Attenuation of Space Radiation-Induced Damage by an Oral Supplementation with a Mixture of Micronutrients in Astronauts

Kedar N Prasad, Jae Ho Kim

Published Date: 2021-09-20

Kedar N Prasad1* and Jae Ho Kim2

1Department of Radiology, University of Colorado Health Sciences Center, Engage Global Inc. 245 El Faisan Drive, San Rafael, CA 94903, USA

2Department of Radiation Oncology, Henry Ford Health System, Detroit, Mi, 48202, United States

*Corresponding Author:
Kedar N Prasad
Department of Radiology, University of Colorado Health Sciences Center, Engage Global Inc. 245 El Faisan Drive
San Rafael, CA 94903, USA
E-mail: knprasad@comcast. net

Received Date: August 30, 2021; Accepted Date: September 13, 2021; Published Date: September 20, 2021

Citation: Prasad NK, Kim HJ (2021) Attenuation of Space Radiation-Induced Damage by an Oral Supplementation with a Mixture of Micronutrients in Astronauts. J Med Phys and Appl Sci Vol.6 No.5: 11.

Visit for more related articles at Journal of Medical Physics and Applied Sciences

Abstract

Context: There are no effective radiation protection strategies to protect the astronauts against space radiation while traveling beyond the earth-orbit.

Objectives: The objectives are to characterize space radiation and its ability to produce reactive oxygen species and inflammation that can increase adverse health effects in astronauts. Because of limitation of using a single or a mixture of few antioxidants, it proposes a comprehensive mixture of micronutrients for prevention and mitigation of radiation damage in astronauts.

Methods: Critical analysis of the published studies on space radiation-induced adverse health effects on astronauts and their countermeasures obtained from the PubMed Central was performed.

Results: While traveling beyond the earth-orbit, astronauts are exposed to galactic cosmic rays (GCRs), solar particles events, and (c) electrons and protons trapped in the Van Allen Belts. Space radiation enhances oxidative stress and inflammation that are associated with increased health risks. Administration of a single or a mixture of few antioxidants may not be adequate for radiation protection astronauts. Therefore, a comprehensive mixture of micronutrients for prevention and mitigation of space radiation injury is proposed.

Conclusion: Astronauts travelling beyond the earth orbit are exposed to space radiation, which enhance oxidative stress and inflammation that can produce adverse health effects. A comprehensive micronutrient mixture may prevent and mitigate adverse health effects in astronauts by simultaneously reducing the above cellular deficits.

Keywords

Space radiation; Reactive oxygen species; Inflammations; Micronutrients; HZE particles

Introduction

Yuri Gagarin of the former USSR was the first astronaut (also called cosmonaut) to travel into outer space and successfully completed one orbit of Earth on 12th April, 1961. Since then, astronauts are spending more and more time on space missions. Between 1969-1972, astronauts landed on the lunar surface in 6 of 7 lunar landing missions. They took 8-12 days to reach moon and spent 21-74 hrs on the lunar surface, depending upon the lunar landing missions. In recent years, astronauts are spending up to 6 months or more at the International Space Station (ISS). To live in such a close environment under microgravity, weightlessness, and exposure to low levels of galactic cosmic radiation (GCR) poses increased health risks to astronauts. These risks include space radiation-induced neoplastic and non-neoplastic diseases, microgravity-induced musculoskeletal atrophy, and psychological stress due to living in isolation.

To The next challenging space exploration mission may include travel to the Moon followed by journey to the Mars. It will take 800 to 1,100 days to reach Mars and astronauts may spend 500 days on the surface of Mars, depending on the final mission design [1]. It is expected that astronauts travelling to Mars will be exposed to much higher doses of space radiation, microgravity, and weightlessness than those who spent a 6-month mission on the ISS. Therefore, the risks of adverse health effects in astronauts are expected to increase much greater than those travelling in the ISS. For the successful space missions, it is imperative to develop safe and effective countermeasures for astronauts on various space missions as well as for astronauts returning to civilian life. In order to accomplish this, it is essential to identify space radiation-induced generation of reactive chemicals that damage the cells.

