#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Measles Immune Suppression: Lessons from the Macaque Model


Measles remains a significant childhood disease, and is associated with a transient immune suppression. Paradoxically, measles virus (MV) infection also induces robust MV-specific immune responses. Current hypotheses for the mechanism underlying measles immune suppression focus on functional impairment of lymphocytes or antigen-presenting cells, caused by infection with or exposure to MV. We have generated stable recombinant MVs that express enhanced green fluorescent protein, and remain virulent in non-human primates. By performing a comprehensive study of virological, immunological, hematological and histopathological observations made in animals euthanized at different time points after MV infection, we developed a model explaining measles immune suppression which fits with the “measles paradox”. Here we show that MV preferentially infects CD45RA memory T-lymphocytes and follicular B-lymphocytes, resulting in high infection levels in these populations. After the peak of viremia MV-infected lymphocytes were cleared within days, followed by immune activation and lymph node enlargement. During this period tuberculin-specific T-lymphocyte responses disappeared, whilst strong MV-specific T-lymphocyte responses emerged. Histopathological analysis of lymphoid tissues showed lymphocyte depletion in the B- and T-cell areas in the absence of apoptotic cells, paralleled by infiltration of T-lymphocytes into B-cell follicles and reappearance of proliferating cells. Our findings indicate an immune-mediated clearance of MV-infected CD45RA memory T-lymphocytes and follicular B-lymphocytes, which causes temporary immunological amnesia. The rapid oligoclonal expansion of MV-specific lymphocytes and bystander cells masks this depletion, explaining the short duration of measles lymphopenia yet long duration of immune suppression.


Vyšlo v časopise: Measles Immune Suppression: Lessons from the Macaque Model. PLoS Pathog 8(8): e32767. doi:10.1371/journal.ppat.1002885
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1002885

Souhrn

Measles remains a significant childhood disease, and is associated with a transient immune suppression. Paradoxically, measles virus (MV) infection also induces robust MV-specific immune responses. Current hypotheses for the mechanism underlying measles immune suppression focus on functional impairment of lymphocytes or antigen-presenting cells, caused by infection with or exposure to MV. We have generated stable recombinant MVs that express enhanced green fluorescent protein, and remain virulent in non-human primates. By performing a comprehensive study of virological, immunological, hematological and histopathological observations made in animals euthanized at different time points after MV infection, we developed a model explaining measles immune suppression which fits with the “measles paradox”. Here we show that MV preferentially infects CD45RA memory T-lymphocytes and follicular B-lymphocytes, resulting in high infection levels in these populations. After the peak of viremia MV-infected lymphocytes were cleared within days, followed by immune activation and lymph node enlargement. During this period tuberculin-specific T-lymphocyte responses disappeared, whilst strong MV-specific T-lymphocyte responses emerged. Histopathological analysis of lymphoid tissues showed lymphocyte depletion in the B- and T-cell areas in the absence of apoptotic cells, paralleled by infiltration of T-lymphocytes into B-cell follicles and reappearance of proliferating cells. Our findings indicate an immune-mediated clearance of MV-infected CD45RA memory T-lymphocytes and follicular B-lymphocytes, which causes temporary immunological amnesia. The rapid oligoclonal expansion of MV-specific lymphocytes and bystander cells masks this depletion, explaining the short duration of measles lymphopenia yet long duration of immune suppression.


Zdroje

1. BeckfordAP, KaschulaRO, StephenC (1985) Factors associated with fatal cases of measles. A retrospective autopsy study. S Afr Med J 68: 858–863.

2. AkramuzzamanSM, CuttsFT, WheelerJG, HossainMJ (2000) Increased childhood morbidity after measles is short-term in urban Bangladesh. Am J Epidemiol 151: 723–735.

3. ShanksGD, LeeSE, HowardA, BrundageJF (2011) Extreme mortality after first introduction of measles virus to the polynesian island of Rotuma, 1911. Am J Epidemiol 173: 1211–1222.

4. Von PirquetCE (1908) Das Verhalten der kutanen Tuberkulin-reaktion während der Masern. Dtsch Med Wochenschr 34: 1297–1300.

