart, liver and kidney were rare compared with IR-only treated group. ...
... disease, can shed light on how ROS may cause or relate to disease, thereby leading to the discovery of new therapeutic agents ( Fig. 3) [53,54]. ...
Article
Reperfusion injury is an inherent response to the restoration of blood flow after ischemia. It is a complex process involving numerous mechanisms occurring in the intracellular and extracellular environments, and it is mediated in part by reactive oxygen species (ROS). The imbalance between the cellular formation of free radicals and cells' capacity to defend against them can cause cardiac tissue injuries. In this context, ROS play an essential role in both the organ injury and repair processes. After reperfusion, infiltration into the myocardium of inflammatory leucocytes, such as macrophages and neutrophils, causes further ROS production beyond the initiation of the inflammatory cascade. In this case, ROS overproduction is crucial in cardiac injury, and it can increase the complications related to cardiac reperfusion. In myocardial tissue, ROS can be produced from several sources, such as xanthine oxidase, cytochrome oxidase, cyclooxygenase, mediated unsaturated fatty acid oxidation, oxidation of catecholamines, mitochondrial oxidation, activation of leukocyte nicotinamide adenine dinucleotide phosphate oxidase, iron release, and reduction?oxidation reaction cycling; all of these sources reduce molecular oxygen in the reperfused myocardium. This review discusses about the molecular and therapeutic aspects of cardiac-reperfusion injuries generated by ROS. Experimental and clinical evidence with respect to the use of ischemic preconditioning, Ca2 +, nitric oxide, and conventional antioxidants in cardiac-reperfusion injury are summarized, and causal therapy approaches with various antioxidants are discussed.
... According to Kuntic et al. (2013), after ionizing radiation, ROS by inducing lipid peroxidation produce cytotoxic aldehydes resulting in inflammatory reactions. This eventually leads to increased synthesis of cytokines, infiltration of mononuclear cells and cellular death (Jamalludin et al., 2007;Zabbarova and Kanai, 2008). In accordance with this, in our experiment the presence of mononuclear cells and fibroblasts was decreased in AMI-protected rats, while necrotic cells in the heart, liver and kidney were rare compared with IR-only treated group. ...
Article
Full-text available
Fullerenol C60(OH)24 nanoparticles (FNP) are promising radioprotectors in prevention of early and late ionizing radiation injury. The aim of this study was to compare the efficacy of FNP and amifostine (AMI) in protection of rats exposed to whole-body X-ray irradiation (7 or 8 Gy). Both compounds (FNP, 100 mg/kg ip; AMI, 300 mg/kg ip) were given 30 min before irradiation throughout the study. The general radioprotective efficacy of FNP and AMI were evaluated in rats irradiated with an absolutely lethal dose of X-rays (8 Gy) and their survival were monitored during the period of 30 days after irradiation. Both compounds were of comparable efficacy. Tissue-protective effects of tested compounds were assessed in rats irradiated with an sublethal dose of X-rays (7 Gy). For this purpose, the animals were sacrificed on the 7th and 28th day after irradiation. Their lung, heart, liver, kidney, small intestine and spleen were taken for histopathological and semiquantitative analysis. Careful examination of established tissue and vascular alteration revealed better radioprotective effects of FNP compared to those of AMI on the small intestine, lung and spleen, while AMI had better radioprotective effects than FNP in protection of the heart, liver and kidney. Results of this study confirmed high radioprotective efficacy of FNP in irradiated rats that was comparable to that of AMI, a well-known radioprotector.
... fatigue, nausea, vomiting, diarrhea, peripheral neuropathy, and cognitive function impairment) that reduce the efficacy of treatment [47]. It is known that radiation-induced damage alters mitochondrial metabolism, inhibits the mitochondrial respiratory chain, and forms highly reactive peroxynitrite (ONO 2 − ) [48]. Once mitochondrial proteins are damaged, the affinity of substrates or enzymes is decreased resulting in mitochondrial dysfunction [45]. ...
Article
Fatigue is one of the most common side-effects accompanying radiotherapy, but arguably the least understood. Radiotherapy-induced fatigue (RIF) is a clinical subtype of cancer treatment-related fatigue. It is described as a pervasive, subjective sense of tiredness persisting over time, interferes with activities of daily living, and is not relieved by adequate rest or sleep. RIF is one of the early side-effects and long-lasting for cancer patients treated with localized radiation. Although the underlying mechanisms of fatigue have been studied in several disease conditions, the etiology, mechanisms, and risk factors of RIF remain elusive, and this symptom remains poorly managed. The purpose of this paper is to review and discuss recent articles that defined, proposed biologic underpinnings and mechanisms to explain the pathobiology of RIF, as well as articles that proposed interventions to manage RIF. Understanding the mechanisms of RIF can describe promising pathways to identify at-risk individuals and identify potential therapeutic targets to alleviate and prevent RIF using a multimodal, multidisciplinary approach.
