Skip to main content

EDITORIAL article

Front. Vet. Sci., 11 September 2023
Sec. Veterinary Infectious Diseases
This article is part of the Research Topic Advances In Host-Pathogen Interactions For Diseases In Animals And Birds View all 9 articles

Editorial: Advances in host-pathogen interactions for diseases in animals and birds

\r\nMrigendra Rajput
Mrigendra Rajput*Neelu ThakurNeelu Thakur
  • Department of Biology, University of Dayton, Dayton, OH, United States

Introduction

Host-virus interactions are complex cross-talk between the virus and its host cell. These interactions are not limited to causing disease or changing the physiology of host cells but they also influence viruses. Studies showed bidirectional changes in viruses and their host which co-evolved together (1, 2). The host body harbors numerous microbes, collectively it is called the microbiome (3, 4). However, the majority of microbiome studies are focused on bacterial populations, missing important viral components which is also known as virome (5). It is estimated that the number of viruses in the host is around ten times more than the host-inhabited bacterial population (6). Interestingly, virus or bacteriophages significantly influence bacterial population or their physiology in the host (7). Studies also indicated that virulence factors in bacteria are acquired from their bacteriophage (810). The overall influence of the virus on the host body and ecosystem is very vast and complex. But the current topic focuses on the interactions between vertebrate hosts like animals with their viruses.

The majority of viruses are commensal in the host's body

The presence of virus in the host body does not always cause diseases. A majority of viruses present in the host body are commensal (11). Host body contains both DNA and RNA virus (12, 13), and these viruses can be localized on the skin (14), peripheral blood (15, 16), or internal organs such as the lungs, liver, spleen, kidney, or heart (17). The number and type of virus in the virome are greatly influenced by the environment, dietary practices, and their location in the body (18, 19). Despite their importance in shaping the overall health of the host, we have very limited information about our virome and its interactions with host cells. The majority of virome study is limited to metagenomic sequencing, which is trying the uncover the least explored part of virome or “viral dark matter” and its potential role in the evolution and shaping of the overall health and immune system of the host (20, 21).

Virus and its symbiotic relation with its host

There are several pieces of evidence showed that the virus and its host can benefit each other. A study with tropical panic grass, fungal endophyte, and fungal virus showed a three-way symbiosis. Where virus-infected fungus confers heat tolerance to both, tropical panic grass and fungal endophyte. Virus-free fungi were unable to confer heat tolerance, but heat tolerance was restored after the virus is reintroduction to fungi (22). Similarly, a study with parasitoid wasp showed obligatory mutualism between virus and parasitoid wasps. Successful development of the parasitoid wasp's egg within the host depends on the presence of the virus which suppresses the host's immune response (23).

Herpesvirus are important virus family which infect humans and animals and they could cause lifelong latency in the host (2426). It is found that herpesvirus latency provides resistance to bacterial infections such as Listeria monocytogenes and Yersinia pestis by activating the host immune response (27). Similarly, bacteriophage plays an important role in regulating bacterial population in the host and participating in immune system development, and maintaining immune homeostasis in the body organ such as in the intestine (2830).

Long-term interactions between the virus and its host left traces in the host genome

The genome of humans and animals encodes from a few to 100,000 different genes (31, 32). It is estimated that around 8% or around one-tenth of the human gene contains pieces of viral DNA (33). These viral DNA fragments incorporated in the host is termed as endogenous viral elements (EVEs) (34). EVEs provide valuable information about their evolution over the time, their host, and geographical distribution (35, 36).

