Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 The impact of biofilms on long-duration space travel should be studied. + 1614 word(s) 1614 2020-07-15 05:47:24 |
2 Format -175 word(s) 1439 2020-07-15 15:13:57 | |
3 format correct -66 word(s) 1373 2020-10-27 09:07:40 | |
4 format correct Meta information modification 1373 2020-11-03 03:36:13 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Landry, K.S.; Morey, J.M.; Bharat, B.; Haney, N.M.; Panesar, S.S. Biofilms in Space. Encyclopedia. Available online: https://encyclopedia.pub/entry/1348 (accessed on 19 April 2024).
Landry KS, Morey JM, Bharat B, Haney NM, Panesar SS. Biofilms in Space. Encyclopedia. Available at: https://encyclopedia.pub/entry/1348. Accessed April 19, 2024.
Landry, Kyle S, Jose M Morey, Bharat Bharat, Nora M Haney, Sandip S Panesar. "Biofilms in Space" Encyclopedia, https://encyclopedia.pub/entry/1348 (accessed April 19, 2024).
Landry, K.S., Morey, J.M., Bharat, B., Haney, N.M., & Panesar, S.S. (2020, July 15). Biofilms in Space. In Encyclopedia. https://encyclopedia.pub/entry/1348
Landry, Kyle S, et al. "Biofilms in Space." Encyclopedia. Web. 15 July, 2020.
Biofilms in Space
Edit

The impacts of biofilms are well known in the medical, agricultural, commercial, and industrial spaces. It less known that biofilms are undermining many facets of space travel and that their effects need to be understood and addressed for future space missions. Biofilms can damage space crew health and spoil limited food supply. Yet, at the same time, they can benefit plant systems for food growth, nutrient development, and other biological systems that are being explored for use in space travel. Various biofilm removal techniques have been studied to mitigate the hazards posed by biofilm persistence during space travel. Because the presence of biofilms can advance or hinder humanity’s space exploration efforts, an understanding of their impacts over the duration of space flights is of paramount importance. 

biofilms space travel bacteria health microbiome pathogens

1. Introduction 

It is well known that space travel subjects crew members to elevated levels of radiation that are known to increase their risk of mutations and cancer [1][2][3]. While NASA can try to protect their astronauts with shielding materials on spacecraft and spacesuits, bacteria have found a way to successfully adapt to these conditions. It has been demonstrated that bacteria can genetically and physically modify their tolerances to lower earth orbit (LEO) conditions, and one of the main mechanisms for this was the formation of biofilms [4][5][6]. While subjected to microgravity, the bacterial populations within biofilms have evolved modifications to genes and gene expression that allows them to survive in hostile environments while also increasing their virulence and pathogenicity factors [7][8][9][10]. Since astronauts will be exposed to bacterial biofilms during long-term space travel, it is imperative that the space exploration community develop an understanding of biofilm formation, persistence, and the potential mitigation of their hazards.

2. Bacterial Biofilm Adaptation to the Extremes of Outer Space

The physical properties and characteristics of a biofilm are responsible for their protective and persistent nature. A bacterial biofilm is generally comprised of three components: 1) extracellular polymetric substances (EPS), 2) vegetative cells, and 3) bacterial remnants [11]. The EPS of a biofilm matrix is a complex mixture of organic material that acts as a structural glue and a physical barrier to disinfectants and antibiotics [12][13]. The EPS substance is a mixture of carbohydrates, proteins, lipids, and extracellular DNA (eDNA) [12][13]. The EPS matrix and its role in the pathogenicity and infection mechanisms is understood; however, the risks and impacts associated with biofilms and space travel is in its infancy.

Over the past decade, there has been an increased awareness of biofilms in space-related environments [14][15][16]. The formation of biofilms on surfaces and the bio-corrosion of space hardware and life-support systems are a significant concern to all space agencies, while also becoming a growing health concern on Earth [14]. Most materials found in space craft are incapable of resisting biofilm formation and require continual maintenance to prevent formation. Additionally, critical systems, such as water pipes, air ducts and life support require service to minimize harmful effects from biofilms [16]. This is especially true for the International Space Station (ISS) or any other craft designed to support human habitation for prolonged periods of time.