GCR radiation by virtue of being high linear energy transfer (LET) radiation causes damage by producing dense ionization in biological molecules that cannot be protected by pharmacological or physiological agents. Because exposure to GCR radiation also generates extensive amounts of reactive oxygen species (ROS) along the track, the damage produced by this component of GCR can be protected by scavengers of ROS. One of such scavengers of ROS is antioxidant.

This review briefly describes (a) characteristics of space radiation, (b) production of ROS during GCR radiation, (c) effects of gravity on the production of ROS, (d) adverse health effects of space radiation, and (e) biological strategies for prevention and mitigation of space radiation-induced cellular damage. This review points out the limitations of administration of a single or a mixture of antioxidants only before irradiation for space radiation protection in astronauts. To address these limitations, the review proposes a comprehensive mixture of micronutrients that can be administered orally before and after exposure to irradiation for prevention and mitigation of radiation injury in astronauts during space travel, during stay on the lunar or Mars surface, and during returns to their civilian life.

Literature Review

Characterization of space radiation

The Space radiation is different from the terrestrial form of radiation with respect LET. Terrestrial radiation such as x-rays and gamma-rays are low LET, whereas space radiation such as high energy particles are high LET. Astronauts in the International Space Station (ISS) at the altitudes of 300-400 km and orbital inclination of 51.60 are exposed to three primary sources of space radiation, (a) galactic cosmic rays (GCRs) consist of high energy protons and heavy ions from outside of our solar system, (b) solar particles events (SPEs) during which high energy particles (electron, neutrons, protons, and heavy ion nuclei) are released, and (c) electrons and protons trapped in the Van Allen Belts outside the spacecraft. During SPE, high energy protons arising from the sun within the region of solar magnetic instability are released for a short period of time, while solar wind consists of low energy protons and electrons [2]. The combination of these forms of radiation produces a complex radiation environment inside and outside of the ISS, and the extent of complexity of radiation depends upon the solar cycle, altitudes, and shielding of each module of the ISS [3].

Galactic cosmic radiation (GCR) is made of the nuclei of atoms that have no electrons and travels nearly speed of light. GCR is composed of 85% hydrogen nuclei (protons), 14% helium nuclei, and 1% high-energy and highly charged high atomic number ions called HZE particles that include heavy ions of carbon, iron, or nickel nuclei. Although HZE particles represent very small part of GCR, they exhibit higher ionizing power, greater penetration ability, and inflicting greater biological damage. GCR can easily penetrate through a typical spacecraft as well as through astronaut skin. Humans are protected from GCR because of earth atmosphere and magnetic field, whereas lunar surface is devoid of these protective atmosphere and magnetic field [4]. Therefore, astronauts on the lunar surface are likely to receive more radiation than on the earth.

Generation of secondary radiations by HZE particles

Technological evolution Interaction between HZE particles and electrons in target atoms, and fragmentation of the incident HZE particles and target nuclei gives rise to secondary radiations, which include photons, protons, neutrons, alpha-particles, and other heavy ions with lower velocity as well as energetic electrons referred to as delta rays [5,6]. Delta rays due to their long range can irradiate not only direct target cells but also neighboring by stander cells. As a matter of fact, the total dose deposited by delta rays could be as high 30-40% of HZE particles [7,8].

Space radiation facilities available to perform experiments on the earth

The HZE particles of galactic cosmic radiation are difficult to obtain for experiments on the earth. However, the Brookhaven National Laboratory in NY has facility to provide high energy ions from proton to gold, the Gunma University Heavy Ion Medical Center in Japan, and the National Center for Oncological Hadron therapy in Italy have facilities to provide high energy C-ion beam. Heavy-Ion Medical Accelerator in Chiba in Japan can provide high energy beam of He, C, Cr, Fe, Ni, Xe, and the GSI Helmholtz Center for Heavy Ion Research in Germany can provide high energy beam of ions from proton to uranium for biological and shielding experiments.