Integration of viral DNA into the host genome is not always harmful. Evidence showed that the presence of EVEs provides antiviral defense against viral infection by encoding several genes such as ribonuclease H (RNase H) or small interfering RNAs (siRNAs) which silence the viral gene in subsequent infection (35, 37, 38). Interestingly, several EVEs such as the Arc gene, play an important role in cognitive function and help to store long-term memory in the host (39, 40). Another study revealed that host cells can use EVEs to induce immunity against tumors (41). Cellular p53 is a nuclear transcription factor with pro-apoptotic function. Cellular stresses such as DNA damage activates p53 and activated p53 promotes cell cycle arrest that allows time for DNA repair or causes apoptosis in the damaged cells (42). Studies showed that p53 activation also increased the expression of EVEs and EVE's expression induced host immune response in the form of higher IFN production, increased T-cell activity, and reduced allograft tumor (41). However, the presence of EVEs is not always beneficial to the host. Studies showed that individuals with inherited chromosomally integrated human herpesvirus 6 (iciHHV-6) gene are more susceptible to angina, a type of chest pain that happens when heart does not receive enough oxygen-rich blood (43).

Host-virus interaction and its outcome

Translocation of virus particles to the host cell is the first step in virus infection. This translocation is initiated by the interaction of distinct molecules present on the outer layer of the virus with specific receptors on the host cell (44). This initial interaction provides signals to the cells which further enhance the virus translocation to the host cell either by receptor-mediated endocytosis or membrane fusion, these mechanism varies widely among different virus families (45, 46). In the host cell, virus releases its genetic material for replication by fusing the viral capsid with the host cell's phagosome or creating pores in the viral capsid by utilizing cellular or viral machinery (47, 48). During this process host immune system recognize the virus by its pattern recognition receptors (PRRs) which specifically interact with pathogen-associated molecular patterns (PAMPs) present in the virus. This recognition triggers a cascade of innate immune responses, including the secretion of interferons/cytokines and the activation of immune cells (49). The outcome of this host–viral interaction depends on several host or viral factors (50). These outcomes can result in the clearing of virus by host immune response (5153), or host immune evasion by virus (54), or establishing acute, chronic, persistent, or latent infection (5557). It is found that a high mutation rate in viruses helps them to evolve, survive and escape from the host's immune response. While these changes may also be responsible for shifting in virus host range (5860).

Damage in viral disease

The outcome of viral disease is greatly influenced by viral factors such as virulence and host factors like susceptibility. Broadly, damage in the host during virus infection can be divided into two, (A) direct damage by the virus when virus replicates in the host cell and cause damage in cells either by hijacking cellular machinery, changing cellular physiology, or damaging cellular components by its structural or non-structural proteins (6163). (B) The virus can also cause indirect damage to the host cell by virus-induced hyperinflammation (64, 65). Virus-induced uncontrol-hyperinflammation has been associated with higher mortality in several virus infections (6668).

Cellular factors in restricting virus replication

Host cells have several inherent antiviral factors such as antiviral proteins which control virus replication. A few of the antiviral proteins are APOBEC3G (Apolipoprotein B mRNA Editing Enzyme, Catalytic Polypeptide-like 3G), ZAP (Zinc Finger Antiviral Protein), SAMHD1 (SAM domain and HD domain-containing protein 1), 2′-5′-Oligoadenylate Synthetase (OAS), Mx Proteins and Tetherin, which is also known as BST-2 (bone marrow stromal antigen 2) (6975).

APOBEC3G exhibits its antiviral effect by inducing hypermutation in viral genomes. APOBEC3G catalyzes the deamination of cytosine (dC) to uracil (dU) in single-stranded DNA, leading to non-functional viral DNA and prevents virus replication (76). While ZAP degrades the viral RNA by binding with its poly A tail. This binding leads to deadenylation of viral RNA and viral RNA is further degraded by exosomes (77). ZAP also selectively recognizes CG-rich viral RNAs and degrades viral RNA by exosomes (70). Additionally, ZAP could suppress virus replication by inducing an antiviral immune response in the host (78).

SAMHD1, another cellular protein exhibits its antiviral effect by hydrolyzing nucleotide triphosphates (dNTPs), dNTPs are needed by the virus to synthesize its genome while the lower concentration of dNTP in cells suppress the virus replication (79). However, a recent study also showed that SAMHD1 can suppress the antiviral host immune response by inhibiting NF-κB (Nuclear factor kappa-light-chain-enhancer of activated B cells) activation, suppressing IFN-I (type-I interferons) response and potentially interfering in virus-specific antibody production (80).

2′-5′-Oligoadenylate Synthetase (OAS), activates RNase L, an endoribonuclease that cleaves single-stranded viral RNAs and suppresses its replication (81), while Mx proteins, upon activation, interact with viral nucleocapsid proteins and disrupts viral replication complexes and thus inhibits the viral life cycle (82). Tetherin, on the other hand, exhibits its antiviral effect by physically “tethering” newly synthesized virus and present its release (83). Above mentioned proteins are a few of the cellular proteins which play an important role in suppressing virus replication. Potentially, there may be numerous more cellular molecules including small interfering RNA (siRNA) which suppress virus replication in the host cell. These molecules need to be studied in detail to understand the underlying pathways and their role in host-virus interactions in suppressing virus replication.

Summary

Host-virus interaction studies provide valuable information about viruses, and their molecular characteristics which facilitate them to mutate, change their virulence, and shift host range. Additionally, these studies help in understanding the effect of virus infection on host cells, cell organelles, and the physiological/metabolic activity of the cells. These studies also help in identifying the cellular molecules which could have antiviral properties. These molecules could be used to suppress the broad range of pathogenic viruses.

Author contributions

MR: Writing—original draft, Writing—review and editing. NT: Writing—original draft, Writing—review and editing.

Acknowledgments

Authors thank Dr. Christopher Chase, South Dakota State University, Brookings, SD for providing valuable information for writing this article. We also thank all the authors who contributed their valuable work to this Research Topic. We appreciate all the reviewers and Research Topic editorial team for their valuable time in reviewing the manuscripts and providing constructive comments. We also want to show our gratitude to the editorial board for approving this topic and we hope this Research Topic will further improve our understanding of host-virus interaction, its effect host body, and on the virus.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Bohálová N, Cantara A, Bartas M, Kaura P, Štastný J, Pečinka P, et al. Tracing dsDNA virus–host coevolution through correlation of their G-Quadruplex-forming sequences. Int J Mol Sci. (2021) 22:3433. doi: 10.3390/ijms22073433

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Lobo FP, Mota BEF, Pena SDJ, Azevedo V, Macedo AM, Tauch A, et al. Virus-host coevolution: common patterns of nucleotide motif usage in Flaviviridae and their hosts. PLoS ONE. (2009) 4:e6282. doi: 10.1371/journal.pone.0006282

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Ursell LK, Metcalf JL, Parfrey LW, Knight R. Defining the human microbiome. Nutr Rev. (2012) 70 (Suppl. 1):S38–44. doi: 10.1111/j.1753-4887.2012.00493

CrossRef Full Text | Google Scholar

4. Bahrndorff S, Alemu T, Alemneh T, Nielsen JL. The microbiome of animals: implications for conservation biology. Int J Genomics. (2016) 2016:5304028. doi: 10.1155/2016/5304028

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Handley SA. The virome: a missing component of biological interaction networks in health and disease. Genome Med. (2016) 8:32. doi: 10.1186/s13073-016-0287-y

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Mukhopadhya I, Segal JP, Carding SR, Hart AL, Hold GL. The gut virome: the ‘missing link' between gut bacteria and host immunity? Therap Adv Gastroenterol. (2019) 12:1756284819836620. doi: 10.1177/1756284819836620

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Carding SR, Davis N, Hoyles L. Review article: the human intestinal virome in health and disease. Aliment Pharmacol Ther. (2017) 46:800–15. doi: 10.1111/apt.14280