... Curcumin also has a radioprotection effect due to its capability to decrease oxidative stress and inflammatory responses, and also confers radiosensitization perhaps via the upregulation of genes responsible for apoptosis [58]. Multiple studies support the intrarectal application of amifostine (WR-2721/WR-1065), a phosphorothioate during external beam RT for prostate cancer for the prevention of radiation-induced rectal injury [59][60][61]. ...
Article
Full-text available
Toxicity induced by radiation therapy is a curse for cancer patients undergoing treatment. It is imperative to understand and define an ideal condition where the positive effects notably outweigh the negative. We used a microarray meta-analysis approach to measure global gene-expression before and after radiation exposure. Bioinformatic tools were used for pathways, network, gene ontology and toxicity related studies. We found 429 differentially expressed genes at fold change >2 and p-value <0.05. The most significantly upregulated genes were synuclein alpha (SNCA), carbonic anhydrase I (CA1), X-linked Kx blood group (XK), glycophorin A and B (GYPA and GYPB), and hemogen (HEMGN), while downregulated ones were membrane-spanning 4-domains, subfamily A member 1 (MS4A1), immunoglobulin heavy constant mu (IGHM), chemokine (C-C motif) receptor 7 (CCR7), BTB and CNC homology 1 transcription factor 2 (BACH2), and B-cell CLL/lymphoma 11B (BCL11B). Pathway analysis revealed calcium-induced T lymphocyte apoptosis and the role of nuclear factor of activated T-cells (NFAT) in regulation of the immune response as the most inhibited pathways, while apoptosis signaling was significantly activated. Most of the normal biofunctions were significantly decreased while cell death and survival process were activated. Gene ontology enrichment analysis revealed the immune system process as the most overrepresented group under the biological process category. Toxicity function analysis identified liver, kidney and heart to be the most affected organs during and after radiation therapy. The identified biomarkers and alterations in molecular pathways induced by radiation therapy should be further investigated to reduce the cytotoxicity and development of fatigue.
... Ultimately, associated increase in the gap junction intercellular communication and release of these reactive products by the responsive cells can propagate non-targeted radiation effects [42]. Therefore, applications of modulators of: (i) mitochondrial redox function; (ii) the ER-stress response; (iii) antioxidant adaptive responses; (iv) renin-angiotensin system; (v) peroxisome proliferaton; (vi) activity of NADPH oxidase 4; and (vii) ROS/RNS-induced programmed cell death, are under recent consideration for mitigation of radiation-related effects [8,20,44,51,[54][55][56]. Thirdly, the targeted and non-targeted epigenetic alterations, up-regulation of inflammatory mediators can lead to a long-term expression of pro-oxidant genes (such as inducible nitric oxide synthase, NOSII), which can further exacerbate the metabolic oxidative/electrophilic stress [51,57,58]. ...
... To address adverse effects of this pro-oxidant shift the concept of antioxidant countermeasure against IR has been widely elaborated. To date, numerous antioxidants were proposed as litigators of the acute radiobiological effects [20,40,41,55,56,59]. So far, there is only one of them approved by the FDA for the radioprotection: this is amifostine (WR2721), an antiradical phosphorothioate [41]. ...
Article
Full-text available
The biological effects of high-dose total body ionizing irradiation [(thereafter, irradiation (IR)] are attributed to primary oxidative breakage of biomolecule targets, mitotic, apoptotic and necrotic cell death in the dose-limiting tissues, clastogenic and epigenetic effects, and cascades of functional and reactive responses leading to radiation sickness defined as the acute radiation syndrome (ARS). The range of remaining and protracted injuries at any given radiation dose as well as the dynamics of post-IR alterations is tissue-specific. Therefore, functional integrity of the homeostatic tissue barriers may decline gradually within weeks in the post-IR period culminating with sepsis and failure of organs and systems. Multiple organ failure (MOF) leading to moribundity is a common sequela of the hemotapoietic form of ARS (hARS). Onset of MOF in hARS can be presented as " two-hit phenomenon " where the " first hit " is the underlying consequences of the IR-induced radiolysis in cells and biofluids, non-septic inflammation, metabolic up-regulation of pro-oxidative metabolic reactions, suppression of the radiosensitive hematopoietic and lymphoid tissues and the damage to gut mucosa and vascular endothelium. While the " second hit " derives from bacterial translocation and spread of the bacterial pathogens and inflammagens through the vascular system leading to septic inflammatory, metabolic responses and a cascade of redox pro-oxidative and adaptive reactions. This sequence of events can create a ground for development of prolonged metabolic, inflammatory, oxidative, nitrative, and carbonyl, electrophilic stress in crucial tissues and thus exacerbate the hARS outcomes. With this perspective, the redox mechanisms, which can mediate the IR-induced protracted oxidative post-translational OPEN ACCESS Antioxidants 2015, 4 135 modification of proteins, oxidation of lipids and carbohydrates and their countermeasures in hARS are subjects of the current review. Potential role of ubiquitous, radioresistant mesenchymal stromal cells in the protracted responses to IR and IR-related septicemia is also discussed.