The main limitation to studying space-related biofilm formation is the ability to simulate space conditions. Even with this major hurdle, researchers have been able to utilize crew time on the ISS and have developed equipment that simulates microgravity to advance the study of biofilms in space [17][18]. One such study, presented by Kim et al., demonstrated that Pseudomonas aeruginosa formed a denser biofilm when grown under microgravity conditions than a biofilm grown on Earth [19][20]. The researchers were also able to demonstrate that the nutrient and gas diffusion rates within a biofilm grown under microgravity significantly impacted the overall cell density of a biofilm [20].

The microgravity conditions associated with LEO has also been shown to increase virulence factors in both Salmonella spp. and Escherichia coli [21][22]. Virulence and pathogenicity factors are tied to a variety of physical, metabolic, and functional gene expression of pathogens [23]. For example, flagella, a feature used for movement, is a key morphological feature that is known to be affected during growth under LEO space conditions [19,20]. Along with movement, the flagella has been shown to stimulate innate immunity, needed for the formation of microcolonies, allow cellular invasion, and promote bacterial surface adhesion [24][25]. For the transcriptome, microgravity has been shown to alter the expression of genes associated with biofilm formation, toxin production and resistance, and sporulation [26]. This also raises the question: what would happen if these enhanced pathogens were transported back to Earth following a deep space mission?

The impact of LEO conditions on the phenotypical characteristics of microorganisms has and continues to be studied to further understand what impact space travel will have on the microbial population about spacecraft. Aboard the Shenzhou VIII spacecraft, a strain of Klebsiella pneumonia was found to have conferred enhanced antibiotic resistance during the mission [27][28]. Interestingly, mutations continued to occur after returning to Earth [27][28]. The authors demonstrated that the mutations improved at least nine virulence/pathogenicity functions of the strain—including, but not limited to, oxidation-reduction capability and biofilm formation. Schiwon and colleagues demonstrated that over 75% of the Staphylococcus and Enterococcus species studied on the ISS demonstrated antibiotic resistance [29]. The research group postulated that most of the pathogens were normal human microflora, likely originating from the crew and cargo and that the LEO microgravity environment and the constant low-dose radiation exposure promoted the mutations. The increased pathogenicity and virulence factors of the human microbiome illustrates a serious challenge for long-duration space travel. There is no pre-flight “sterilization” process for crew members and their cargo that would limit microbial contamination and mutation during space flight [30]. To further emphasize the point, it should be noted that the standard 3-week quarantine procedures for crew members were ineffective at removing and/or limiting exposure to microorganisms that were exposed to LEO conditions [31][32].

The phenomenon known as anhydrobiosis has also been associated with bacterial biofilms in space. Contrary to the belief that water is needed to sustain bacterial life, many studies have shown that upon drying, certain bacteria are able to exist in a suspended state with little metabolic activity. Upon rehydration, the organisms are reactivated [33][34]. An experiment by Billi et al. demonstrated that dried biofilms of Chroococcidiopsis, when compared to their multi-layer planktonic counterparts, were able to recover faster after exposure to Mars-like conditions [35]. It was speculated that the drying process protected the organisms by minimizing the impact of free radicals and other reactive species that are present in Martian environments. Similar to bacteria suspended in anhydrobiosis, bacterial spores, when comprised in a biofilm, have also been shown to survive exposure to outer space conditions. Horneck et al. demonstrated that over the span of 6 years in outer space, Bacillus subtilis spores survived on the bottom layer of a biofilm [36]. A protective mechanism similar to anhydrobiosis is believed to protect bacterial spores from free radicals and cosmic galactic radiation.

3. Potential Benefits

Not all biofilms have negative impacts for space travel. In fact, biofilms may provide us with clues on how we as humans can tolerate space flight. For example, Rettberg et al. used a biofilm “dosimeter” to determine if adequate UV radiation was being experienced by astronauts on their space missions to produce adequate vitamin D [42][43]. The results from the biofilm-based studies indicated that the amount of vitamin D synthesis was inadequate and oral supplementation or sunlamp UV exposure on long-duration missions was recommended. These recommendations are now routinely used during space flight.

It has been proposed that biofilms formed via bacterization could be used to promote competitive ecologies within space systems [38]. Intentional bacterial seeding has also been proposed for environmental remediation and human health on Earth [44][45][46][47][48]. This idea has been proposed and studied, though it is in its infancy, for space-based applications. For example, Ichikawa et al. describes a biofilm reactor experiment used on spaceflight missions, which uses bacteria to clean up the nitrogenous byproducts produced by aquatic organisms [49]. Other applications may involve the seeding of beneficial bacteria in waste reactors, on various food production systems, and even seeding the astronaut’s intestinal tract prior, during, and after space flight. While these ideas are noteworthy, the interplay between space and bacterial colonization needs further exploration. This is especially true since the long-term effect of radiation on beneficial bacteria has not been studied.

References

  1. Durante, M.; Cucinotta, F.A. Physical basis of radiation protection in space travel. Rev. Mod. Phys. 2011, 83, 1245. [Google Scholar] [CrossRef]
  2. Seed, T.; Kumar, S.; Whitnall, M.; Srinivasan, V.; Singh, V.; Elliott, T.; Landauer, M.; Miller, A.; Chang, C.-M.; Inal, C. New strategies for the prevention of radiation injury: Possible implications for countering radiation hazards of long-term space travel. J. Radiat. Res. 2002, 43, S239–S244. [Google Scholar] [CrossRef] [PubMed]
  3. Peng, Y.; Nagasawa, H.; Warner, C.; Bedford, J.S. Genetic susceptibility: Radiation effects relevant to space travel. Health Phys. 2012, 103, 607–620. [Google Scholar] [CrossRef] [PubMed]
  4. Flemming, H.C.; Wingender, J.; Szewzyk, U.; Steinberg, P.; Rice, S.A.; Kjelleberg, S. Biofilms: An emergent form of bacterial life. Nat. Rev. Microbiol. 2016, 14, 563–575. [Google Scholar] [CrossRef] [PubMed]
  5. Panitz, C.; Frosler, J.; Wingender, J.; Flemming, H.C.; Rettberg, P. Tolerances of deinococcus geothermalis biofilms and planktonic cells exposed to space and simulated martian conditions in low earth orbit for almost two years. Astrobiology 2019, 19, 979–994. [Google Scholar] [CrossRef] [PubMed]
  6. Tirumalai, M.R.; Karouia, F.; Tran, Q.; Stepanov, V.G.; Bruce, R.J.; Ott, C.M.; Pierson, D.L.; Fox, G.E. The adaptation of Escherichia coli cells grown in simulated microgravity for an extended period is both phenotypic and genomic. NPJ Microgravity 2017, 3, 15. [Google Scholar] [CrossRef]
  7. Wilson, J.W.; Ott, C.M.; Honer zu Bentrup, K.; Ramamurthy, R.; Quick, L.; Porwollik, S.; Cheng, P.; McClelland, M.; Tsaprailis, G.; Radabaugh, T.; et al. Space flight alters bacterial gene expression and virulence and reveals a role for global regulator Hfq. Proc. Natl. Acad. Sci. USA 2007, 104, 16299–16304. [Google Scholar] [CrossRef]
  8. Taylor, P.W.; Sommer, A.P. Towards rational treatment of bacterial infections during extended space travel. Int. J. Antimicrob. Agents 2005, 26, 183–187. [Google Scholar] [CrossRef]
  9. Mermel, L.A. Infection prevention and control during prolonged human space travel. Clin. Infect. Dis. 2013, 56, 123–130. [Google Scholar] [CrossRef]
  10. Rosenzweig, J.A.; Abogunde, O.; Thomas, K.; Lawal, A.; Nguyen, Y.-U.; Sodipe, A.; Jejelowo, O. Spaceflight and modeled microgravity effects on microbial growth and virulence. Appl. Microbiol. Biotechnol. 2010, 85, 885–891. [Google Scholar] [CrossRef]
  11. Hall-Stoodley, L.; Costerton, J.W.; Stoodley, P. Bacterial biofilms: From the natural environment to infectious diseases. Nat. Rev. Microbiol. 2004, 2, 95–108. [Google Scholar] [CrossRef] [PubMed]
  12. Das, T.; Sharma, P.K.; Busscher, H.J.; van der Mei, H.C.; Krom, B.P. Role of extracellular DNA in initial bacterial adhesion and surface aggregation. Appl. Environ. Microbiol. 2010, 76, 3405–3408. [Google Scholar] [CrossRef] [PubMed]
  13. Fulaz, S.; Vitale, S.; Quinn, L.; Casey, E. Nanoparticle-Biofilm interactions: The role of the eps matrix. Trends Microbiol. 2019, 27, 915–926. [Google Scholar] [CrossRef] [PubMed]
  14. Gu, J.D.; Roman, M.; Esselman, T.; Mitchell, R. The role of microbial biofilms in deterioration of space station candidate materials. Int. Biodeterior. Biodegrad. 1998, 41, 25–33. [Google Scholar] [CrossRef]
  15. Castro, V.A.; Thrasher, A.N.; Healy, M.; Ott, C.M.; Pierson, D.L. Microbial characterization during the early habitation of the International Space Station. Microb. Ecol. 2004, 47, 119–126. [Google Scholar] [CrossRef]
  16. Koenig, D.W.; Pierson, D.L. Microbiology of the space shuttle water system. Water Sci. Technol. 1997, 35, 59–64. [Google Scholar] [CrossRef]
  17. McLean, R.J.; Cassanto, J.M.; Barnes, M.B.; Koo, J.H. Bacterial biofilm formation under microgravity conditions. FEMS Microbiol. Lett. 2001, 195, 115–119. [Google Scholar] [CrossRef]
  18. Frosler, J.; Panitz, C.; Wingender, J.; Flemming, H.C.; Rettberg, P. Survival of deinococcus geothermalis in biofilms under desiccation and simulated space and martian conditions. Astrobiology 2017, 17, 431–447. [Google Scholar] [CrossRef]
  19. Kim, W.; Tengra, F.K.; Young, Z.; Shong, J.; Marchand, N.; Chan, H.K.; Pangule, R.C.; Parra, M.; Dordick, J.S.; Plawsky, J.L.; et al. Spaceflight promotes biofilm formation by Pseudomonas aeruginosa. PLoS ONE 2013, 8, e62437. [Google Scholar] [CrossRef]
  20. Kim, W.; Tengra, F.K.; Shong, J.; Marchand, N.; Chan, H.K.; Young, Z.; Pangule, R.C.; Parra, M.; Dordick, J.S.; Plawsky, J.L. Effect of spaceflight on pseudomonas aeruginosa final cell density is modulated by nutrient and oxygen availability. BMC Microbiol. 2013, 13, 241. [Google Scholar] [CrossRef]
  21. Chopra, V.; Fadl, A.; Sha, J.; Chopra, S.; Galindo, C.; Chopra, A. Alterations in the virulence potential of enteric pathogens and bacterial–host cell interactions under simulated microgravity conditions. J. Toxicol. Environ. Health Part A 2006, 69, 1345–1370. [Google Scholar] [CrossRef] [PubMed]
  22. Rahme, L.G.; Stevens, E.J.; Wolfort, S.F.; Shao, J.; Tompkins, R.G.; Ausubel, F.M. Common virulence factors for bacterial pathogenicity in plants and animals. Science 1995, 268, 1899–1902. [Google Scholar] [CrossRef] [PubMed]
  23. Haiko, J.; Westerlund-Wikström, B. The role of the bacterial flagellum in adhesion and virulence. Biology 2013, 2, 1242–1267. [Google Scholar] [CrossRef] [PubMed]
  24. DRAKE, D.; MONTIE, T.C. Flagella, motility and invasive virulence of pseudomonas aeruginosa. Microbiology 1988, 134, 43–52. [Google Scholar] [CrossRef]
  25. Morrison, M.D.; Fajardo-Cavazos, P.; Nicholson, W.L. Comparison of bacillus subtilis transcriptome profiles from two separate missions to the International Space Station. NPJ Microgravity 2019, 5, 1. [Google Scholar] [CrossRef]
  26. Guo, Y.; Li, J.; Liu, J.; Wang, T.; Li, Y.; Yuan, Y.; Zhao, J.; Chang, D.; Fang, X.; Li, T.; et al. Effects of space environment on genome, transcriptome, and proteome of klebsiella pneumoniae. Arch. Med. Res. 2015, 46, 609–618. [Google Scholar] [CrossRef]
  27. Li, J.; Liu, F.; Wang, Q.; Ge, P.; Woo, P.C.; Yan, J.; Zhao, Y.; Gao, G.F.; Liu, C.H.; Liu, C. Genomic and transcriptomic analysis of NDM-1 Klebsiella pneumoniae in spaceflight reveal mechanisms underlying environmental adaptability. Sci. Rep. 2014, 4, 6216. [Google Scholar] [CrossRef]
  28. Schiwon, K.; Arends, K.; Rogowski, K.M.; Furch, S.; Prescha, K.; Sakinc, T.; Van Houdt, R.; Werner, G.; Grohmann, E. Comparison of antibiotic resistance, biofilm formation and conjugative transfer of Staphylococcus and Enterococcus isolates from International Space Station and Antarctic Research Station Concordia. Microb. Ecol. 2013, 65, 638–651. [Google Scholar] [CrossRef]
  29. Checinska Sielaff, A.; Urbaniak, C.; Mohan, G.B.M.; Stepanov, V.G.; Tran, Q.; Wood, J.M.; Minich, J.; McDonald, D.; Mayer, T.; Knight, R.; et al. Characterization of the total and viable bacterial and fungal communities associated with the international space station surfaces. Microbiome 2019, 7, 50. [Google Scholar] [CrossRef]
  30. Taylor, G.R.; Henney, M.R.; Ellis, W.L. Changes in the fungal autoflora of apollo astronauts. Appl. Microbiol. 1973, 26, 804–813. [Google Scholar] [CrossRef]
  31. Taylor, G.R. Recovery of medically important microorganisms from apollo astronauts. Aerosp. Med. 1974, 45, 824–828. [Google Scholar] [PubMed]
  32. Crowe, J.H.; Hoekstra, F.A.; Crowe, L.M. Anhydrobiosis. Annu. Rev. Physiol. 1992, 54, 579–599. [Google Scholar] [CrossRef] [PubMed]