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Plinkett G, Rose DJ, Durfee TJ, Blattner FR. Sequence of Shiga toxin 2 phage 933W from Escherichia coli O157:H7: Shiga toxin as a phage late-gene product. J Bacteriol. (1999) 181:1767–78. doi: 10.1128/JB.181.6.1767-1778.1999

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Pant A, Das B, Bhadra RK. CTX phage of Vibrio cholerae: genomics and applications. Vaccine. (2020) 38 (Suppl. 1):A7–A12. doi: 10.1016/j.vaccine.2019.06.034

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Rajadhyaksha AB, Srinivasa Rao S. The role of phage in the transduction of the toxinogenic factor in Corynebacterium diphtheriae. Microbiology. (1965) 40:421–9. doi: 10.1099/00221287-40-3-421

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Vu DL, Kaiser L. The concept of commensal viruses almost 20 years later: redefining borders in clinical virology. Clin Microbiol Infect. (2017) 23:688–90. doi: 10.1016/j.cmi.2017.03.005

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Kapusinszky B, Minor P, Delwart E. Nearly constant shedding of diverse enteric viruses by two healthy infants. J Clin Microbiol. (2012) 50:3427–34. doi: 10.1128/JCM.01589-12

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Harvey E, Holmes EC. Diversity and evolution of the animal virome. Nat Rev Microbiol. (2022) 20:321–34. doi: 10.1038/s41579-021-00665-x

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Hannigan GD, Meisel JS, Tyldsley AS, Zheng Q, Hodkinson BP, SanMiguel AJ, et al. The human skin double-stranded DNA virome: topographical and temporal diversity, genetic enrichment, and dynamic associations with the host microbiome. mBio. (2015) 1–13. doi: 10.1128/mBio.01578-15

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Moustafa A, Xie C, Kirkness E, Biggs W, Wong E, Turpaz Y, et al. The blood DNA virome in 8,000 humans. PLoS Pathog. (2017) 13:e1006292. doi: 10.1371/journal.ppat.1006292

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Stremlau MH, Andersen KG, Folarin OA, Grove JN, Odia I, Ehiane PE, et al. Discovery of novel rhabdoviruses in the blood of healthy individuals from West Africa. PLoS Negl Trop Dis. (2015) 9:e0003631. doi: 10.1371/journal.pntd.0003631

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Kumata R, Ito J, Takahashi K, Suzuki T, Sato K. A tissue level atlas of the healthy human virome. BMC Biol. (2020) 18:55. doi: 10.1186/s12915-020-00785-5

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Minot S, Sinha R, Chen J, Li H, Keilbaugh SA, Wu GD, et al. The human gut virome: inter-individual variation and dynamic response to diet. Genome Res. (2011) 21:1616–25. doi: 10.1101/gr.122705.111

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Garmaeva S, Gulyaeva A, Sinha T, Shkoporov AN, Clooney AG, Stockdale SR, et al. Stability of the human gut virome and effect of gluten-free diet. Cell Rep. (2021) 35:109132. doi: 10.1016/j.celrep.2021.109132

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Santiago-Rodriguez TM, Hollister EB. Unraveling the viral dark matter through viral metagenomics. Front Immunol. (2022) 13:1005107. doi: 10.3389/fimmu.2022.1005107

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Liang G, Bushman FD. The human virome: assembly, composition and host interactions. Nat Rev Microbiol. (2021) 19:514–27. doi: 10.1038/s41579-021-00536-5

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Márquez LM, Redman RS, Rodriguez RJ, Roossinck MJ. A virus in a fungus in a plant: three-way symbiosis required for thermal tolerance. Science. (2007) 315:513–5. doi: 10.1126/science.1136237

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Edson KM, Vinson SB, Stoltz DB, Summers MD. Virus in a parasitoid wasp: suppression of the cellular immune response in the parasitoid's host. Science. (1981) 211:582–3. doi: 10.1126/science.7455695

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Azab W, Dayaram A, Greenwood AD, Osterrieder N. How host specific are herpesviruses? Lessons from herpesviruses infecting wild and endangered mammals. Annu Rev Virol. (2018) 5:53–68. doi: 10.1146/annurev-virology-092917-043227

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Ostler JB, Jones C. The Bovine Herpesvirus 1 latency-reactivation cycle, a chronic problem in the cattle industry. Viruses. (2023) 15:552. doi: 10.3390/v15020552

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Cohen JI. Herpesvirus latency. J Clin Invest. (2020) 130:3361–9. doi: 10.1172/JCI136225

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Barton ES, White DW, Cathelyn JS, Brett-McClellan KA, Engle M, Diamond MS, et al. Herpesvirus latency confers symbiotic protection from bacterial infection. Nature. (2007) 447:326–9. doi: 10.1038/nature05762

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Liu L, Gong T, Tao W, Lin B, Li C, Zheng X, et al. Commensal viruses maintain intestinal intraepithelial lymphocytes via noncanonical RIG-I signaling. Nat Immunol. (2019) 20:1681–91. doi: 10.1038/s41590-019-0513-z

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Łusiak-Szelachowska M, Weber-Dabrowska B, Jończyk-Matysiak E, Wojciechowska R, Górski A. Bacteriophages in the gastrointestinal tract and their implications. Gut Pathog. (2017) 9:44. doi: 10.1186/s13099-017-0196-7

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Satyam R, Bhardwaj T, Jha NK, Jha SK, Nand P. Phage interaction with the mammalian immune system. In: Górski A, Miedzybrodzki R, Borysowski J, editors. Phage Therapy: A Practical Approach. Cham: Springer International Publishing (2019). p. 91–122.

Google Scholar

31. Salzberg SL. Open questions: how many genes do we have? BMC Biol. (2018) 16:94. doi: 10.1186/s12915-018-0564-x

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Bai Y, Sartor M, Cavalcoli J. Current status and future perspectives for sequencing livestock genomes. J Anim Sci Biotechnol. (2012) 3:8. doi: 10.1186/2049-1891-3-8

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Frank JA, Singh M, Cullen HB, Kirou RA, Benkaddour-Boumzaouad M, Cortes JL, et al. Evolution and antiviral activity of a human protein of retroviral origin. Science. (2022) 378:422–8. doi: 10.1126/science.abq7871

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Veglia AJ, Bistolas KSI, Voolstra CR, Hume BCC, Ruscheweyh H-J, Planes S, et al. Endogenous viral elements reveal associations between a non-retroviral RNA virus and symbiotic dinoflagellate genomes. Commun Bio. (2023) 6:566. doi: 10.1038/s42003-023-04917-9

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Kaján GL, Doszpoly A, Tarján ZL, Vidovszky MZ, Papp T. Virus-host coevolution with a focus on animal and human DNA viruses. J Mol Evol. (2020) 88:41–56. doi: 10.1007/s00239-019-09913-4

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Aiewsakun P, Katzourakis A. Endogenous viruses: connecting recent and ancient viral evolution. Virology. (2015) 479–80:26–37. doi: 10.1016/j.virol.2015.02.011

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Aliyari R, Ding SW. RNA-based viral immunity initiated by the Dicer family of host immune receptors. Immunol Rev. (2009) 227:176–88. doi: 10.1111/j.1600-065X.2008.00722.x

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Grandi N, Tramontano E. Human endogenous retroviruses are ancient acquired elements still shaping innate immune responses. Front Immunol. (2018) 9:2039. doi: 10.3389/fimmu.2018.02039

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Pastuzyn ED, Day CE, Kearns RB, Kyrke-Smith M, Taibi AV, McCormick J, et al. The neuronal gene arc encodes a repurposed retrotransposon Gag protein that mediates intercellular RNA transfer. Cell. (2018) 172:275–88.e18. doi: 10.1016/j.cell.2017.12.024

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Kedrov AV, Durymanov M, Anokhin KV. The Arc gene: retroviral heritage in cognitive functions. Neurosci Biobehav Rev. (2019) 99:275–81. doi: 10.1016/j.neubiorev.2019.02.006

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Zhou X, Singh M, Santos GS, Guerlavais V, Carvajal LA, Aivado M, et al. Pharmacologic activation of p53 triggers viral mimicry response thereby abolishing tumor immune evasion and promoting antitumor immunity. Cancer Discov. (2021) 11:3090–105. doi: 10.1158/2159-8290.CD-20-1741

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Ozaki T, Nakagawara A. Role of p53 in cell death and human cancers. Cancers. (2011) 3:994–1013. doi: 10.3390/cancers3010994

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Gravel A, Dubuc I, Morissette G, Sedlak RH, Jerome KR, Flamand L. Inherited chromosomally integrated human herpesvirus 6 as a predisposing risk factor for the development of angina pectoris. Proc Nat Acad Sci USA. (2015) 112:8058–63. doi: 10.1073/pnas.1502741112

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Casasnovas JM. Virus-receptor interactions and receptor-mediated virus entry into host cells. Subcell Biochem. (2013) 68:441–66. doi: 10.1007/978-94-007-6552-8_15

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Barrow E, Nicola AV, Liu J. Multiscale perspectives of virus entry via endocytosis. Virol J. (2013) 10:177. doi: 10.1186/1743-422X-10-177

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Harrison SC. Viral membrane fusion. Nat Struct Mol Biol. (2008) 15:690–8. doi: 10.1038/nsmb.1456

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Yamauchi Y, Greber UF. Principles of virus uncoating: cues and the snooker ball. Traffic. (2016) 17:569–92. doi: 10.1111/tra.12387

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Bostina M, Levy H, Filman DJ, Hogle JM. Poliovirus RNA is released from the capsid near a twofold symmetry axis. J Virol. (2011) 85:776–83. doi: 10.1128/JVI.00531-10

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Carty M, Guy C, Bowie AG. Detection of viral infections by innate immunity. Biochem Pharmacol. (2021) 183:114316. doi: 10.1016/j.bcp.2020.114316

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Rouse BT, Sehrawat S. Immunity and immunopathology to viruses: what decides the outcome? Nat Rev Immunol. (2010) 10:514–26. doi: 10.1038/nri2802

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Guidotti LG, Chisari FV. Noncytolytic control of viral infections by the innate and adaptive immune response. Annu Rev Immunol. (2001) 19:65–91. doi: 10.1146/annurev.immunol.19.1.65

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Mueller SN, Rouse BT. 27 - Immune responses to viruses. In: Rich RR, Fleisher TA, Shearer WT, Schroeder HW, Frew AJ, Weyand CM, editors. Clinical Immunology (Third Edition), Mosby, Vol. 1 (2008). p. 421–31. doi: 10.1016/B978-0-323-04404-2.10027-2

CrossRef Full Text | Google Scholar

53. Freihorst J, Ogra PL. Mucosal immunity and viral infections. Ann Med. (2001) 33:172–7. doi: 10.3109/07853890109002074

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Alcami A, Koszinowski UH. Viral mechanisms of immune evasion. Trends Microbiol. (2000) 8:410–8. doi: 10.1016/S0966-842X(00)01830-8

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Rai KR, Shrestha P, Yang B, Chen Y, Liu S, Maarouf M, et al. Acute infection of viral pathogens and their innate immune escape. Front Microbiol. (2021) 12:672026. doi: 10.3389/fmicb.2021.672026

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Zheng Y, Wu J, Ding C, Xu K, Yang S, Li L. Disease burden of chronic hepatitis B and complications in China from 2006 to 2050: an individual-based modeling study. Virol J. (2020) 17:132. doi: 10.1186/s12985-020-01393-z

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Streeck H, Maestri A, Habermann D, Crowell TA, Esber AL, Son G, et al. Dissecting drivers of immune activation in chronic HIV-1 infection. EBioMedicine. (2022) 83:104182. doi: 10.1016/j.ebiom.2022.104182

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Longdon B, Brockhurst MA, Russell CA, Welch JJ, Jiggins FM. The evolution and genetics of virus host shifts. PLoS Pathog. (2014) 10:e1004395. doi: 10.1371/journal.ppat.1004395

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Anishchenko M, Bowen RA, Paessler S, Austgen L, Greene IP, Weaver SC. Venezuelan encephalitis emergence mediated by a phylogenetically predicted viral mutation. Proc Natl Acad Sci USA. (2006) 103:4994–9. doi: 10.1073/pnas.0509961103

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Merani S, Petrovic D, James I, Chopra A, Cooper D, Freitas E, et al. Effect of immune pressure on hepatitis C virus evolution: insights from a single-source outbreak. Hepatology. (2011) 53:396–405. doi: 10.1002/hep.24076

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Castelló A, Alvarez E, Carrasco L. The multifaceted poliovirus 2A protease: regulation of gene expression by picornavirus proteases. J Biomed Biotechnol. (2011) 2011:369648. doi: 10.1155/2011/369648

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Kimura H, Kawada J, Ito Y. Epstein-Barr virus-associated lymphoid malignancies: the expanding spectrum of hematopoietic neoplasms. Nagoya J Med Sci. (2013) 75:169–79.

PubMed Abstract | Google Scholar

63. Walker AP, Fodor E. Interplay between influenza virus and the host RNA polymerase II transcriptional machinery. Trends Microbiol. (2019) 27:398–407. doi: 10.1016/j.tim.2018.12.013

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Priya SP, Sunil PM, Varma S, Brigi C, Isnadi MAR, Jayalal JA, et al. Direct, indirect, post-infection damages induced by coronavirus in the human body: an overview. Virus Dis. (2022) 33:429–44. doi: 10.1007/s13337-022-00793-9

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Kuypers FA. Hyperinflammation, apoptosis, and organ damage. Exp Biol Med. (2022) 247:1112–23. doi: 10.1177/15353702221090454

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Hojyo S, Uchida M, Tanaka K, Hasebe R, Tanaka Y, Murakami M, et al. How COVID-19 induces cytokine storm with high mortality. Inflamm Regen. (2020) 40:37. doi: 10.1186/s41232-020-00146-3

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Ramatillah DL, Gan SH, Pratiwy I, Sulaiman SAS, Jaber AAS, Jusnita N, et al. Impact of cytokine storm on severity of COVID-19 disease in a private hospital in West Jakarta prior to vaccination. PLoS ONE. (2022) 17:e0262438. doi: 10.1371/journal.pone.0262438

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Ryabkova VA, Churilov LP, Shoenfeld Y. Influenza infection, SARS, MERS and COVID-19: cytokine storm – The common denominator and the lessons to be learned. Clin Immunol. (2021) 223:108652. doi: 10.1016/j.clim.2020.108652

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Wang X, Ao Z, Chen L, Kobinger G, Peng J, Yao X. The cellular antiviral protein APOBEC3G interacts with HIV-1 reverse transcriptase and inhibits its function during viral replication. J Virol. (2012) 86:3777–86. doi: 10.1128/JVI.06594-11

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Meagher JL, Takata M, Gonçalves-Carneiro D, Keane SC, Rebendenne A, Ong H, et al. Structure of the zinc-finger antiviral protein in complex with RNA reveals a mechanism for selective targeting of CG-rich viral sequences. Proc Nat Acad Sci USA. (2019) 116:24303–9. doi: 10.1073/pnas.1913232116

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Yu CH, Bhattacharya A, Persaud M, Taylor AB, Wang Z, Bulnes-Ramos A, et al. Nucleic acid binding by SAMHD1 contributes to the antiretroviral activity and is enhanced by the GpsN modification. Nat Commun. (2021) 12:731. doi: 10.1038/s41467-021-21023-8

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Kristiansen H, Scherer Christina A, McVean M, Iadonato Shawn P, Vends S, Thavachelvam K, et al. Extracellular 2′-5′ oligoadenylate synthetase stimulates RNase L-independent antiviral activity: a novel mechanism of virus-induced innate immunity. J Virol. (2010) 84:11898–904. doi: 10.1128/JVI.01003-10

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Verhelst J, Hulpiau P, Saelens X. Mx proteins: antiviral gatekeepers that restrain the uninvited. Microbiol Mol Biol Rev. (2013) 77:551–66. doi: 10.1128/MMBR.00024-13

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Neil SJ. The antiviral activities of tetherin. Curr Top Microbiol Immunol. (2013) 371:67–104. doi: 10.1007/978-3-642-37765-5_3

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Momin T, Villasenor A, Singh A, Darweesh M, Singh A, Rajput M. ZFP36 ring finger protein like 1 significantly suppresses human coronavirus OC43 replication. PeerJ. (2023) 11:e14776. doi: 10.7717/peerj.14776

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Okada A, Iwatani Y. APOBEC3G-mediated G-to-A hypermutation of the HIV-1 genome: the missing link in antiviral molecular mechanisms. Front Microbiol. (2016) 7:2027. doi: 10.3389/fmicb.2016.02027

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Guo X, Carroll J-WN, Macdonald MR, Goff SP, Gao G. The zinc finger antiviral protein directly binds to specific viral mRNAs through the CCCH zinc finger motifs. J Virol. (2004) 78:12781–7. doi: 10.1128/JVI.78.23.12781-12787.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Zhang B, Goraya MU, Chen N, Xu L, Hong Y, Zhu M, et al. Zinc finger CCCH-type antiviral protein 1 restricts the viral replication by positively regulating type I interferon response. Front Microbiol. (2020) 11:1912. doi: 10.3389/fmicb.2020.01912

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Lahouassa H, Daddacha W, Hofmann H, Ayinde D, Logue EC, Dragin L, et al. SAMHD1 restricts the replication of human immunodeficiency virus type 1 by depleting the intracellular pool of deoxynucleoside triphosphates. Nat Immunol. (2012) 13:223–8. doi: 10.1038/ni.2236

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Chen S, Bonifati S, Qin Z, Gelais CS, Wu L. SAMHD1 suppression of antiviral immune responses. Trends Microbiol. (2019) 27:254–67. doi: 10.1016/j.tim.2018.09.009

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Silverman RH. Viral encounters with 2′,5′-oligoadenylate synthetase and RNase L during the interferon antiviral response. J Virol. (2007) 81:12720–9. doi: 10.1128/JVI.01471-07

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Kochs G, Janzen C, Hohenberg H, Haller O. Antivirally active MxA protein sequesters La Crosse virus nucleocapsid protein into perinuclear complexes. Proc Natl Acad Sci USA. (2002) 99:3153–8. doi: 10.1073/pnas.052430399

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Le Tortorec A, Willey S, Neil SJ. Antiviral inhibition of enveloped virus release by tetherin/BST-2: action and counteraction. Viruses. (2011) 3:520–40. doi: 10.3390/v3050520

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: host-virus interaction, virome, antiviral protein, endogenous viral elements, virus symbiosis

Citation: Rajput M and Thakur N (2023) Editorial: Advances in host-pathogen interactions for diseases in animals and birds. Front. Vet. Sci. 10:1282110. doi: 10.3389/fvets.2023.1282110

Received: 23 August 2023; Accepted: 30 August 2023;
Published: 11 September 2023.

Edited and reviewed by: Michael Kogut, United States Department of Agriculture, United States

Copyright © 2023 Rajput and Thakur. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Mrigendra Rajput, mrajput1@udayton.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.