#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Establishment of HSV1 Latency in Immunodeficient Mice Facilitates Efficient Reactivation


Although mouse models have been very useful in studies of HSV1 latency, the inability to efficiently reactivate latent HSV1 in vivo has impeded studies of reactivation. Reasoning that reactivation would be much more efficient in the absence of T cells, we exploited IVIG to promote survival of latently infected Rag mice lacking B and T cells. We established a threshold inoculum dose that was higher for B6- compared to 129-Rag mice, which determined whether HSV1 could be efficiently reactivated in vivo resulting in encephalitis. We showed directly that memory T cells are required to control spontaneous and induced reactivation in mice inoculated at high dose but are dispensable for maintaining latency in low dose inoculated mice. Incorporating different knockout strains into the Rag latency model by adoptive transfer of cells or crossbreeding will facilitate studying the role of various cellular genes involved in regulating neuronal gene expression and innate and adaptive immunity in the control of HSV1 reactivation. The potential of this powerful latency model to unravel the molecular and immune mechanisms regulating latency will be realized only after it is adopted and refined by researchers in the field.


Vyšlo v časopise: Establishment of HSV1 Latency in Immunodeficient Mice Facilitates Efficient Reactivation. PLoS Pathog 11(3): e32767. doi:10.1371/journal.ppat.1004730
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004730

Souhrn

Although mouse models have been very useful in studies of HSV1 latency, the inability to efficiently reactivate latent HSV1 in vivo has impeded studies of reactivation. Reasoning that reactivation would be much more efficient in the absence of T cells, we exploited IVIG to promote survival of latently infected Rag mice lacking B and T cells. We established a threshold inoculum dose that was higher for B6- compared to 129-Rag mice, which determined whether HSV1 could be efficiently reactivated in vivo resulting in encephalitis. We showed directly that memory T cells are required to control spontaneous and induced reactivation in mice inoculated at high dose but are dispensable for maintaining latency in low dose inoculated mice. Incorporating different knockout strains into the Rag latency model by adoptive transfer of cells or crossbreeding will facilitate studying the role of various cellular genes involved in regulating neuronal gene expression and innate and adaptive immunity in the control of HSV1 reactivation. The potential of this powerful latency model to unravel the molecular and immune mechanisms regulating latency will be realized only after it is adopted and refined by researchers in the field.


Zdroje

1. Yao H-W, Ling P, Chen S-H, Tung Y-Y, Chen S-H. Factors affecting herpes simplex virus reactivation from the explanted mouse brain. Virology 2012; 433: 116–123. doi: 10.1016/j.virol.2012.07.018 22884293

2. Nicoll MP, Proença JT, Efstathiou S The molecular basis of herpes simplex virus latency. FEMS Microbiology Reviews 2012; 36: 684–705. doi: 10.1111/j.1574-6976.2011.00320.x 22150699

3. Ramakrishna C, Openshaw H, Cantin EM. The case for immunomodulatory approaches in treating HSV encephalitis. Future Virology 2013; 8: 259–272. 23956785

4. Whitley RJ. Herpes simplex encephalitis: Adolescents and adults. Antiviral Research 2006; 71: 141–148. 16675036

5. Tyler KL. Herpes simplex virus infections of the central nervous system: encephalitis and meningitis, including Mollaret's. Herpes 2004; 11 Suppl 2: 57A–64A. 15319091

6. Bernstein DI, Bellamy AR, Hook EW, Levin MJ, Wald A, et al. Epidemiology, Clinical Presentation, and Antibody Response to Primary Infection With Herpes Simplex Virus Type 1 and Type 2 in Young Women. Clinical Infectious Diseases 2013; 56: 344–351. doi: 10.1093/cid/cis891 23087395

7. Fatahzadeh M, Schwartz RA. Human herpes simplex virus infections: Epidemiology, pathogenesis, symptomatology, diagnosis, and management. Journal of the American Academy of Dermatology 2007; 57: 737–763. 17939933

8. Azwa A, Barton SE. Aspects of herpes simplex virus: a clinical review. J Fam Plann Reprod Health Care 2009; 35: 237–242. doi: 10.1783/147118909789587376 19849918