Studies performed using HZE radiation facilities on the earth

Extensive studies on the adverse health effects of HZE particles utilizing high energy heavy ions have been performed primarily in rodents. However, there are several uncertainties regarding the extrapolation of the results from the above studies to astronauts traveling in lower-earth orbit (LEO) and beyond. They include use of mono-energetic high atomic number ions radiation, higher dose rate, and total dose [9]. The use of sequential irradiation with more than one high energy ions is better than one monoenergetic ion, but it cannot mimic the effects of complex space radiation. In contrast, astronauts are exposed to complex radiation environment of particles and ions, together with microgravity and social isolation. These conditions are difficult to reproduce on the earth for laboratory experiments. Nevertheless, experimental data on the adverse health effects of mono energetic HZE particle in rodents suggest that such effects may be found in astronauts who would stay inside the spacecraft for more than 6 months, who perform extravehicular activities, and who may reside on lunar surface for several days.

GCR radiation doses received by astronauts during extravehicular activities

Astronauts are exposed to higher doses of space radiation during extravehicular activities than during the same period inside the spacecraft despite extravehicular space suits, which provides insufficient shielding against such radiation [10].

Doses received by astronauts on the lunar surface

Between 1969-1072, astronauts successfully landed on lunar surface in 6 of 7 lunar landing missions. The average dose to astronauts at the lunar surface varied from 180 mSv to 1140 mSv (Life Sciences Data Archive at JSC3) (Table 1).

Mission Duration to the moon Lunar surface duration   Average dose to astronauts
Apollo 11              8 days, 3hrs, 13 mins        21 hrs, 36 mins 0.18 rad (180 mSv)
Apollo 12   10 days, 4 hrs, 31 mins       31 hrs, 31 mins 0.58 rad (580 mSv) 
       Apollo 14          9 days, 1 min             33hrs, 31 mins 1.14 rad (1140 mSv)
         Apollo 15     10 days, 1 hrs, 11 mins           66 hrs, 54 mins 0.30 rad (300 mSv)
Apollo 16      11 days, 1 hrs. 51 mins          71 hrs. 2 mins 0.51 rad (510 mSv)
Apollo 17     12 days, 13 hrs, 51 mins 74 hrs., 59 mins 0.55 rad (550 mSv)

Table 1: Describes time to reach to the moon, time spent on lunar surface, and average dose in rad as well as in mSv (in parenthesis).

Astronauts in deep space are exposed to both solar particle events (SPE) and galactic cosmic rays (GCR). Dose rates from SPE and GCR are around (0.5 mSv)/day inside the ISS. The average dose to 7 deceased Apollo crew was +/- 5.9 mSv 1.5 mSv (range 1.8 mSv to 11.4 mSv) [11].

The dose to astronauts depends upon the time spent in the spacecraft, extravehicular activities or on the lunar surfaces. Generally, effective dose is low; however, the biological effects are cumulative. High-energy protons and charged particles can damage shielding as well as biological system [3,12]. In recent years, astronauts are spending up to 6 months or more at the International Space Station (ISS). To live in such a close environment under microgravity and weightlessness, exposure to low levels of space radiation poses additional health risks to astronauts. Astronauts received an effective dose in the range of 50-2000 mSv over 6-12 months stay in ISS. Astronauts would receive approximately 290 mSv during an average duration stay of 15.6 days in ISS [13].

Reactive (ROS) production during exposure to galactic cosmic radiation

An excellent review has described the involvement of increased oxidative stress and inflammation in damage produced by HZE particles in in vitro and in vivo [14]. High LET HZE particles produce direct effect by ionizing cellular molecules and indirectly by producing reactive oxygen species (ROS) from hydrolysis of water, which generate various oxidizing species such as e-aq (hydrated electron), •OH, H•, H2, H2O2 [15, 16]. In the presence of oxygen, e-aq and H• species are rapidly converted to superoxide (O2 •-) and perhydroxyl (HO2 •). Organic radical (R•), when combines with O2, produce Peroxyradical (RO2 ) [17]. The burst of these ROS occur along the track of HZE particles [18]. Space radiation can stimulate the activity of inducible nitric oxide synthase in the target cells [19], leading to generation of large amounts of nitric oxide (•NO), which can combine with O•-2 to form peroxynitrite (ONNO-), which can damage nucleic acid, lipid, protein, and thiol [20]. Peroxynitrite and other reactive nitrogen species (RNS), when generated during inflammation, can damage nucleic acid, protein and lipid in neighboring bystander cells that can lead to genomic instability or cell death [21].

Unlike low-LET radiations (x-rays, γ-rays), which decrease energy deposition exponentially as a function of tissue penetration, the energy deposit of HZE particles is characterized by a relatively low entrance dose in the target tissue and a sharp maximum deposit at the end of range known as a Bragg peak. Because of the spread of oxidative stress from cells directly targeted with HZE to neighboring non-targeted cells, the increased oxidative stress continues to persist in both targeted and non-targeted cells. This in turn promotes genetic instability that may enhance the risks of long-term late effects including cancer and degenerative disease [14].

ROS production during exposure to microgravity

The interaction between the effects of HZE particles and microgravity in producing ROS is not fully defined. Exposure to microgravity alone contributes to oxidative stress leading to bone loss [22]. The analysis of the urine of astronauts on the Russian space station MIR and on the shuttle for a short- duration missions revealed opposite effect on the levels of oxidative stress during in-flight and post-flight [23]. The levels of oxidative damage were less during in-flight, whereas they increased during post-flight. These results were interpreted to mean that decreased production of ROS was due to the reduced energy production by the astronauts during in-flight, while increased oxidative stress during post-flight was due to decreased antioxidant defense system. Indeed, reduced activities of antioxidant enzymes were detected for several days after space flight [24].

Microgravity increased lipid peroxidation in human erythrocyte membrane and decreased blood antioxidant levels in cosmonauts [25,26]. Long-term missions increased triglyceride levels and ratio of HDL/LDL cholesterol (high- density lipoprotein/low-density lipoprotein) [27]. Since space radiation generates extensive amounts ROS and RNS (reactive nitrogen species), it is likely that the combination of space radiation and microgravity together with other space stressors may increase the risk of cardiovascular disease, including atherosclerosis in astronauts.

ROS production during exposure to hypergravity

It is essential to note that astronauts are also exposed to hypergravity during the launch of the spacecraft and upon re- entry to the earth environment, but no significant studies on changes in the levels of oxidative stress have been performed. In rats, increased levels of MDA (malondialdehyde), a marker of oxidative stress, were found in the heart and kidney tissues [28,29]. Exposure to hypergravity increased oxidative stress and decreased the number of Purkinje neurons and impaired motor co-ordination in rat neonates [30].

Induction of inflammation as a response to cellular radiation damage

Ionizing radiation is known to induce chronic inflammation via radiation-induced cell damage [31]. GCR disrupted synaptic integrity and increased neuroinflammation, which persisted 6 months after irradiation in rats [32]. As soon as cell injury irrespective of inducing agents occurs, acute inflammation releases anti-inflammation cytokines at the site of injury to help in the healing process. When the cell damage is repaired, acute inflammation is turned off. In the event, the cell injury is not healed, chronic inflammation, which releases pro-inflammatory cytokines, complement protein, adhesion molecules, and ROS occurs. Space radiation-induced cellular damage caused by ROS can be attenuated by antioxidants; however, injury caused by ionization of vital cell molecules cannot be reduced by antioxidants, therefore, cellular damage persists, which initiates chronic inflammation. Both increased oxidative stress and chronic inflammation play an important role in initiating and progression of space radiation-induced adverse health effects. Thus, decreasing oxidative stress and chronic inflammation at the same time may decrease the acute and late health risks in astronauts. In order to achieve this goal, increasing the levels of antioxidant enzymes as well as dietary and endogenous antioxidant compounds at the same time for neurodegenerative diseases such as Alzheimer’s disease have been proposed [33]. The same idea would be applicable to astronaut protection.

Adverse health effects of space radiation

Possible health risks to astronauts include cancer, damage to the central nervous system, cardiovascular disease, cataract, acute radiation sickness, and genetic damage [3,12,34].

Effects of space radiation on the brain function: It is essential that astronauts maintain normal neuronal function during long space journey to Moon or Mars for the success of missions. Effects of HZE particles on the brain function have been investigated primarily on rodents [35]. Exposure to HZE particles increased the presence of dense fibrillary proteins and beta-amyloid fragments in the serum of mice that are associated with Alzheimer’s disease [36]. Irradiation of rats with HZE particles induced cognitive dysfunction and behavioral deficits that are linked to the damage of structure and synaptic integrity of the specific region of the brain [37-40]. These adverse neuronal functions persisted over one year, suggesting that HZE particles can cause permanent damage in the brain [32]. Most studies have used mono-energetic HZE particle radiation. One study has investigated the effects of sequential irradiation with protons, 16 O ions, and 28 Si ions on behavioral and cognitive performance in male and female mice [41]. Results showed that mice of both sexes exhibited depressed behavior and impaired cognitive function. In addition, cortical levels of microglia activation marker CD68 (Cluster of Differentiation 68) increased after irradiation in females but not in males indicating the presence of neuroinflammation, while the levels of BDNF (brain-derived neurotrophic factor) decreased after irradiation in males, but not in females. Gut microbiome also changed after irradiation in these mice.

Space radiation on cancer risk: Space radiation can increase the risk of cancer in astronauts during civilian life, especially after long duration flight because of exposure to complex radiation environment of low and high LET radiation together with microgravity inside and outside of the spacecraft. There are no human data available on this issue. However, animal studies suggest that the risk of cancer exists in astronauts exposed to space radiation. Exposure to HZE particles induced malignant lymphoma and harderian gland tumor in the eye’s orbit of mice [34, 42]. Also, exposure to proton radiation and Fe-ions radiation induced premalignant changes and malignant tumors of myeloid origin of the bone marrow [42].

Effects of space radiation on DNA damage and chromosomal damage: Galactic cosmic radiation causes dense ionization in DNA along the radiation tracks and induces “complex DNA damage” or clustered DNA damage. This form of DNA damage is difficult to repair when compared to normal DNA damage [43- 45]. Gamma-H2AX, a phosphorylated histone protein, is a marker of DNA double-strand breaks (DSBs). Tracks of the gamma-H2AX foci are found in the nuclei of lymphoblast’s [46] and fibroblasts in astronauts after space flight [47].This form of DNA damage can lead to cancer or cell death. The frequency of chromosomal anomalies appeared to be higher at post-flight than at pre-flight among astronauts, especially after space flight of longer than 180 days [48,49]. Thus, space radiation can cause genetic instability that can increase the risk of cancer. Simultaneous exposure to microgravity and x-rays or carbon ions increased chromosomal damage more than that observed with microgravity, x-rays or carbon ions alone.

Effects of space radiation on the eye: Astronauts have increased risk of cataract due to exposure to GCR during space travel [50-52]. High LET radiation is known to have a high radiobiological effect (RBE) on the development of cataract.

Effects of space radiation on the immune system: Because of radiation-induced impaired immune system, astronauts have increased risk of serious infections including debilitating dental infection and urinary tract infection [53-58]. Reduction in wholeblood counts following exposure to SPE particles was greater in pigs than in ferrets and mice [59]. Combination of microgravity and SPE radiation can produce additive or synergistic effects on the immune system of murine models [60-63].

Studies on protection of astronauts against galactic cosmic rays

Antioxidants administered before irradiation with HZE particles: Dr. Kennedy and her colleagues at the University of Pennsylvania have performed pioneering studies on radiation protection using high doses of mono-energetic HZE particles from one of the facilities on the earth [34,64]. These studies have utilized one antioxidant or a mixture containing antioxidants and some B-vitamins. There may be several limitations of using one antioxidant in astronauts, although it is effective in protecting rodents against HZE particles radiation. This issue will be presented later in the manuscript. The mixture used in the above studies contains most dietary and endogenous antioxidants, only 2 B-vitamins, selenomethionine and Bowman-Birk inhibitor. This mixture produced significant protection against monoenergetic HZE particles radiation in rodents and cell culture models. For the studies on astronauts, adding other antioxidants, vitamin D3, all B-vitamins may be essential for protection against acute and long-term damage produced by space radiation (see the rationale below).

Extensive studies on micronutrients, phytonutrients, and selenium when administered before monoenergetic HZE particles irradiation in animal and cell culture models have been performed. A few such studies are described here.

Treatment of human cells in culture with selenome-thionine for 18 hrs. Before irradiation prevented monoenergetic HZE particles-induced rise in increased oxidative stress as well as it reduced lethality of irradiated cells [65]. Administration of diet containing vitamin A one week before accelerated 56Fe ion-irradiation significantly reduced the expression of inflammation-related genes and reduced the incidence of cancer by half in rats [66]. Irradiation of hippocampal cells in culture with 1 Gy of high energy 56Fe ions produced extensive amounts of free radicals [67]. Treatment of these cells with alpha-lipoic acid before or after irradiation markedly attenuated the levels of ROS. Treatment of middle-aged mice with alpha-lipoic acid 3 weeks before irradiation with 3 Gy of 56Fe ions reduced radiation-induced impairment of special memory retention ability [68]. Supplementation with diet rich in polyphenolic compounds from blueberry or strawberry extract 8 weeks before irradiation with 1.5 Gy or 2 Gy of energetic 56Fe ions protected against radiation-induced behavior impairment [69].

Dietary supplementation with a mixture of antioxidant containing n-acetylcysteine (NAC), coenzyme Q10, glutathione, alpha-lipoic acid, vitamin C, vitamin E succinate, B-vitamins (B1-thiamine, B3-niacin), BBI (Bowman-Birk inhibitor), and selenomethione in various combinations before irradiation provided protection against monoenergetic H ZE particles radiation in mic e and cell culture models [34,42]. One CT scan, which delivers 10 mGy of gamma-radiation, i nduced double-strand D NA breaks (DSBs) in the peripheral lymphocytes of patients. Administration of a mixture of multiple antioxidants (a commercial preparation) 60 minutes before the CT scan reduced the number of cells with DSBs by over 50% [70].

Antioxidants administered after irradiation with gammaradiation: Family of vitamin E, administered before irradiation is more effective than alpha-tocopherol in providing radiation protection in animal models [71]. This micronutrients also effective when administered after irradiation [72]. For an optimal radiation protection, timing of administration after irradiation is very important. Dietary supplementation with a mixture of antioxidants (selenomethionine, sodium ascorbate, N-acetylcysteine, alpha-lipoic acid, vitamin E succinate, and coenzyme Q10) when administered 24 hrs after total body irradiation with gamma-radiation provided better radiation protection than that administered immediately after irradiation [73]. Similar observations were reported in another study [74]. Diet supplemented with dried plum powder reduced space radiation and microgravity-induced bone loss by decreasing oxidative stress and inflammation in mice [75].

Limitations of administering antioxidants only before or after irradiation

In most studies, antioxidants were administered before or after irradiation. Existence of long-lived free radicals after irradiation [76] suggested that post-irradiation treatment with antioxidants together with pre-irradiation treatment would be useful in optimally reducing radiation damage. However, timing of postirradiation treatment is important. Post-irradiation treatment with antioxidants soon after irradiation may interfere the action of anti-inflammatory cytokines that are released to help in the repair of cellular damage in the early of phase of tissue repair. Studies on rodents have shown that administration of antioxidant mixture 24 hours after irradiation provide better protection than that administered soon after irradiation [73,74]. No studies have been performed to evaluate the effectiveness of antioxidants when administered orally before and after irradiation in the same animal or cell culture models.

Limitations of administering a single antioxidant for radiation protection in astronauts

The administration of a single antioxidant is effective in prevention or mitigation of radiation injury in animal models; it may not provide radiation protection in the astronauts because of several limitations. Some of them are described here.

(a) Space radiation produces high levels of ROS and RNS. Administration of a single antioxidant in a high internal oxidative environment of astronauts would be oxidized, which then would act as a pro-oxidant rather than as an antioxidant [75].

(b) Different antioxidants are distributed differently in the subcellular compartments of cells, all of which must be protected. Administration of a single antioxidant cannot accumulate in all parts of the cell in sufficient amounts to provide adequate protection in astronauts [76].