5. TamashiroVG, PerezHH, GriffinDE (1987) Prospective study of the magnitude and duration of changes in tuberculin reactivity during uncomplicated and complicated measles. Pediatr Infect Dis J 6: 451–454.

6. LisseI, SambB, WhittleH, JensenH, SoumareM, et al. (1998) Acute and long-term changes in T-lymphocyte subsets in response to clinical and subclinical measles. A community study from rural Senegal. Scand J Infect Dis 30: 17–21.

7. RyonJJ, MossWJ, MonzeM, GriffinDE (2002) Functional and phenotypic changes in circulating lymphocytes from hospitalized Zambian children with measles. Clin Diagn Lab Immunol 9: 994–1003.

8. Premenko-LanierM, RotaPA, RhodesGH, BelliniWJ, McChesneyMB (2004) Protection against challenge with measles virus (MV) in infant macaques by an MV DNA vaccine administered in the presence of neutralizing antibody. J Infect Dis 189: 2064–2071.

9. BankampB, HodgeG, McChesneyMB, BelliniWJ, RotaPA (2008) Genetic changes that affect the virulence of measles virus in a rhesus macaque model. Virology 373: 39–50.

10. HirschRL, GriffinDE, JohnsonRT, CooperSJ, Lindo de SorianoI, et al. (1984) Cellular immune responses during complicated and uncomplicated measles virus infections of man. Clin Immunol Immunopathol 31: 1–12.

11. WardBJ, JohnsonRT, VaisbergA, JaureguiE, GriffinDE (1991) Cytokine production in vitro and the lymphoproliferative defect of natural measles virus infection. Clin Immunol Immunopathol 61: 236–248.

12. GriffinDE, WardBJ (1993) Differential CD4 T cell activation in measles. J Infect Dis 168: 275–281.

13. Fugier-VivierI, Servet-DelpratC, RivaillerP, RissoanM-C, LiuY-J, et al. (1997) Measles virus suppresses cell-mediated immunity by interfering with the survival and functions of dendritic and T cells. J Exp Med 186: 813–823.

14. GrosjeanI, CauxC, BellaC, BergerI, WildF, et al. (1997) Measles virus infects human dendritic cells and blocks their allostimulatory properties for CD4+ T cells. J Exp Med 186: 801–812.

15. SchnorrJ-J, XanthakosS, KeikavoussiP, KampgenE, Ter MeulenV, et al. (1997) Induction of maturation of human blood dendritic cell precursors by measles virus is associated with immmunosuppression. Proc Natl Acad Sci U S A 94: 5326–5331.

16. GriffinDE, WardBJ, JaureguiE, JohnsonRT, VaisbergA (1989) Immune activation in measles. N Engl J Med 320: 1667–1672.

17. FerreiraCS, FrenzkeM, LeonardVH, WelsteadGG, RichardsonCD, et al. (2010) Measles virus infection of alveolar macrophages and dendritic cells precedes spread to lymphatic organs in transgenic mice expressing human signaling lymphocytic activation molecule (SLAM, CD150). J Virol 84: 3033–3042.

18. LemonK, De VriesRD, MesmanAW, McQuaidS, Van AmerongenG, et al. (2011) Early target cells of measles virus after aerosol infection of non-human primates. PLoS Pathog 7: e1001263.

19. De VriesRD, MesmanAW, GeijtenbeekTBH, DuprexWP, De SwartRL (2012) The pathogenesis of measles. Curr Opin Virol In press: DOI 10.1016/j.coviro.2012.03.005..

20. TatsuoH, OnoN, YanagiY (2001) Morbilliviruses use signaling lymphocyte activation molecules (CD150) as cellular receptors. J Virol 75: 5842–5850.

21. NoyceRS, BondreDG, HaMN, LinLT, SissonG, et al. (2011) Tumor cell marker PVRL4 (nectin 4) is an epithelial cell receptor for measles virus. PLoS Pathog 7: e1002240.

22. MühlebachMD, MateoM, SinnPL, PruferS, UhligKM, et al. (2011) Adherens junction protein nectin-4 is the epithelial receptor for measles virus. Nature 480: 530–3.

23. RacanielloV (2011) An exit strategy for measles virus. Science 334: 1650–1651.

24. SullivanJL, BarryDW, LucasSJ, AlbrechtP (1975) Measles infection of human mononuclear cells. I. Acute infection of peripheral blood lymphocytes and monocytes. J Exp Med 142: 773–784.

25. HuddlestoneJR, LampertPW, OldstoneMBA (1980) Virus-lymphocyte interactions: infection of Tg and Tm subsets by measles virus. Clin Immunol Immunopathol 15: 502–509.

26. ForthalDN, AarnaesS, BlandingJ, De la MazaL, TillesJG (1992) Degree and length of viremia in adults with measles. J Infect Dis 166: 421–424.

27. De SwartRL, LudlowM, De WitteL, YanagiY, Van AmerongenG, et al. (2007) Predominant infection of CD150+ lymphocytes and dendritic cells during measles virus infection of macaques. PLoS Pathog 3: e178.

28. CondackC, GrivelJ-C, DevauxP, MargolisL, CattaneoR (2007) Measles virus vaccine attenuation: suboptimal infection of lymphatic tissue and tropism alteration. J Infect Dis 196: 541–549.

29. De SwartRL (2009) Measles studies in the macaque model. Curr Top Microbiol Immunol 330: 55–72.

30. De VriesRD, LemonK, LudlowM, McQuaidS, YükselS, et al. (2010) In vivo tropism of attenuated and pathogenic measles virus expressing green fluorescent protein in macaques. J Virol 84: 4714–4724.

31. CocksBG, ChangCC, CarballidoJM, YsselH, De VriesJE, et al. (1995) A novel receptor involved in T-cell activation. Nature 376: 260–263.

32. De SalortJ, SintesJ, LlinasL, Matesanz-IsabelJ, EngelP (2011) Expression of SLAM (CD150) cell-surface receptors on human B-cell subsets: from pro-B to plasma cells. Immunol Lett 134: 129–136.

33. McChesneyMB, MillerCJ, RotaPA, ZhuY, AntipaL, et al. (1997) Experimental measles I. Pathogenesis in the normal and the immunized host. Virology 233: 74–84.

34. WarthinAS (1931) Occurrence of numerous large giant cells in the tonsils and pharyngeal mucosa in the prodromal stage of measles. Arch Pathol 11: 864–874.

35. De VriesRD, YükselS, OsterhausADME, De SwartRL (2010) Specific CD8+ T-lymphocytes control dissemination of measles virus. Eur J Immunol 40: 388–395.

36. Van BinnendijkRS, PoelenMCM, KuijpersKC, OsterhausADME, UytdeHaagFGCM (1990) The predominance of CD8+ T cells after infection with measles virus suggests a role for CD8+ class I MHC-restricted cytotoxic T lymphocytes (CTL) in recovery from measles. J Immunol 144: 2394–2399.

37. MongkolsapayaJ, JayeA, CallanMFC, MagnusenAF, McMichaelAJ, et al. (1999) Antigen-specific expansion of cytotoxic T lymphocytes in acute measles virus infection. J Virol 73: 67–71.

38. MoussallemTM, GuedesF, FernandesER, PagliariC, LancellottiCLP, et al. (2007) Lung involvement in childhood measles: severe immune dysfunction revealed by quantitative immunohistochemistry. Hum Pathol 38: 1239–1247.

39. SallustoF, LenigD, ForsterR, LippM, LanzavecchiaA (1999) Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401: 708–712.

40. Schneider-SchauliesS, Schneider-SchauliesJ (2009) Measles virus-induced immunosuppression. Curr Top Microbiol Immunol 330: 243–269.

41. HahmB (2009) Hostile communication of measles virus with host innate immunity and dendritic cells. Curr Top Microbiol Immunol 330: 271–287.

42. GriffinDE (2010) Measles virus-induced suppression of immune responses. Immunol Rev 236: 176–189.

43. SteineurMP, GrosjeanI, BellaC, KaiserlianD (1998) Langerhans cells are susceptible to measles virus infection and actively suppress T cell proliferation. Eur J Dermatol 8: 413–420.

44. Servet-DelpratC, VidalainP-O, BausingerH, ManieS, Le DeistF, et al. (2000) Measles virus induces abnormal differentiation of CD40 ligand-activated human dendritic cells. J Immunol 164: 1753–1760.

45. VidalainP-O, AzocarO, Rabourdin-CombeC, Servet-DelpratC (2001) Measles virus-infected dendritic cells develop immunosuppressive and cytotoxic activities. Immunobiol 204: 629–638.

46. De WitteL, De VriesRD, Van der VlistM, YükselS, LitjensM, et al. (2008) DC-SIGN and CD150 have distinct roles in transmission of measles virus from dendritic cells to T-lymphocytes. PLoS Pathog 4: e1000049.

47. Van der VlistM, De WitteL, De VriesRD, LitjensM, De JongMAWP, et al. (2011) Human Langerhans cells capture measles virus through Langerin and present viral antigens to CD4(+) T-cells but are incapable of cross-presentation. Eur J Immunol 41: 2619–2631.

48. AvotaE, AvotsA, NiewieskS, KaneLP, BommhardtU, et al. (2001) Disruption of Akt kinase activation is important for immunosuppression induced by measles virus. Nat Med 7: 725–731.

49. McCulloughB, KrakowkaS, KoestnerA (1974) Experimental canine distemper virus-induced lymphoid depletion. Am J Pathol 74: 155–170.

50. von MesslingV, SpringfeldC, DevauxP, CattaneoR (2003) A ferret model of canine distemper virus virulence and immunosuppression. J Virol 77: 12579–12591.

51. BeinekeA, PuffC, SeehusenF, BaumgartnerW (2009) Pathogenesis and immunopathology of systemic and nervous canine distemper. Vet Immunol Immunopathol 127: 1–18.

52. De SwartRL, VosHW, UytdeHaagFGCM, OsterhausADME, Van BinnendijkRS (1998) Measles virus fusion protein- and hemagglutinin-transfected cell lines are a sensitive tool for the detection of specific antibodies by a FACS-measured immunofluorescence assay. J Virol Methods 71: 35–44.

53. SimpanenE, vanER, IsomakiH (1977) Remission of juvenile rheumatoid arthritis (Still's disease) after measles. Lancet 2: 987–988.

54. YoshiokaK, MiyataH, MakiS (1981) Transient remission of juvenile rheumatoid arthritis after measles. Acta Paediatr Scand 70: 419–420.

55. LinCY, LinMT, HsiehYL, TsaoLY (1988) Transient disappearance of immunologic disorders and remission after intercurrent measles infections in children with chronic idiopathic thrombocytopenic purpura. J Clin Immunol 8: 207–213.

56. MossWJ, ScottS, NdhlovuZ, MonzeM, CuttsFT, et al. (2009) Suppression of human immunodeficiency virus type 1 viral load during acute measles. Pediatr Infect Dis J 28: 63–65.

57. RuelTD, AchanJ, GasasiraAF, CharleboisED, MehbratuT, et al. (2008) HIV RNA suppression among HIV-infected Ugandan children with measles. J Acquir Immune Defic Syndr 48: 225–227.

58. KretschmerR, JanewayCA, RosenFS (1968) Immunologic amnesia. Study of an 11-year-old girl with recurrent severe infections associated with dysgammaglobulinemia, lymphopenia and lymphocytotoxic antibody, resulting in loss of immunologic memory. Pediat Res 2: 7–16.

59. LangermansJA, AndersenP, van SoolingenD, VervenneRA, FrostPA, et al. (2001) Divergent effect of bacillus Calmette-Guerin (BCG) vaccination on Mycobacterium tuberculosis infection in highly related macaque species: implications for primate models in tuberculosis vaccine research. Proc Natl Acad Sci U S A 98: 11497–11502.

60. NguyenDT, De WitteL, LudlowM, YükselS, WiesmullerK-H, et al. (2010) The synthetic bacterial lipopeptide Pam3CSK4 modulates respiratory syncytial virus infection independent of TLR activation. PLoS Pathog 6: e1001049.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2012 Číslo 8
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#