  33. Crowe, L.M.; Crowe, J.H. Anhydrobiosis: A strategy for survival. Adv. Space Res. 1992, 12, 239–247. [Google Scholar] [CrossRef]
  34. Billi, D.; Staibano, C.; Verseux, C.; Fagliarone, C.; Mosca, C.; Baque, M.; Rabbow, E.; Rettberg, P. Dried biofilms of desert strains of chroococcidiopsis survived prolonged exposure to space and mars-like conditions in low earth orbit. Astrobiology 2019, 19, 1008–1017. [Google Scholar] [CrossRef] [PubMed]
  35. Horneck, G.; Bucker, H.; Reitz, G. Long-term survival of bacterial spores in space. Adv. Space Res. 1994, 14, 41–45. [Google Scholar] [CrossRef]
  36. Roman, M.C.; Minton-Summers, S. Assessment of biofilm formation in the international space station water recovery and management system. Life Support Biosph. Sci. 1998, 5, 45–51. [Google Scholar] [PubMed]
  37. Gonzales, A.; Schuerger, A.; Barford, C.; Mitchell, R. Engineering strategies for the design of plant nutrient delivery systems for use in space: Approaches to countering microbiological contamination. Adv. Space Res. 1996, 18, 5–20. [Google Scholar] [CrossRef]
  38. Song, B.; Leff, L.G. Identification and characterization of bacterial isolates from the Mir space station. Microbiol. Res. 2005, 160, 111–117. [Google Scholar] [CrossRef]
  39. Wang, H.; Yan, Y.; Rong, D.; Wang, J.; Wang, H.; Liu, Z.; Wang, J.; Yang, R.; Han, Y. Increased biofilm formation ability in Klebsiella pneumoniae after short-term exposure to a simulated microgravity environment. Microbiologyopen 2016, 5, 793–801. [Google Scholar] [CrossRef]
  40. Mehta, P.; Bhayani, D. Impact of space environment on stability of medicines: Challenges and prospects. J. Pharm. Biomed. Anal. 2017, 136, 111–119. [Google Scholar] [CrossRef]
  41. Rettberg, P.; Horneck, G.; Zittermann, A.; Heer, M. Biological dosimetry to determine the UV radiation climate inside the MIR station and its role in vitamin D biosynthesis. Adv. Space Res. 1998, 22, 1643–1652. [Google Scholar] [CrossRef]
  42. Rettberg, P.; Horneck, G. Biologically weighted measurement of UV radiation in space and on Earth with the biofilm technique. Adv. Space Res. 2000, 26, 2005–2014. [Google Scholar] [CrossRef]
  43. De Vos, W.M. Microbial biofilms and the human intestinal microbiome. NPJ Biofilms Microbiomes 2015, 1, 15005. [Google Scholar] [CrossRef] [PubMed]
  44. Macfarlane, S.; Dillon, J. Microbial biofilms in the human gastrointestinal tract. J. Appl. Microbiol. 2007, 102, 1187–1196. [Google Scholar] [CrossRef]
  45. Cao, W.; Zhang, H.; Wang, Y.; Pan, J. Bioremediation of polluted surface water by using biofilms on filamentous bamboo. Ecol. Eng. 2012, 42, 146–149. [Google Scholar] [CrossRef]
  46. Xu, X.-Y.; Feng, L.-J.; Zhu, L.; Xu, J.; Ding, W.; Qi, H.-Y. Biofilm formation and microbial community analysis of the simulated river bioreactor for contaminated source water remediation. Environ. Sci. Pollut. Res. 2012, 19, 1584–1593. [Google Scholar] [CrossRef]
  47. Seneviratne, C.J.; Zhang, C.F.; Samaranayake, L.P. Dental plaque biofilm in oral health and disease. Chin. J. Dent. Res. 2011, 14, 87. [Google Scholar]
  48. Ichikawa, K.; Nakamura, H.K.; Ogawa, N.; Sakimura, T.; Kuroda, M. R&D of long-term life support system by using electrochemically activated biofilm reactor of aquatic animals for space examinations. Biol. Sci. Space 1999, 13, 348–350. [Google Scholar]
  49. Paredes, J.; Alonso-Arce, M.; Schmidt, C.; Valderas, D.; Sedano, B.; Legarda, J.; Arizti, F.; Gomez, E.; Aguinaga, A.; Del Pozo, J.L.; et al. Smart central venous port for early detection of bacterial biofilm related infections. Biomed. Microdevices 2014, 16, 365–374.
More
Information
Subjects: Microbiology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 994
Revisions: 4 times (View History)
Update Date: 03 Nov 2020
1000/1000