(c) Vitamin E is more effective scavenger of free radicals in reduced oxygen pressure, whereas beta-carotene and vitamin A are more effective in higher oxygen pressure of the cells [77]. Therefore, administration of one antioxidant may not provide adequate protection in whole body.

(d) Elevation of both antioxidant enzymes and dietary and endogenous antioxidant compounds are needed to achieve maximal protection against radiation-induced oxidative and inflammatory damage. This is due to the fact that they act by different mechanisms. Antioxidant compounds neutralize free radicals by donating electrons to those molecules with unpaired electrons, whereas antioxidant enzymes destroy H2O2 by catalysis, converting them to harmless molecules such as water and oxygen. Administration of a single antioxidant cannot achieve this goal.

(e) Administration of a single antioxidant cannot protect both the aqueous and lipid compartments of the cell against radiation injury.

(f) Different antioxidants increase the production of different protective proteins in the cells by altering the expression of different microRNAs [78]. For example, some antioxidants can activate Nrf2 by upregulating miR-200a that inhibits its target protein Keap1, whereas others activate Nrf2 by down-regulating miR-21 that binds with 3’-UTR Nrf2 mRNA [79]. Thus, different antioxidants activate Nrf2 (Nuclear Factor-Erythroid-2-Related Factor 2) by different mechanisms. Administration of a single antioxidant cannot accomplish the above objective.

Administration of a single antioxidant compound failed to yield expected benefits that were observed in animal models. For examples, administration of vitamin E alone produced no effect in patients with Parkinson’s disease [80,81]. It also had no effect on cognitive function in patients with Alzheimer’s disease, but it produced minimal benefits in early phase of this disease [82,83]. Administration of beta-carotene alone in male heavy tobacco smokers increased the risk of lung cancer [84]. These studies suggest that administration of a single antioxidant is unlikely to provide any significant protection against space radiation in astronauts.

How to simultaneously reduce oxidative stress and inflammation

In order to reduce oxidative stress and inflammation at the same time, it is essential to simultaneously enhance the levels of antioxidant enzymes and dietary and endogenous antioxidant compounds [85]. Oral supplementation with a mixture of antioxidants can increase the levels of dietary and endogenous antioxidant compounds; however, enhancing the levels of antioxidant enzymes requires an activation of a nuclear transcriptional factor Nrf2. A brief description of activation processes is presented here.

Processes of Activation of Nrf2: Under normal physiological conditions, activation of Nrf2, one of the nuclear transcriptional factors, requires ROS. Activated Nrf2 dissociates itself from Keap1-CuI-Rbx1 complex and migrates to the nucleus where it heterodimerizes with a small Maf protein and binds with ARE (antioxidant response element) leading to increased transcription of cytoprotective enzymes including antioxidant enzymes [86, 87]. It appears that during prolonged oxidative stress commonly observed in human chronic diseases, activation of Nrf2 becomes resistant to ROS [88,90]. This is evidenced by the fact that increased oxidative stress continues to occur in chronic diseases despite the presence of Nrf2. However, some antioxidants such as vitamin E [91], alpha-lipoic acid [92], curcumin [93], resveratrol [94,95], omega-3-fatty acids, [96,97], glutathione [98], n-acetylcysteine [99], and coenzyme Q10 [100] activate ROS-resistant Nrf2.

An activation of Nrf2 alone is not sufficient to increase the levels of antioxidant enzymes. Activated Nrf2 must bind to ARE to increase the transcription of cytoprotective enzymes including antioxidant enzymes. It is interesting to note that the binding ability of Nrf2 to ARE was impaired in aged rats and this defect was restored by supplementation with alpha-lipoic acid [92]. It is unknown whether the binding ability of activated Nrf2 would be impaired in astronauts during prolonged stay inside the spacecraft or long after space journey when they adopt civilian life.

It has been reported that activation of Nrf2 decreases oxidative stress as well inflammation [101,102]. Many antioxidant compounds also attenuate inflammation [103-108]. Levels of oxidative stress and inflammation are closely linked. Inflammatory responses are necessary for healing of oxidative damage of the cells, and it turned off after healing is complete. However, oxidative damage of cells is not healed, chronic inflammation responses occur. Such inflammatory responses release ROS, proinflammatory cytokines, adhesion molecules and complement proteins, all of which are toxic to the cells.

Proposed mixture of micronutrients for reducing adverse health-effects of space radiation

A comprehensive mixture of micronutrients containing vitamin A, mixed carotenoids, vitamin C, alpha-tocopheryl acetate, alpha-tocopheryl succinate, vitamin D3, alpha-lipoic acid, N-acetylcysteine, coenzyme Q10, omega-3 fatty acids, curcumin, resveratrol, all B-vitamins, and minerals selenomithionine, and zinc for prevention and mitigation of space radiation is proposed [33,85]. This mixture would increase the levels of antioxidant enzymes by activating the ROS-resistant Nrf2 and enhancing the levels of dietary and endogenous antioxidant compounds. Such a micronutrient mixture may optimally reduce oxidative stress and chronic inflammation at the same time, and thereby, provided maximal protection against space radiation in astronauts. The question arises whether any mixture of antioxidants has produced beneficial effects in human diseases. Two clinical studies support the value of a mixture of micronutrient in producing beneficial effects in certain human diseases in which oxidative stress and inflammation plays a major role in their pathogenesis. For example, administration of multiple micronutrients reduced the risk of cancer in men [109] and delayed the progression of HIV disease by prolonging the time period for initiating the anti-viral therapy [110]. Therefore, it is likely that the proposed micronutrient mixture may reduce the adverse effects of space radiation by reducing oxidative stress and inflammation at the same time in astronauts during as well as after space journey. Pre-clinical studies administering this micronutrient mixture before and after exposure to HZE particles radiation should be performed to test its effectiveness in radiation protection.

Discussion and Conclusion

The approach of methodology is theoretical. The data was collected from the secondary sources like published books, articles etc. Critical analysis of the published studies on space radiation-induced adverse health effects on astronauts and their countermeasures obtained from the PubMed Central was performed.

Astronauts are exposed to space radiation during travel into lowearth orbit and beyond. Three primary sources of space radiation include (a) galactic cosmic rays (GCR) consists of high energy protons, Helium nuclei, and high-energy and highly charged ions called HZE particles that include heavy ions of carbon, iron, or nickel nuclei released from outside of our solar system, (b) solar particles events (SPEs) during which high energy particles (electron, neutrons, protons, and heavy nuclei) are released, and (c) electrons and protons trapped in the Van Allen Belts outside the spacecraft. The combination of these forms of radiation produces a complex radiation environment inside and outside of the International Space Station (ISS). Despite extravehicular space suits, astronauts are exposed to higher doses of space radiation during extravehicular activities than during the same period inside the spacecraft. The risk of cancer, brain damage, cardiovascular disease, cataract, and genetic damage after exposure to space radiation exist in astronauts. Space radiation produces damage by ionizing vital molecules in the cells and by generating extensive amounts of ROS along radiation track. Although administration of a single or a mixture of few antioxidants prevents and mitigates ROS-induced radiation damage in animal and cell culture models, they may not provide an optimal protection in astronauts. The reasons for this discrepancy have been discussed. Therefore, a comprehensive mixture of micronutrients containing dietary and endogenous antioxidants, resveratrol, curcumin, n-acetylcysteine, and minerals selenomethione and Zn is proposed for prevention and mitigation of space radiation injury in the astronauts during space travel as well as during civilian life. The proposed micronutrient mixture can be prepared in the form of paste and placed in a tube for the astronauts while travelling in space or spending time on the lunar surface. Astronauts can resume micronutrient mixture in capsules when they return to civilian life.

Conflict of Interest

The author is Chief Scientific Officer of Engage Global of Utah. This company sells nutritional products to consumers.

References

open access journals, open access scientific research publisher, open access publisher
Select your language of interest to view the total content in your interested language

Viewing options

Flyer image

Share This Article

paper.io

agar io

wowcappadocia.com
cappadocia-hotels.com
caruscappadocia.com
brothersballoon.com
balloon-rides.net

wormax io