Apoptosis- and survival-related gene mRNA profile in peripheral blood leukocytes in children with acute EBV infectious mononucleosis

Cover Page


Cite item

Full Text

Abstract

Acute EBV-associated mononucleosis develops mainly in children and in patients with functionally impaired immune system. Consequently, it may result in developing secondary immunodeficiency, neoplasms as well as diverse alterations in cell-mediated immune reaction. Despite extensively examining molecular mechanisms of EBV infection, it is also necessary seek for new molecular and genetic factors underlying pathogenesis of EBV-mediated mononucleosis and EBV-associated malignant cell transformation is necessary, which might be used in clinical practice to monitor clinical score as well as predictive parameters for EBV-associated complications such as immunocompromised conditions and neoplasms. Here, we proposed to use our splicing sensitive DNA microarrays to perform a comprehensive semi-quantitative mRNA expression analysis for major apoptosis- and survival-related signaling components in peripheral blood leukocytes collected from children with acute EBV infectious mononucleosis as well as during recovery period. Using such DNA microchips allowed to assess both total (denoted by Σ) and separate transcript expression resulting from alternative splicing. It was shown that the balance of mRNA levels in acute phase of EBV-infectious mononucleosis was shifted towards upregulated expression of anti-apoptotic factors and components of of NF-κB-linked pro-survival signaling able to profoundly augment apoptosis resistance. Moreover, some EBV-associated changes (BIM/BCL2L11-Σ, PUMA/ BBC3-NM_001127241, BID-Σ, CASP3-Σ, NFKB1-Σ, RELA-Σ) were in agreement with the data published before. In addition, we also found previously unknown changes in level of EBV-associated coding and noncoding transcripts (DCR1/ TNFRSF10C-NM_003841, DR5/TNFRSF10B-NR_027140, CASP6 beta/CASP6-NM_032992, CASP7-NM_033338). Analyzing their properties allowed to suggest that they play an important role in the pathogenesis of EBV-associated mononucleosis. However, at asymptomatic recovery stage, level of some mRNA expression was kept altered compared to healthy volunteers (DCR2/TNFRSF10D-NM_003840, CASP8-Σ, CASP3-Σ, BIM/BCL2L11-Σ, BCL2-NM_000633, MCL1-Σ, BCL-W/BCL2L2-Σ, BCL-XL/BCL2L1-NM_138578, BIRC2-NM_001166, XIAP-NM_001167, TRAF2-NM_021138, MAP3K14-Σ, NFKB1-Σ), which may point at postponed EBV-associated molecular consequences. On one hand, such changes may be due to long-lasting anti-EBV immune response, whereas, on the other hand, they might be influenced by EBV-associated factors facilitating virus persistence. Overall, we identified the molecular features predisposing to chronic course of EBV-infection. The data obtained further expand our understanding about the molecular pathogenetic mechanisms for EBV infectious mononucleosis.

About the authors

N. A. Sakharnov

Blokhina Scientific Research Institute of Epidemiology and Microbiology of Nizhny Novgorod

Author for correspondence.
Email: saharnov@nniiem.ru
http://www.nniiem.ru

Nikolai A. Sakharnov, Researcher, Laboratory of Molecular Biology and Biotechnology

603950, Nizhny Novgorod, Malaya Yamskaya str., 71

Phone: +7 (831) 469-79-46 (office); +7 950 624-87-12 (mobile). Fax: +7 (831) 469-79-20.

Russian Federation

O. V. Utkin

Blokhina Scientific Research Institute of Epidemiology and Microbiology of Nizhny Novgorod

Email: utkino2004@mail.ru

PhD (Biology), Head of the Laboratory of Molecular Biology and Biotechnology

Nizhny Novgorod Russian Federation

E. N. Filatova

Blokhina Scientific Research Institute of Epidemiology and Microbiology of Nizhny Novgorod

Email: filatova@nniiem.ru

PhD (Biology), Leading Researcher, Laboratory of Molecular Biology and Biotechnology

Nizhny Novgorod

Russian Federation

D. I. Knyazev

Blokhina Scientific Research Institute of Epidemiology and Microbiology of Nizhny Novgorod

Email: Dmitry-Kn@yandex.ru

PhD (Biology), Senior Researcher, Laboratory of Molecular Biology and Biotechnology

Nizhny Novgorod Russian Federation

N. B. Presnyakova

Blokhina Scientific Research Institute of Epidemiology and Microbiology of Nizhny Novgorod

Email: presnyakova_nb@mail.ru

Researcher, Laboratory of Molecular Biology and Biotechnology

Nizhny Novgorod Russian Federation

References

  1. Кудин А.П., Романовская Т.Р., Белевцев М.В. Состояние специфического иммунитета при инфекционном мононуклеозе у детей // Медицинский журнал. 2007. Т. 1, № 19. С. 102–106.
  2. Кускова Т.К., Белова Е.Г. Семейство герпесвирусов на современном этапе // Лечащий врач. 2004. Т. 5. С. 64–69.
  3. Уткин О.В., Новиков В.В. Регуляция апоптоза с помощью альтернативного сплайсинга матричной РНК // Российский биотерапевтический журнал. 2007. Т. 6, № 2. C. 13–20.
  4. Уткин О.В., Новиков В.В. Рецепторы смерти в модуляции апоптоза // Успехи современной биологии. 2012. Т. 132, № 4. С. 381–390.
  5. Филатова Е.Н., Уткин О.В. Роль некодирующих изоформ мРНК белок-кодирующих генов в регуляции генной экспрессии // Генетика. 2018. Т. 54, № 8. С. 879–887. doi: 10.1134/S0016675818080052
  6. Филатова Е.Н., Уткин О.В. Современные подходы к моделированию герпесвирусной инфекции // Журнал МедиАль. 2014. Т. 2, № 12. С. 172–197.
  7. Anderton E., Yee J., Smith P., Crook T., White R.E., Allday M.J. Two Epstein–Barr virus (EBV) oncoproteins cooperate to repress expression of the proapoptotic tumor-suppressor Bim: clues to the pathogenesis of Burkitt’s lymphoma. Oncogene, 2008, vol. 27, no. 4, pp. 421–433. doi: 10.1038/sj.onc.1210668
  8. Barblu L., Smith N., Durand S., Scott-Algara D., Boufassa F., Delfraissy J.F., Cimarelli A., Lamwbotte O., Herbeuval J.P. Reduction of death receptor 5 expression and apoptosis of CD4+ T cells from HIV controllers. Clin. Immunol., 2014, vol. 155, no. 1, pp. 17–26. doi: 10.1016/j.clim.2014.07.010
  9. Carmilleri-Broet B.S., Davi F., Feuillard J., Bourgeois C., Seilhean D., Hauw J.J., Rapha l M. High expression of latent membrane protein 1 of Epstein–Barr virus and BCL-2 oncoprotein in acquired immunodeficiency syndrome-related primary brain lymphomas. Blood, 1995, vol. 86, no. 2, pp. 432–435
  10. Chang M.S., Kim D.H., Roh J.K., Middeldorp J.M., Kim Y.S., Kim S., Han S., Kim C.W., Lee B.L., Kim W.H., Woo J.H. Epstein–Barr virus-encoded BARF1 promotes proliferation of gastric carcinoma cells through regulation of NF-κB. J. Virol., 2013, vol. 87, no. 19, pp. 10515–10523. doi: 10.1128/JVI.00955-13
  11. Chanut A., Duguet F., Marfak A., David A., Petit B., Parrens M., Durand-Panteix S., Boulin-Deveza M., Gachard N., YoulyouzMarfak I., Bordessoule D., Feuillard J., Faumont N. RelA and RelB cross-talk and function in Epstein–Barr virus transformed B cells. Leukemia, 2014, vol. 28, no. 4, pp. 871–879. doi: 10.1038/leu.2013.274
  12. Choy E.Y., Siu K.L., Kok K.H., Lung R.W., Tsang C.M., To K.F., Kwong D.L., Tsao S.W., Jin D.Y. An Epstein–Barr virus-encoded microRNA targets PUMA to promote host cell survival. J. Exp. Med., 2008, vol. 205, no. 11, pp. 2551–2560. doi: 10.1084/jem.20072581
  13. Cohen J.I., Fauci A.S., Varmus H., Nabel G.J., Epstein–Barr virus: an important vaccine target for cancer prevention. Sci. Transl. Med., 2011, vol. 3: 107fs7. doi: 10.1126/scitranslmed.3002878
  14. Collison A., Foster P.S., Mattes J. Emerging role of tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) as a key regulator of inflammatory responses. Clin. Exp. Pharmacol. Physiol., 2009, vol. 36, no. 11, pp. 1049–1053. doi: 10.1111/j.1440-1681.2009.05258.x
  15. Devergne O., Hatzivassiliou E., Izumi K.M., Kaye K.M., Kleijnen M.F., Kieff E., Mosialos G. TRAF1, TRAF2 and TRAF3 effect NF-kB activation by an Epstein–Barr Virus LMP1 domain important for B lymphocyte transformation. Mol. Cell Biol., 1996, vol. 16, pp. 7098–7107. doi: 10.1128/MCB.16.12.7098
  16. Devergne O., McFarland E.C., Mosialos G., Izumi K.M., Ware C.F., Kieff E. Role of the TRAF binding site and NF-kB activation in Epstein–Barr virus latent membrane protein 1-induced cell gene expression. J. Virol., 1998, vol. 72, pp. 7900–7908.
  17. Dojcinov S.D., Fend F., Quintanilla-Martinez L. EBV-positive lymphoproliferations of B-, Tand NK-cell derivation in nonimmunocompromised hosts. Pathogens, 2018, vol. 7, p. 28. doi: 10.3390/pathogens7010028
  18. Du J., Liang X., Liu Y., Qu Z., Gao L., Han L., Liu S., Cui M., Shi Y., Zhang Z., Yu L., Cao L., Ma C., Zhang L., Chen Y., Sun W. Hepatitis B virus core protein inhibits TRAIL-induced apoptosis of hepatocytes by blocking DR5 expression. Cell Death Differ., 2009, vol. 16, no. 2, pp. 219–229. doi: 10.1038/cdd.2008.144
  19. Eliopoulos A.G., Stack M., Dawson C.W., Kaye K.M., Hodgkin L., Sinota S., Rowe M., Young L.S. Epstein–Barr virus-encoded LMP1 and CD40 mediate IL-6 production in epithelial cells via an NF-kB pathway involving TNF receptor associated factors. Oncogene, 1997, vol. 14, no. 24, pp. 2899–2916. doi: 10.1038/sj.onc.1201258
  20. Eliopoulos A.G., Young L.S. LMP1 structure and signal transduction. Semin. Cancer Biol., 2001, vol. 11, pp. 435–444. doi: 10.1006/scbi.2001.0410
  21. Floettmann J.E., Rowe M. Epstein–Barr virus latent membrane protein-1 (LMP1) C-terminus activation region 2 (CTAR2) maps to the far C-terminus and requires oligomerisation for NF-κB activation. Oncogene, 1997, vol. 15, pp. 1851–1858. doi: 10.1038/sj.onc.1201359
  22. Fu Q., He C., Mao Z.R. Epstein–Barr virus interactions with the Bcl-2 protein family and apoptosis in human tumor cells. Journal of Zhejiang University. Science B. Biomedicine and Biotechnology, 2013, vol. 14, no. 1, pp. 8–24. doi: 10.1631/jzus.B1200189
  23. Gires O., Zimber-Strobl U., Gonnella R., Ueffing M., Marschall G., Zeidler R., Pich D., Hammerschmidt W. Latent membrane protein 1 of Epstein–Barr virus mimics a constitutively active receptor molecule. EMBO J., 1997, vol. 16, pp. 6131–6140. doi: 10.1093/emboj/16.20.6131
  24. Harold C., Cox D., Riley K.J. Epstein–Barr viral microRNAs target caspase 3. Virol. J., 2016, vol. 13: v145. doi: 10.1186/s12985-016-0602-7
  25. Hayward S.D. Viral interactions with the Notch pathway. Semin. Cancer Biol., 2004, vol. 14, no. 5, pp. 387–396. doi: 10.1016/j.semcancer.2004.04.018
  26. Hjalgrim H., Askling J., Sørensen P., Madsen M., Rosdahl N., Storm H.H., Hamilton-Dutoit S., Eriksen L.S., Frisch M., Ekbom A., Melbye M. Risk of Hodgkin’s disease and other cancers after infectious mononucleosis. J. Natl. Cancer Inst., 2000, vol. 92, no. 18, pp. 1522–1528
  27. Irmler M., Thome M., Hahne M., Schneider P., Hofmann K., Steiner V., Bodmer J.L., Schroter M., Burns K., Mattmann C., Rimoldi D., French L.E., Tschopp J. Inhibition of death receptor signals by cellular FLIP. Nature, 1997, vol. 388, no. 6638, pp. 190–195. doi: 10.1038/40657
  28. Iyori M., Zhang T., Pantel H., Gagne B.A., Sentman C.L. TRAIL/DR5 plays a critical role in NK cell-mediated negative regulation of dendritic cell cross-priming of T cells. J. Immunol., 2011, vol. 187, no. 6, pp. 3087–3095. doi: 10.4049/jimmunol.1003879
  29. Kaye K.M., Devergne O., Harada J.N., Izumi K.M., Yalamanchili R., Kieff E., Mosialos G. Tumour necrosis factor receptor associated factor 2 is a mediator of NF-kB activation by latent infection membrane protein 1, the Epstein–Barr virus transforming protein. Proc. Natl. Acad. Sci. USA, 1996, vol. 93, pp. 11085–11090
  30. Knyazev D.I., Starikova V.D., Utkin О.V., Solntsev L.A., Sakharnov N.A., Efimov E.I. Splicing-sensitive DNA-microarrays: peculiarities and application in biomedical research. CTM, 2015, vol. 7, no. 4. pp. 162–172. doi: 10.17691/stm2015.7.4.23
  31. Kohlhof H., Hampel F., Hoffmann R., Burtscher H., Weidle U.H., Holzel M., Eick D., Zimber-Strobl U., Strobl L. J. Notch1, Notch 2 and Epstein–Barr virus-encoded nuclear antigen 2 signaling differentially affects proliferation and survival of Epstein– Barr virus-infected B cells. Blood, 2009, vol. 113, no. 22, pp. 5506–5515. doi: 10.1182/blood-2008-11-190090
  32. Lantner F., Starlets D., Gore Y., Flaishon L., Yamit-Hezi A., Dikstein R., Leng L., Bucala R., Machluf Y., Oren M., Shachar I. CD74 induces TAp63 expression leading to B-cell survival. Blood, 2007, vol. 110, no. 13, pp. 4303–4311. doi: 10.1182/blood-2007-04-087486
  33. Lee A.W., Champagne N., Wang X., Su X.D., Goodyer C., Leblanc A.C. Alternatively spliced caspase-6B isoform inhibits the activation of caspase-6A. J. Biol. Chem., 2010, vol. 285, no. 42, pp. 31974–31984. doi: 10.1074/jbc.M110.152744
  34. Lee Y., Rio D.C. Mechanisms and regulation of alternative pre-mRNA splicing. Annu. Rev. Biochem., 2015, vol. 84, pp. 291–323. doi: 10.1146/annurev-biochem-060614-034316
  35. Luftig M., Yasui T., Soni V., Kang M.S., Jacobson N., Cahir-McFarland E., Seed B., Kieff E. Epstein–Barr virus latent infection membrane protein 1 TRAF-binding site induces NIK/IKK alpha-dependent noncanonical NF-kappaB activation. Proc. Natl. Acad. Sci. USA, 2004, vol. 101, no. 1. pp. 141–146. doi: 10.1073/pnas.2237183100
  36. McCarthy D. J., Smyth G. K. Testing significance relative to a fold-change threshold is a TREAT. Bioinformatics, 2009, vol. 25, no. 6, pp. 765–771. doi: 10.1093/bioinformatics/btp053
  37. Nandakumar A., Uwatoko F., Yamamoto M., Tomita K., Majima H.J., Akiba S., Koriyama C. Radiation-induced Epstein– Barr virus reactivation in gastric cancer cells with latent EBV infection. Tumor Biol., 2017, vol. 39, no. 7: 1010428317717718. doi: 10.1177/1010428317717718
  38. Paschos K., Smith P., Anderton E. Middeldorp J.M., White R.E., Allday M.J. Epstein–Barr virus latency in B cells leads to epigenetic repression and CpG methylation of the tumor suppressor gene bim. PLoS Pathog., 2009, vol. 5, no. 6: 1000492. doi: 10.1371/journal.ppat.1000492
  39. Portis T., Longnecker R. Epstein–Barr virus (EBV) LMP2A mediates B-lymphocyte survival through constitutive activation of the Ras/PI3K/AKT pathway. Oncogene, 2004, vol. 23, no. 53, pp. 8619–8628. doi: 10.1038/sj.onc.1207905
  40. Pratt Z.L., Zhang J., Sugden B. Simultaneously induce and inhibit oncogene of Epstein–Barr virus can the latent membrane protein 1 (LMP1) apoptosis in B cells. J. Virol., 2012, vol. 86, no. 8, pp. 4380–4393. doi: 10.1128/JVI.06966-11
  41. Schneider F., Neugebauer J., Griese J., Liefold N., Kutz H., Brise ñ o C., Kieser A. The viral oncoprotein LMP1 exploits TRADD for signaling by masking its apoptotic activity. PLoS Biol., 2008. vol. 6, no. 1: 8. doi: 10.1371/journal.pbio.0060008
  42. Schröfelbauer B., Polley S., Behar M. Ghosh G., Hoffmann A. NEMO ensures signaling specificity of the pleiotropic IKKβ by directing its kinase activity toward IκBα. Mol. Cell, 2012, vol. 47, pp. 111–121. doi: 10.1016/j.molcel.2012.04.020
  43. Schwerk C., Schulze-Osthoff K. Regulation of apoptosis by alternative pre-mRNA splicing. Mol. Cell, 2005, vol. 19, pp. 1–13. doi: 10.1016/j.molcel.2005.05.026
  44. Shinozaki-Ushiku A., Kunita A., Isogai M., Hibiya T., Ushiku T., Takada K., Fukayama M. Profiling of virus-encoded micro-RNAs in Epstein–Barr virus-associated gastric carcinoma and their roles in gastric carcinogenesis. J. Virol., 2015, vol. 89, no. 10, pp. 5581–5591. doi: 10.1128/JVI.03639-14
  45. Snow A.L., Lambert S. L., Natkunam Y., Esquivel C.O., Krams S.M., Martinez O.M. EBV can protect latently infected B cell lymphomas from death receptor-induced apoptosis. J. Immunol., 2006, vol. 177, pp. 3283–3293. doi: 10.4049/jimmunol.177.5.3283
  46. Solntsev L.A., Starikova V.D., Sakharnov N.A., Knyazev D.I., Utkin O.V. Strategy of probe selection for studying mRNAs that participate in receptor-mediated apoptosis signaling. Mol. Biol., 2015, vol. 49, no. 3, pp. 457–465. doi: 10.7868/S0026898415030167
  47. Steelman L.S., Pohnert S.C., Shelton J.G., Franklin R.A., Bertrand F.E., McCubrey J.A. JAK/STAT, Raf/MEK/ERK, PI3K/ Akt and BCR-ABL in cell cycle progression and leukemogenesis. Leukemia, 2004, vol. 18, no. 2, pp. 189–218. doi: 10.1038/sj.leu.2403241
  48. Tepper C.G., Seldin M.F. Modulation of caspase-8 and FLICE-inhibitory protein expression as a potential mechanism of Epstein–Barr virus tumorigenesis in Burkitt’s lymphoma. Blood, 1999, vol. 94, no. 5, pp. 1727–1737.
  49. Williams E.J., Embleton N.D., Clark J.E., Bythell M., Ward Platt M.P., Berrington J.E. Viral infections: contributions to late fetal death, stillbirth, and infant death. J. Pediatr., 2013, vol. 163, no. 2, pp. 424–428. doi: 10.1016/j.jpeds.2013.02.004
  50. Wu Z., Aryee M.J. Subset quantile normalization using negative control features. J. Comput. Biol., 2010, vol. 17, no. 10, pp. 1385–1395. doi: 10.1089/cmb.2010.0049
  51. Yachie A. Cytologic analysis of Epstein–Barr virus-associated T/Natural killer-cell lymphoproliferative diseases. Front. Pediatr., 2018, vol. 6: 327. doi: 10.3389/fped.2018.00327
  52. Zhu D.M., Shi J., Liu S., Liu Y., Zheng D. HIV infection enhances TRAIL-induced cell death in macrophage by down-regulating decoy receptor expression and generation of reactive oxygen species. PLoS One, 2011, vol. 6, no. 4: e18291. doi: 10.1371/journal.pone.0018291
  53. Zimber-Strobl U., Strobl L.J. EBNA2 and Notch signaling in Epstein–Barr virus mediated immortalization of B lymphocytes. Semin. Cancer Biol., 2001, vol. 11, no. 6, pp. 423–434. doi: 10.1006/scbi.2001.0409
  54. Zuo J., Thomas W.A., Haigh T.A., Fitzsimmons L., Long H.M., Hislop A.D., Taylor G.S., Rowe M. Epstein–Barr virus evades CD4+ T cell responses in lytic cycle through BZLF1-mediated down-regulation of CD74 and the cooperation of vBcl-2. PLoS Pathog., 2011, vol. 7, no. 12: 1002455. doi: 10.1371/journal.ppat.1002455

Supplementary files

Supplementary Files
Action
1. JATS XML

Copyright (c) 2020 Sakharnov N.A., Utkin O.V., Filatova E.N., Knyazev D.I., Presnyakova N.B.

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.

СМИ зарегистрировано Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор).
Регистрационный номер и дата принятия решения о регистрации СМИ: серия ПИ № ФС 77 - 64788 от 02.02.2016.


This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies