Yonsei Med J.  2018 Mar;59(2):176-186. 10.3349/ymj.2018.59.2.176.

Systems Biology-Based Platforms to Accelerate Research of Emerging Infectious Diseases

Affiliations
  • 1Department of Biomedical Sciences, College of Medicine, Korea University Guro Hospital, Seoul, Korea. oshin@korea.ac.kr
  • 2College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Korea.

Abstract

Emerging infectious diseases (EIDs) pose a major threat to public health and security. Given the dynamic nature and significant impact of EIDs, the most effective way to prevent and protect against them is to develop vaccines in advance. Systems biology approaches provide an integrative way to understand the complex immune response to pathogens. They can lead to a greater understanding of EID pathogenesis and facilitate the evaluation of newly developed vaccine-induced immunity in a timely manner. In recent years, advances in high throughput technologies have enabled researchers to successfully apply systems biology methods to analyze immune responses to a variety of pathogens and vaccines. Despite recent advances, computational and biological challenges impede wider application of systems biology approaches. This review highlights recent advances in the fields of systems immunology and vaccinology, and presents ways that systems biology-based platforms can be applied to accelerate a deeper understanding of the molecular mechanisms of immunity against EIDs.

Keyword

Emerging infectious diseases; systems biology; systems vaccinology; vaccines; immunity

MeSH Terms

*Communicable Diseases, Emerging
Humans
*Immunity
Research
Systems Biology/*methods
Vaccines/*immunology
Vaccines

Figure

  • Fig. 1 The process of systems vaccinology approaches. Systems biology approaches applied to clinical trials can lead to the generation of new hypotheses that can be tested and ultimately lead to better vaccine development. For example, correlates of vaccine-induced immunity in clinical trials can be profiled in detail with high-throughput technologies, such as RNA-sequencing, proteomics, and metabolomics. The high-throughput data thus generated can be integrated using bioinformatics tools and used to create hypotheses about the biological mechanisms underlying vaccine efficacy and immunogenicity. Such hypotheses can then be tested with animal models or in vitro human systems. The insights gained from experimentation can then guide the identification of biomarker and the design and development of new vaccines. Thus, systems vaccinology approaches can provide translational solutions for novel and enhanced vaccine development.

  • Fig. 2 Potential biases for systems vaccinology data analysis. The application of systems biology approaches to the fields of immunology and vaccinology faces many challenges and potential limitations. The potentially challenging factors for systems vaccinology analysis can be divided into different categories, such as cellular, population, molecular, individual, and technical aspects.

  • Fig. 3 Use of reverse vaccinology tools in vaccine design. The workflow of vaccine design using reverse vaccinology is shown. First, B cells or plasma cells can be isolated from infected or vaccinated individuals and PCR amplification of antibody gene heavy and light chains can lead to human B cell repertoire analysis, which enables the identification of protective antibodies. Using sequence information of antibody variable regions, the interaction of antibodies with their target antigen can be structurally characterized and used to predict the protective epitope using crystallography. The protective epitope can then be engineered to produce an optimized immunogen with enhanced delivery format, such as nanoparticle technologies or adjuvants. Finally, this new reverse vaccinology-based antigen can be tested in humans.