9. Stranska R, Schuurman R, Nienhuis E, Goedegebuure IW, Polman M, et al. Survey of acyclovir-resistant herpes simplex virus in the Netherlands: prevalence and characterization. J Clin Virol 2005; 32: 7–18. 15572000

10. van der Beek MT, Laheij AM, Raber-Durlacher JE, von dem Borne PA, Wolterbeek R, et al. Viral loads and antiviral resistance of herpesviruses and oral ulcerations in hematopoietic stem cell transplant recipients. Bone Marrow Transplant 2012; 47: 1222–1228. doi: 10.1038/bmt.2012.2 22327137

11. Frobert E, Burrel S, Ducastelle-Lepretre S, Billaud G, Ader F, et al. Resistance of herpes simplex viruses to acyclovir: An update from a ten-year survey in France. Antiviral Research 2014; 111: 36–41. doi: 10.1016/j.antiviral.2014.08.013 25218782

12. Duan R, de Vries RD, van Dun JM, van Loenen FB, Osterhaus ADME, et al. Acyclovir Susceptibility and Genetic Characteristics of Sequential Herpes Simplex Virus Type 1 Corneal Isolates from Patients with Recurrent Herpetic Keratitis. Journal of Infectious Diseases 2009; 200: 1402–1414. doi: 10.1086/606028 19795980

13. van Velzen M, van de Vijver DAMC, van Loenen FB, Osterhaus ADME, Remeijer L, et al. Acyclovir Prophylaxis Predisposes to Antiviral-Resistant Recurrent Herpetic Keratitis. Journal of Infectious Diseases 2013; 208: 1359–1365. doi: 10.1093/infdis/jit350 23901090

14. Cantin E, Tanamachi B, Openshaw H. Role for gamma interferon in control of herpes simplex virus type 1 reactivation. J Virol 1999; 73: 3418–3423. 10074196

15. Sawtell NM, Thompson RL. Rapid in vivo reactivation of herpes simplex virus in latently infected murine ganglionic neurons after transient hyperthermia. J Virol 1992; 66: 2150–2156. 1312625

16. Liu T, Tang Q, Hendricks RL. Inflammatory infiltration of the trigeminal ganglion after herpes simplex virus type 1 corneal infection. J Virol 1996; 70: 264–271. 8523535

17. van Lint AL, Kleinert L, Clarke SRM, Stock A, Heath WR, et al. Latent Infection with Herpes Simplex Virus Is Associated with Ongoing CD8+ T-Cell Stimulation by Parenchymal Cells within Sensory Ganglia. J Virol 2005; 79: 14843–14851. 16282484

18. Lekstrom-Himes JA, LeBlanc RA, Pesnicak L, Godleski M, Straus SE. Gamma interferon impedes the establishment of herpes simplex virus type 1 latent infection but has No impact on its maintenance or reactivation in mice [In Process Citation]. J Virol 2000; 74: 6680–6683. 10864685

19. Tan IL, McArthur JC, Venkatesan A, Nath A. Atypical manifestations and poor outcome of herpes simplex encephalitis in the immunocompromised. Neurology 2012; 79: 2125–2132. doi: 10.1212/WNL.0b013e3182752ceb 23136265

20. Webre JM, Hill JM, Nolan NM, Clement C, McFerrin HE, et al.Rabbit and Mouse Models of HSV-1 Latency, Reactivation, and Recurrent Eye Diseases. Journal of Biomedicine and Biotechnology 2012; 2012: 18.

21. Schiff D, Rosenblum MK. Herpes simplex encephalitis (HSE) and the immunocompromised: a clinical and autopsy study of HSE in the settings of cancer and human immunodeficiency virus-type 1 infection [see comments]. Human Pathology 1998; 29: 215–222. 9496822

22. Lundberg P, Ramakrishna C, Brown J, Tyszka JM, Hamamura M, et al. The Immune Response to Herpes Simplex Virus Type 1 Infection in Susceptible Mice is a Major Cause of CNS Pathology Resulting in Fatal Encephalitis. J Virol 2008; 82: 7078–7088. doi: 10.1128/JVI.00619-08 18480436

23. Jordan SC, Toyoda M, Vo AA. Intravenous Immunoglobulin a Natural Regulator of Immunity and Inflammation. Transplantation 2009; 88: 1–6. doi: 10.1097/TP.0b013e3181a9e89a 19584672

24. Imbach P, Lazarus AH, Khne T. Intravenous immunoglobulins induce potentially synergistic immunomodulations in autoimmune disorders. Vox Sanguinis 2010; 98: 385–394. doi: 10.1111/j.1423-0410.2009.01264.x 19821958

25. Ferrara G, Zumla A, Maeurer M. Intravenous Immunoglobulin (IVIg) for Refractory and Difficult-to-treat Infections. The American Journal of Medicine 2012; 125: 1036.e1031–1036.e1038. doi: 10.1016/j.amjmed.2012.01.023 22608788

26. Keller MA, Stiehm ER. Passive immunity in prevention and treatment of infectious diseases. Clin Microbiol Rev 2000; 13: 602–614. 11023960

27. Ramakrishna C, Newo ANS, Shen Y- W, Cantin E. Passively Administered Pooled Human Immunoglobulins Exert IL-10 Dependent Anti-Inflammatory Effects that Protect against Fatal HSV Encephalitis. PLoS Pathog 2011; 7: e1002071. doi: 10.1371/journal.ppat.1002071 21655109

28. Zerboni L, Che X, Reichelt M, Qiao Y, Gu H, et al. Herpes Simplex Virus 1 Tropism for Human Sensory Ganglion Neurons in the Severe Combined Immunodeficiency Mouse Model of Neuropathogenesis. Journal of Virology 2013; 87: 2791–2802. doi: 10.1128/JVI.01375-12 23269807

29. Lundberg P, Openshaw H, Wang M, Yang HJ, Cantin E. Effects of CXCR3 Signaling on Development of Fatal Encephalitis and Corneal and Periocular Skin Disease in HSV-Infected Mice Are Mouse-Strain Dependent. Invest Ophthalmol Vis Sci 2007; 48: 4162–4170. 17724202

30. Yao H-W, Ling P, Tung Y-Y, Hsu S-M, Chen S-H. In Vivo Reactivation of Latent Herpes Simplex Virus 1 in Mice Can Occur in the Brain before in the Trigeminal Ganglion. Journal of Virology. 2014; 88(19):11264–70 doi: 10.1128/JVI.01616-14 25031345

31. Feldman LT, Ellison AR, Voytek CC, Yang L, Krause P, et al. Spontaneous molecular reactivation of herpes simplex virus type 1 latency in mice. PNAS. 2002; 99(2):978–83 11773630

32. Ma JZ, Russell TA, Spelman T, Carbone FR, Tscharke DC. Lytic Gene Expression Is Frequent in HSV-1 Latent Infection and Correlates with the Engagement of a Cell-Intrinsic Transcriptional Response. PLoS Pathog 2014; 10: e1004237. doi: 10.1371/journal.ppat.1004237 25058429

33. Margolis TP, Elfman FL, Leib D, Pakpour N, Apakupakul K, et al. Spontaneous Reactivation of Herpes Simplex Virus Type 1 in Latently Infected Murine Sensory Ganglia. J Virol 2007; 81: 11069–11074. 17686862

34. Krawczyk A, Arndt MAE, Grosse-Hovest L, Weichert W, Giebel B, et al. Overcoming drug-resistant herpes simplex virus (HSV) infection by a humanized antibody. Proceedings of the National Academy of Sciences. 2013; 110(17):6760–5 doi: 10.1073/pnas.1220019110 23569258

35. Koyuncu OO, Hogue IB, Enquist LW. Virus Infections in the Nervous System. Cell Host & Microbe 2013; 13: 379–393.

36. Frenzel K, Ganepola S, Michel D, Thiel E, Krüger D, et al. Antiviral function and efficacy of polyvalent immunoglobulin products against CMV isolates in different human cell lines. Medical Microbiology and Immunology. 2012; 201(3):277–86 doi: 10.1007/s00430-012-0229-2 22297735

37. Pavan PR, Ennis FA. The Elimination of Herpes Simplex Plaques by Antibody and the Emergence of Resistant Strains. The Journal of Immunology 1977; 118: 2167–2175. 193992

38. Oakes JE, Lausch RN. Monoclonal antibodies suppress replication of herpes simplex virus type 1 in trigeminal ganglia. J Virol 1984; 51: 656–661. 6088790

39. Sawtell NM. Comprehensive quantification of herpes simplex virus latency at the single-cell level. J Virol 1997; 71: 5423–5431. 9188614

40. Sawtell NM. The Probability of In Vivo Reactivation of Herpes Simplex Virus Type 1 Increases with the Number of Latently Infected Neurons in the Ganglia. Journal of Virology 1998; 72: 6888–6892. 9658140

41. Hoshino Y, Qin J, Follmann D, Cohen JI, Straus SE. The number of herpes simplex virus-infected neurons and the number of viral genome copies per neuron correlate with the latent viral load in ganglia. Virology 2008; 372: 56–63. 18045638

42. Ellison AR, Yang L, Voytek C, Margolis TP. Establishment of latent herpes simplex virus type 1 infection in resistant, sensitive, and immunodeficient mouse strains. Virology 2000; 268: 17–28. 10683323

43. Chakraborty S, Nazmi A, Dutta K, Basu A. Neurons under viral attack: Victims or warriors? Neurochemistry International 2010; 56: 727–735. doi: 10.1016/j.neuint.2010.02.016 20206655

44. Sahin U, Ferhi O, Jeanne M, Benhenda S, Berthier C, et al. Oxidative stress–induced assembly of PML nuclear bodies controls sumoylation of partner proteins. The Journal of Cell Biology 2014; 204: 931–945. doi: 10.1083/jcb.201305148 24637324

45. Cho H, Proll SC, Szretter KJ, Katze MG, Gale M, et al. Differential innate immune response programs in neuronal subtypes determine susceptibility to infection in the brain by positive-stranded RNA viruses. Nature Medicine. 2013; 19(4):458–64 doi: 10.1038/nm.3108 23455712

46. Margolis TP, Imai Y, Yang L, Vallas V, Krause PR. Herpes Simplex Virus Type 2 (HSV-2) Establishes Latent Infection in a Different Population of Ganglionic Neurons than HSV-1: Role of Latency-Associated Transcripts. J Virol 2007; 81: 1872–1878. 17151134

47. Bertke AS, Ma A, Margolis MS, Margolis TP. Different Mechanisms Regulate Productive Herpes Simplex Virus 1 (HSV-1) and HSV-2 Infections in Adult Trigeminal Neurons. Journal of Virology 2013; 87: 6512–6516. doi: 10.1128/JVI.00383-13 23514893

48. Boutell C, Everett RD. Regulation of alphaherpesvirus infections by the ICP0 family of proteins. Journal of General Virology 2013; 94: 465–481. doi: 10.1099/vir.0.048900-0 23239572

49. Randow F, MacMicking JD, James LC. Cellular Self-Defense: How Cell-Autonomous Immunity Protects Against Pathogens. Science 2013; 340: 701–706. doi: 10.1126/science.1233028 23661752

50. Lafaille FG, Pessach IM, Zhang S-Y, Ciancanelli MJ, Herman M, et al. Impaired intrinsic immunity to HSV-1 in human iPSC-derived TLR3-deficient CNS cells. Nature 2012; 491: 769–773. doi: 10.1038/nature11583 23103873

51. van Lint A, Ayers M, Brooks AG, Coles RM, Heath WR, et al. Herpes Simplex Virus-Specific CD8+ T Cells Can Clear Established Lytic Infections from Skin and Nerves and Can Partially Limit the Early Spread of Virus after Cutaneous Inoculation. J Immunol 2004; 172: 392–397. 14688347

52. Lang A, Nikolich-Zugich J. Development and Migration of Protective CD8+ T Cells into the Nervous System following Ocular Herpes Simplex Virus-1 Infection. J Immunol 2005; 174: 2919–2925. 15728503

53. Orr MT, Mathis MA, Lagunoff M, Sacks JA, Wilson CB. CD8 T Cell Control of HSV Reactivation from Latency Is Abrogated by Viral Inhibition of MHC Class I. Cell Host & Microbe 2007; 2: 172–180.

54. Jugovic P, Hill AM, Tomazin R, Ploegh H, Johnson DC. Inhibition of major histocompatibility complex class I antigen presentation in pig and primate cells by herpes simplex virus type 1 and 2 ICP47. J Virol 1998; 72: 5076–5084. 9573278

55. Knickelbein JE, Khanna KM, Yee MB, Baty CJ, Kinchington PR, et al. Noncytotoxic Lytic Granule-Mediated CD8+ T Cell Inhibition of HSV-1 Reactivation from Neuronal Latency. Science 2008; 322: 268–271. doi: 10.1126/science.1164164 18845757

56. Held K, Derfuss T. Control of HSV-1 latency in human trigeminal ganglia—current overview. Journal of Neurovirology 2011; 17: 518–527. doi: 10.1007/s13365-011-0063-0 22139603

57. St. Leger A, Hendricks R. CD8+ T cells patrol HSV-1-infected trigeminal ganglia and prevent viral reactivation. Journal of Neurovirology 2011; 17: 528–534. doi: 10.1007/s13365-011-0062-1 22161682

58. Himmelein S, St Leger A, Knickelbein J, Rowe A, Freeman M, et al. Circulating herpes simplex type 1 (HSV-1)-specific CD8+ T cells do not access HSV-1 latently infected trigeminal ganglia. Herpesviridae 2011; 2: 5. doi: 10.1186/2042-4280-2-5 21429183

59. Gebhardt T, Wakim LM, Eidsmo L, Reading PC, Heath WR, et al. Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus. Nat Immunol 2009; 10: 524–530. doi: 10.1038/ni.1718 19305395

60. Wakim LM, Woodward-Davis A, Bevan MJ. Memory T cells persisting within the brain after local infection show functional adaptations to their tissue of residence. Proc Natl Acad Sci U S A 2010; 107: 17872–17879. doi: 10.1073/pnas.1010201107 20923878

61. Egan K, Wu S, Wigdahl B, Jennings S. Immunological control of herpes simplex virus infections. Journal of NeuroVirology 2013; 19: 328–345. doi: 10.1007/s13365-013-0189-3 23943467

62. Cantin E, Tanamachi B, Openshaw H, Mann J, Clarke K. Gamma interferon (IFN-gamma) receptor null-mutant mice are more susceptible to herpes simplex virus type 1 infection than IFN-gamma ligand null-mutant mice. J Virol 1999; 73: 5196–5200. 10233988

63. Stock AT, Jones CM, Heath WR, Carbone FR. Rapid recruitment and activation of CD8+ T cells after herpes simplex virus type 1 skin infection. Immunol Cell Biol 2011; 89: 143–148. doi: 10.1038/icb.2010.66 20458339

64. F Catez dr, Picard C, Held K, Gross S, Rousseau A, et al. HSV-1 Genome Subnuclear Positioning and Associations with Host-Cell PML-NBs and Centromeres Regulate LAT Locus Transcription during Latency in Neurons. PLoS Pathogens 2012; 8: e1002852. doi: 10.1371/journal.ppat.1002852 22912575

65. Catez F ric, Rousseau A, Labetoulle M, Lomonte P. Detection of the Genome and Transcripts of a Persistent DNA Virus in Neuronal Tissues by Fluorescent In situ Hybridization Combined with Immunostaining. JoVE. 2014; (83): e51091. doi: 10.3791/51091 24514006

66. Lachmann RH, Brown C, Efstathiou S. A murine RNA polymerase I promoter inserted into the herpes simplex virus type 1 genome is functional during lytic, but not latent, infection. J Gen Virol 1996; 77 (Pt 10): 2575–2582. 8887493

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2015 Číslo 3
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#