Reference

1. Morse SS, Schluederberg A. From the National Institute of Allergy and Infectious Diseases, the Fogarty International Center of the National Institutes of Health, and the Rockefeller University. Emerging viruses: the evolution of viruses and viral diseases. J Infect Dis. 1990; 162:1–7.
2. Morse SS, Hughes JM. Developing an integrated epidemiologic approach to emerging infectious diseases. Epidemiol Rev. 1996; 18:1–3.
Article
3. Woolhouse M, Scott F, Hudson Z, Howey R, Chase-Topping M. Human viruses: discovery and emergence. Philos Trans R Soc Lond B Biol Sci. 2012; 367:2864–2871.
Article
4. Woolhouse ME. Population biology of emerging and re-emerging pathogens. Trends Microbiol. 2002; 10:10 Suppl. S3–S7.
Article
5. Woolhouse ME, Gowtage-Sequeria S. Host range and emerging and reemerging pathogens. Emerg Infect Dis. 2005; 11:1842–1847.
Article
6. Grimm SK, Ackerman ME. Vaccine design: emerging concepts and renewed optimism. Curr Opin Biotechnol. 2013; 24:1078–1088.
Article
7. Nakaya HI, Pulendran B. Vaccinology in the era of high-throughput biology. Philos Trans R Soc Lond B Biol Sci. 2015; 370:20140146.
Article
8. Koff WC, Burton DR, Johnson PR, Walker BD, King CR, Nabel GJ, et al. Accelerating next-generation vaccine development for global disease prevention. Science. 2013; 340:1232910.
Article
9. Westerhoff HV, Winder C, Messiha H, Simeonidis E, Adamczyk M, Verma M, et al. Systems biology: the elements and principles of life. FEBS Lett. 2009; 583:3882–3890.
Article
10. Nakaya HI, Li S, Pulendran B. Systems vaccinology: learning to compute the behavior of vaccine induced immunity. Wiley Interdiscip Rev Syst Biol Med. 2012; 4:193–205.
Article
11. Li S, Nakaya HI, Kazmin DA, Oh JZ, Pulendran B. Systems biological approaches to measure and understand vaccine immunity in humans. Semin Immunol. 2013; 25:209–218.
Article
12. Pulendran B, Li S, Nakaya HI. Systems vaccinology. Immunity. 2010; 33:516–529.
Article
13. Pulendran B, Oh JZ, Nakaya HI, Ravindran R, Kazmin DA. Immunity to viruses: learning from successful human vaccines. Immunol Rev. 2013; 255:243–255.
Article
14. Almeida SL. Trending now: re-emerging infectious disease update. J Emerg Nurs. 2015; 41:104–108.
Article
15. Howard CR, Fletcher NF. Emerging virus diseases: can we ever expect the unexpected? Emerg Microbes Infect. 2012; 1:e46.
Article
16. Dash AP, Bhatia R, Sunyoto T, Mourya DT. Emerging and re-emerging arboviral diseases in Southeast Asia. J Vector Borne Dis. 2013; 50:77–84.
17. Morse SS. Factors in the emergence of infectious diseases. Emerg Infect Dis. 1995; 1:7–15.
Article
18. Lauring AS, Frydman J, Andino R. The role of mutational robustness in RNA virus evolution. Nat Rev Microbiol. 2013; 11:327–336.
Article
19. Webster RG, Bean WJ, Gorman OT, Chambers TM, Kawaoka Y. Evolution and ecology of influenza A viruses. Microbiol Rev. 1992; 56:152–179.
Article
20. Yoon SW, Webby RJ, Webster RG. Evolution and ecology of influenza A viruses. Curr Top Microbiol Immunol. 2014; 385:359–375.
Article
21. Short KR, Richard M, Verhagen JH, van Riel D, Schrauwen EJ, van den Brand JM, et al. One health, multiple challenges: the inter-species transmission of influenza A virus. One Health. 2015; 1:1–13.
Article
22. Wilson ME. Travel and the emergence of infectious diseases. Emerg Infect Dis. 1995; 1:39–46.
Article
23. Wilson ME. The traveller and emerging infections: sentinel, courier, transmitter. J Appl Microbiol. 2003; 94:1S–11S.
Article
24. Hwang GM, Mahoney PJ, James JH, Lin GC, Berro AD, Keybl MA, et al. A model-based tool to predict the propagation of infectious disease via airports. Travel Med Infect Dis. 2012; 10:32–42.
Article
25. Cho SY, Kang JM, Ha YE, Park GE, Lee JY, Ko JH, et al. MERS-CoV outbreak following a single patient exposure in an emergency room in South Korea: an epidemiological outbreak study. Lancet. 2016; 388:994–1001.
Article
26. Park WB, Perera RA, Choe PG, Lau EH, Choi SJ, Chun JY, et al. Kinetics of serologic responses to MERS coronavirus infection in humans, South Korea. Emerg Infect Dis. 2015; 21:2186–2189.
Article
27. Beugnet F, Chalvet-Monfray K. Impact of climate change in the epidemiology of vector-borne diseases in domestic carnivores. Comp Immunol Microbiol Infect Dis. 2013; 36:559–566.
Article
28. Medlock JM, Hansford KM, Versteirt V, Cull B, Kampen H, Fontenille D, et al. An entomological review of invasive mosquitoes in Europe. Bull Entomol Res. 2015; 105:637–663.
Article
29. Murray NE, Quam MB, Wilder-Smith A. Epidemiology of dengue: past, present and future prospects. Clin Epidemiol. 2013; 5:299–309.
30. Stone BL, Tourand Y, Brissette CA. Brave new worlds: the expanding universe of Lyme disease. Vector Borne Zoonotic Dis. 2017; 17:619–629.
Article
31. Ostfeld RS, Brunner JL. Climate change and Ixodes tick-borne diseases of humans. Philos Trans R Soc Lond B Biol Sci. 2015; 370:20140051.
32. Choi YK, Pascua PN, Song MS. Swine influenza viruses: an Asian perspective. Curr Top Microbiol Immunol. 2013; 370:147–172.
Article
33. Chen R, Holmes EC. Avian influenza virus exhibits rapid evolutionary dynamics. Mol Biol Evol. 2006; 23:2336–2341.
Article
34. Su S, Gu M, Liu D, Cui J, Gao GF, Zhou J, et al. Epidemiology, evolution, and pathogenesis of H7N9 Influenza viruses in five epidemic waves since 2013 in China. Trends Microbiol. 2017; 25:713–728.
Article
35. Wang X, Jiang H, Wu P, Uyeki TM, Feng L, Lai S, et al. Epidemiology of avian influenza A H7N9 virus in human beings across five epidemics in mainland China, 2013-17: an epidemiological study of laboratory-confirmed case series. Lancet Infect Dis. 2017; 17:822–832.
Article
36. Liu Q, He B, Huang SY, Wei F, Zhu XQ. Severe fever with thrombocytopenia syndrome, an emerging tick-borne zoonosis. Lancet Infect Dis. 2014; 14:763–772.
Article
37. Zhan J, Wang Q, Cheng J, Hu B, Li J, Zhan F, et al. Current status of severe fever with thrombocytopenia syndrome in China. Virol Sin. 2017; 32:51–62.
Article
38. Yun SM, Lee WG, Ryou J, Yang SC, Park SW, Roh JY, et al. Severe fever with thrombocytopenia syndrome virus in ticks collected from humans, South Korea, 2013. Emerg Infect Dis. 2014; 20:1358–1361.
Article
39. Takahashi T, Maeda K, Suzuki T, Ishido A, Shigeoka T, Tominaga T, et al. The first identification and retrospective study of Severe Fever with Thrombocytopenia Syndrome in Japan. J Infect Dis. 2014; 209:816–827.
Article
40. Shin SY, Cho OH, Bae IG. Bone marrow suppression and hemophagocytic histiocytes are common findings in Korean severe fever with thrombocytopenia syndrome patients. Yonsei Med J. 2016; 57:1286–1289.
Article
41. Oh WS, Yoo JR, Kwon KT, Kim HI, Lee SJ, Jun JB, et al. Effect of early plasma exchange on survival in patients with severe fever with thrombocytopenia syndrome: a multicenter study. Yonsei Med J. 2017; 58:867–871.
Article
42. Gai ZT, Zhang Y, Liang MF, Jin C, Zhang S, Zhu CB, et al. Clinical progress and risk factors for death in severe fever with thrombocytopenia syndrome patients. J Infect Dis. 2012; 206:1095–1102.
Article
43. Kim WY, Choi W, Park SW, Wang EB, Lee WJ, Jee Y, et al. Nosocomial transmission of severe fever with thrombocytopenia syndrome in Korea. Clin Infect Dis. 2015; 60:1681–1683.
Article
44. Bao CJ, Guo XL, Qi X, Hu JL, Zhou MH, Varma JK, et al. A family cluster of infections by a newly recognized bunyavirus in eastern China, 2007: further evidence of person-to-person transmission. Clin Infect Dis. 2011; 53:1208–1214.
Article
45. Tang X, Wu W, Wang H, Du Y, Liu L, Kang K, et al. Human-to-human transmission of severe fever with thrombocytopenia syndrome bunyavirus through contact with infectious blood. J Infect Dis. 2013; 207:736–739.
Article
46. Zaki AM, van Boheemen S, Bestebroer TM, Osterhaus AD, Fouchier RA. Isolation of a novel coronavirus from a man with pneumonia in Saudi Arabia. N Engl J Med. 2012; 367:1814–1820.
Article
47. Azhar EI, Lanini S, Ippolito G, Zumla A. The middle east respiratory syndrome coronavirus-a continuing risk to global health security. Adv Exp Med Biol. 2017; 972:49–60.
48. de Wit E, van Doremalen N, Falzarano D, Munster VJ. SARS and MERS: recent insights into emerging coronaviruses. Nat Rev Microbiol. 2016; 14:523–534.
Article
49. Memish ZA, Mishra N, Olival KJ, Fagbo SF, Kapoor V, Epstein JH, et al. Middle East respiratory syndrome coronavirus in bats, Saudi Arabia. Emerg Infect Dis. 2013; 19:1819–1823.
Article
50. Gubler DJ. Dengue, urbanization and globalization: the unholy trinity of the 21(st) century. Trop Med Health. 2011; 39:4 Suppl. 3–11.
Article
51. Diamond MS, Pierson TC. Molecular insight into dengue virus pathogenesis and its implications for disease control. Cell. 2015; 162:488–492.
Article
52. Halstead SB. Dengue. Lancet. 2007; 370:1644–1652.
Article
53. Park JH, Lee DW. Dengue fever in South Korea, 2006-2010. Emerg Infect Dis. 2012; 18:1525–1527.
Article
54. Miki S, Lee WC, Lee MJ. A comparative study of the trends of imported Dengue cases in Korea and Japan 2011-2015. J Clin Med Res. 2017; 9:650–653.
Article
55. Musso D, Gubler DJ. Zika virus. Clin Microbiol Rev. 2016; 29:487–524.
Article
56. Mlakar J, Korva M, Tul N, Popović M, Poljšak-Prijatelj M, Mraz J, et al. Zika virus associated with microcephaly. N Engl J Med. 2016; 374:951–958.
Article
57. Lim SK, Lim JK, Yoon IK. An update on Zika virus in Asia. Infect Chemother. 2017; 49:91–100.
Article
58. Jang HC, Park WB, Kim UJ, Chun JY, Choi SJ, Choe PG, et al. First imported case of Zika virus infection into Korea. J Korean Med Sci. 2016; 31:1173–1177.
Article
59. Aderem A, Adkins JN, Ansong C, Galagan J, Kaiser S, Korth MJ, et al. A systems biology approach to infectious disease research: innovating the pathogen-host research paradigm. MBio. 2011; 2:e00325–e00310.
Article
60. Mei B, Ding X, Xu HZ, Wang MT. Global gene expression changes in human peripheral blood after H7N9 infection. Gene. 2014; 551:255–260.
Article
61. Wang Y, Lupiani B, Reddy SM, Lamont SJ, Zhou H. RNA-seq analysis revealed novel genes and signaling pathway associated with disease resistance to avian influenza virus infection in chickens. Poult Sci. 2014; 93:485–493.
Article
62. Ertl R, Klein D. Transcriptional profiling of the host cell response to feline immunodeficiency virus infection. Virol J. 2014; 11:52.
Article
63. Jones M, Dry IR, Frampton D, Singh M, Kanda RK, Yee MB, et al. RNA-seq analysis of host and viral gene expression highlights interaction between varicella zoster virus and keratinocyte differentiation. PLoS Pathog. 2014; 10:e1003896.
Article
64. Rossetto CC, Tarrant-Elorza M, Verma S, Purushothaman P, Pari GS. Regulation of viral and cellular gene expression by Kaposi’s sarcoma-associated herpesvirus polyadenylated nuclear RNA. J Virol. 2013; 87:5540–5553.
Article
65. Juranic Lisnic V, Babic Cac M, Lisnic B, Trsan T, Mefferd A, Das Mukhopadhyay C, et al. Dual analysis of the murine cytomegalovirus and host cell transcriptomes reveal new aspects of the virus-host cell interface. PLoS Pathog. 2013; 9:e1003611.
Article
66. Park SJ, Kumar M, Kwon HI, Seong RK, Han K, Song JM, et al. Dynamic changes in host gene expression associated with H5N8 avian influenza virus infection in mice. Sci Rep. 2015; 5:16512.
Article
67. Nowakowski TJ, Pollen AA, Di Lullo E, Sandoval-Espinosa C, Bershteyn M, Kriegstein AR. Expression analysis highlights AXL as a candidate Zika virus entry receptor in neural stem cells. Cell Stem Cell. 2016; 18:591–596.
Article
68. Onorati M, Li Z, Liu F, Sousa AMM, Nakagawa N, Li M, et al. Zika virus disrupts Phospho-TBK1 localization and mitosis in human neuroepithelial stem cells and radial glia. Cell Rep. 2016; 16:2576–2592.
Article
69. Ahmed N, Gottschalk S. How to design effective vaccines: lessons from an old success story. Expert Rev Vaccines. 2009; 8:543–546.
Article
70. Querec TD, Akondy RS, Lee EK, Cao W, Nakaya HI, Teuwen D, et al. Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat Immunol. 2009; 10:116–125.
Article
71. Nakaya HI, Wrammert J, Lee EK, Racioppi L, Marie-Kunze S, Haining WN, et al. Systems biology of vaccination for seasonal influenza in humans. Nat Immunol. 2011; 12:786–795.
Article
72. Oh JZ, Ravindran R, Chassaing B, Carvalho FA, Maddur MS, Bower M, et al. TLR5-mediated sensing of gut microbiota is necessary for antibody responses to seasonal influenza vaccination. Immunity. 2014; 41:478–492.
Article
73. Ravindran R, Khan N, Nakaya HI, Li S, Loebbermann J, Maddur MS, et al. Vaccine activation of the nutrient sensor GCN2 in dendritic cells enhances antigen presentation. Science. 2014; 343:313–317.
Article
74. Nakaya HI, Hagan T, Duraisingham SS, Lee EK, Kwissa M, Rouphael N, et al. Systems analysis of immunity to influenza vaccination across multiple years and in diverse populations reveals shared molecular signatures. Immunity. 2015; 43:1186–1198.
Article
75. Nakaya HI, Clutterbuck E, Kazmin D, Wang L, Cortese M, Bosinger SE, et al. Systems biology of immunity to MF59-adjuvanted versus nonadjuvanted trivalent seasonal influenza vaccines in early childhood. Proc Natl Acad Sci U S A. 2016; 113:1853–1858.
Article
76. Kazmin D, Nakaya HI, Lee EK, Johnson MJ, van der Most R, van den Berg RA, et al. Systems analysis of protective immune responses to RTS,S malaria vaccination in humans. Proc Natl Acad Sci U S A. 2017; 114:2425–2430.
Article
77. Li S, Sullivan NL, Rouphael N, Yu T, Banton S, Maddur MS, et al. Metabolic phenotypes of response to vaccination in humans. Cell. 2017; 169:862–877.e17.
Article
78. Hoek KL, Samir P, Howard LM, Niu X, Prasad N, Galassie A, et al. A cell-based systems biology assessment of human blood to monitor immune responses after influenza vaccination. PLoS One. 2015; 10:e0118528.
Article
79. Mizukami T, Momose H, Kuramitsu M, Takizawa K, Araki K, Furuhata K, et al. System vaccinology for the evaluation of influenza vaccine safety by multiplex gene detection of novel biomarkers in a preclinical study and batch release test. PLoS One. 2014; 9:e101835.
Article
80. Bucasas KL, Franco LM, Shaw CA, Bray MS, Wells JM, Niño D, et al. Early patterns of gene expression correlate with the humoral immune response to influenza vaccination in humans. J Infect Dis. 2011; 203:921–929.
Article
81. Pulendran B. Learning immunology from the yellow fever vaccine: innate immunity to systems vaccinology. Nat Rev Immunol. 2009; 9:741–747.
Article
82. Gaucher D, Therrien R, Kettaf N, Angermann BR, Boucher G, Filali-Mouhim A, et al. Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses. J Exp Med. 2008; 205:3119–3131.
Article
83. O'Hagan DT, Friedland LR, Hanon E, Didierlaurent AM. Towards an evidence based approach for the development of adjuvanted vaccines. Curr Opin Immunol. 2017; 47:93–102.
84. Vesikari T, Knuf M, Wutzler P, Karvonen A, Kieninger-Baum D, Schmitt HJ, et al. Oil-in-water emulsion adjuvant with influenza vaccine in young children. N Engl J Med. 2011; 365:1406–1416.
Article
85. Vesikari T, Forstên A, Herbinger KH, Cioppa GD, Beygo J, Borkowski A, et al. Safety and immunogenicity of an MF59(®)-adjuvanted A/H5N1 pre-pandemic influenza vaccine in adults and the elderly. Vaccine. 2012; 30:1388–1396.
Article
86. Six A, Bellier B, Thomas-Vaslin V, Klatzmann D. Systems biology in vaccine design. Microb Biotechnol. 2012; 5:295–304.
Article
87. Henn AD, Wu S, Qiu X, Ruda M, Stover M, Yang H, et al. High-resolution temporal response patterns to influenza vaccine reveal a distinct human plasma cell gene signature. Sci Rep. 2013; 3:2327.
Article
88. Chaussabel D, Pascual V, Banchereau J. Assessing the human immune system through blood transcriptomics. BMC Biol. 2010; 8:84.
Article
89. Rappuoli R. Reverse vaccinology. Curr Opin Microbiol. 2000; 3:445–450.
Article
90. Rappuoli R, Black S, Lambert PH. Vaccine discovery and translation of new vaccine technology. Lancet. 2011; 378:360–368.
Article
91. Korber B, LaBute M, Yusim K. Immunoinformatics comes of age. PLoS Comput Biol. 2006; 2:e71.
Article
92. Sievers SA, Scharf L, West AP Jr, Bjorkman PJ. Antibody engineering for increased potency, breadth and half-life. Curr Opin HIV AIDS. 2015; 10:151–159.
Article
Full Text Links
  • YMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr