#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Translocates into the Myocardium and Forms Unique Microlesions That Disrupt Cardiac Function


Hospitalization for community-acquired pneumonia carries a documented risk for adverse cardiac events. These occur during infection and contribute to elevated mortality rates in convalescent individuals up to 1 year thereafter. We describe a previously unrecognized pathogenic mechanism by which Streptococcus pneumoniae, the leading cause of community-acquired pneumonia, causes direct cardiotoxicity and forms microscopic bacteria-filled lesions within the heart. Microlesions were detected in experimentally infected mice and rhesus macaques, as well as in heart sections from humans who succumbed to invasive pneumococcal disease (IPD). Cardiac microlesion formation required interaction of the bacterial adhesin CbpA with host Laminin receptor and bacterial cell wall with Platelet-activating factor receptor. Microlesion formation also required the pore-forming toxin pneumolysin. When infected mice were rescued with antibiotics, we observed robust signs of collagen deposition at former lesion sites. Thus, microlesions and the scarring that occurs thereafter may explain why adverse cardiac events occur during and following IPD.


Vyšlo v časopise: Translocates into the Myocardium and Forms Unique Microlesions That Disrupt Cardiac Function. PLoS Pathog 10(9): e32767. doi:10.1371/journal.ppat.1004383
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004383

Souhrn

Hospitalization for community-acquired pneumonia carries a documented risk for adverse cardiac events. These occur during infection and contribute to elevated mortality rates in convalescent individuals up to 1 year thereafter. We describe a previously unrecognized pathogenic mechanism by which Streptococcus pneumoniae, the leading cause of community-acquired pneumonia, causes direct cardiotoxicity and forms microscopic bacteria-filled lesions within the heart. Microlesions were detected in experimentally infected mice and rhesus macaques, as well as in heart sections from humans who succumbed to invasive pneumococcal disease (IPD). Cardiac microlesion formation required interaction of the bacterial adhesin CbpA with host Laminin receptor and bacterial cell wall with Platelet-activating factor receptor. Microlesion formation also required the pore-forming toxin pneumolysin. When infected mice were rescued with antibiotics, we observed robust signs of collagen deposition at former lesion sites. Thus, microlesions and the scarring that occurs thereafter may explain why adverse cardiac events occur during and following IPD.


Zdroje

1. Corrales-MedinaVF, SuhKN, RoseG, ChirinosJA, DoucetteS, et al. (2011) Cardiac complications in patients with community-acquired pneumonia: a systematic review and meta-analysis of observational studies. PLoS Med 8: e1001048.

2. Corrales-MedinaVF, MusherDM, WellsGA, ChirinosJA, ChenL, et al. (2012) Cardiac complications in patients with community-acquired pneumonia: incidence, timing, risk factors, and association with short-term mortality. Circulation 125: 773–781.

3. Corrales-MedinaVF, SerpaJ, RuedaAM, GiordanoTP, BozkurtB, et al. (2009) Acute bacterial pneumonia is associated with the occurrence of acute coronary syndromes. Medicine 88: 154–159.

4. KaplanV, ClermontG, GriffinMF, KasalJ, WatsonRS, et al. (2003) Pneumonia: still the old man's friend? Arch Intern Med 163: 317–323.

5. KumarS, WangL, FanJ, KraftA, BoseME, et al. (2008) Detection of 11 common viral and bacterial pathogens causing community-acquired pneumonia or sepsis in asymptomatic patients by using a multiplex reverse transcription-PCR assay with manual (enzyme hybridization) or automated (electronic microarray) detection. J Clin Microbiol 46: 3063–3072.

6. MusherDM, RuedaAM, KakaAS, MaparaSM (2007) The association between pneumococcal pneumonia and acute cardiac events. Clin Infect Dis 45: 158–165.

7. Corrales-MedinaVF, MusherDM, ShachkinaS, ChirinosJA (2013) Acute pneumonia and the cardiovascular system. Lancet 381: 496–505.

8. BoydJH, MathurS, WangY, BatemanRM, WalleyKR (2006) Toll-like receptor stimulation in cardiomyoctes decreases contractility and initiates an NF-kappaB dependent inflammatory response. Cardiol Res 72: 384–393.

9. RolliJ, Rosenblatt-VelinN, LiJ, LoukiliN, LevrandS, et al. (2010) Bacterial flagellin triggers cardiac innate immune responses and acute contractile dysfunction. PloS One 5: e12687.

10. FillonS, SoulisK, RajasekaranS, Benedict-HamiltonH, RadinJN, et al. (2006) Platelet-activating factor receptor and innate immunity: uptake of Gram-positive bacterial cell wall into host cells and cell-specific pathophysiology. J Immunol 177: 6182–6191.

11. HsuHE, ShuttKA, MooreMR, BeallBW, BennettNM, et al. (2009) Effect of pneumococcal conjugate vaccine on pneumococcal meningitis. N Eng J Med 360: 244–256.

12. CundellDR, GerardNP, GerardC, Idanpaan-HeikkilaI, TuomanenEI (1995) Streptococcus pneumoniae anchor to activated human cells by the receptor for platelet-activating factor. Nature 377: 435–438.

13. OrihuelaCJ, MahdaviJ, ThorntonJ, MannB, WooldridgeKG, et al. (2009) Laminin receptor initiates bacterial contact with the blood brain barrier in experimental meningitis models. J Clin Invest 119: 1638–1646.

14. MitchellTJ, AndrewPW (1997) Biological properties of pneumolysin. Microb Drug Resist 3: 19–26.

15. FarrandAJ, LaChapelleS, HotzeEM, JohnsonAE, TwetenRK (2010) Only two amino acids are essential for cytolytic toxin recognition of cholesterol at the membrane surface. Proc Natl Acad Sci USA 107: 4341–4346.

16. BraunJS, NovakR, HerzogKH, BodnerSM, ClevelandJL, et al. (1999) Neuroprotection by a caspase inhibitor in acute bacterial meningitis. Nat Med 5: 298–302.

17. YoshimuraA, LienE, IngallsRR, TuomanenE, DziarskiR, et al. (1999) Cutting edge: recognition of Gram-positive bacterial cell wall components by the innate immune system occurs via Toll-like receptor 2. J Immunol 163: 1–5.

18. Kinsella K, He W (2009) An Aging World: 2008. In: U.S. Census Bureau IPR, editor. Washington D.C.: U.S. Goverment Printing Office.

19. ChengAG, KimHK, BurtsML, KrauszT, SchneewindO, et al. (2009) Genetic requirements for Staphylococcus aureus abscess formation and persistence in host tissues. FASEB J 23: 3393–3404.

20. NelsonS, HappelKI, ZhangP, MyersL, DufourJP, et al. (2013) Effect of bacterial pneumonia on lung simian immunodeficiency virus (SIV) replication in alcohol consuming SIV-infected rhesus macaques. Alcohol Clin Exp Res 37: 969–977.

21. OrihuelaCJ, MahdaviJ, ThorntonJ, MannB, WooldridgeKG, et al. (2009) Laminin receptor initiates bacterial contact with the blood brain barrier in experimental meningitis models. J Clin Invest 119: 1638–1646.

22. LuL, MaY, ZhangJR (2006) Streptococcus pneumoniae recruits complement factor H through the amino terminus of CbpA. J Biol Chem 281: 15464–15474.

23. McNeelaEA, BurkeA, NeillDR, BaxterC, FernandesVE, et al. (2010) Pneumolysin activates the NLRP3 inflammasome and promotes proinflammatory cytokines independently of TLR4. PLoS Pathog 6: e1001191.

24. FangR, TsuchiyaK, KawamuraI, ShenY, HaraH, et al. (2011) Critical roles of ASC inflammasomes in caspase-1 activation and host innate resistance to Streptococcus pneumoniae infection. J Immunol 187: 4890–4899.

25. HoegenT, TremelN, KleinM, AngeleB, WagnerH, et al. (2011) The NLRP3 inflammasome contributes to brain injury in pneumococcal meningitis and is activated through ATP-dependent lysosomal cathepsin B release. J Immunol 187: 5440–5451.

26. HuL, JoshiSB, LiyanageMR, PansalawattaM, AldersonMR, et al. (2013) Physical characterization and formulation development of a recombinant pneumolysoid protein-based pneumococcal vaccine. J Pharm Sci 102: 387–400.

27. LuoR, MannB, LewisWS, RoweA, HeathR, et al. (2005) Solution structure of choline binding protein A, the major adhesin of Streptococcus pneumoniae. EMBO J 24: 34–43.

28. MannB, ThorntonJ, HeathR, WadeKR, TwetenRK, et al. (2013) Broadly Protective Protein-Based Pneumococcal Vaccine Comprised of Pneumolysin Toxoid-CbpA Peptide Recombinant Fusion Protein. J Infect Dis 209: 1116–25.

29. LuL, LammME, LiH, CorthesyB, ZhangJR (2003) The human polymeric immunoglobulin receptor binds to Streptococcus pneumoniae via domains 3 and 4. J Biol Chem 278: 48178–48187.

30. ArenalA, HernandezJ, Perez-DavidE, Rubio-GuivernauJL, Ledesma-CarbayoMJ, et al. (2012) Do the spatial characteristics of myocardial scar tissue determine the risk of ventricular arrhythmias? Cardiol Res 94: 324–332.

31. DenekeT, MullerKM, LemkeB, LawoT, CalcumB, et al. (2005) Human histopathology of electroanatomic mapping after cooled-tip radiofrequency ablation to treat ventricular tachycardia in remote myocardial infarction. J Cardiovasc Electrophysiol 16: 1246–1251.

32. de BakkerJM, van CapelleFJ, JanseMJ, WildeAA, CoronelR, et al. (1988) Reentry as a cause of ventricular tachycardia in patients with chronic ischemic heart disease: electrophysiologic and anatomic correlation. Circulation 77: 589–606.

33. VermaA, MarroucheNF, SchweikertRA, SalibaW, WazniO, et al. (2005) Relationship between successful ablation sites and the scar border zone defined by substrate mapping for ventricular tachycardia post-myocardial infarction. J Cardiovasc Electrophysiol 16: 465–471.

34. WuKC (2012) Assessing risk for ventricular tachyarrhythmias and sudden cardiac death: is there a role for cardiac MRI? Circ Cardiovasc Imaging 5: 2–5.

35. BoydAR, ShivshankarP, JiangS, BertonMT, OrihuelaCJ (2012) Age-related defects in TLR2 signaling diminish the cytokine response by alveolar macrophages during murine pneumococcal pneumonia. Exp Gerontol 47: 507–518.

36. RoschJW, BoydAR, HinojosaE, PestinaT, HuY, et al. (2010) Statins protect against fulminant pneumococcal infection and cytolysin toxicity in a mouse model of sickle cell disease. J Clin Invest 120: 627–635.

37. ChopraV, RogersMA, BuistM, GovindanS, LindenauerPK, et al. (2012) Is statin use associated with reduced mortality after pneumonia? A systematic review and meta-analysis. Am J Med 125: 1111–1123.

38. SmithCC, DavidsonSM, LimSY, SimpkinJC, HothersallJS, et al. (2007) Necrostatin: a potentially novel cardioprotective agent? Cardiovasc Drugs Ther 21: 227–233.

39. KennedyCL, SmithDJ, LyrasD, ChakravortyA, RoodJI (2009) Programmed cellular necrosis mediated by the pore-forming alpha-toxin from Clostridium septicum. PLoS Pathog 5: e1000516.

40. AuthemanD, WyderM, PopoffM, D'HerdeK, ChristenS, et al. (2013) Clostridium perfringens beta-toxin induces necrostatin-inhibitable, calpain-dependent necrosis in primary porcine endothelial cells. PloS One 8: e64644.

41. KaczmarekA, VandenabeeleP, KryskoDV (2013) Necroptosis: the release of damage-associated molecular patterns and its physiological relevance. Immunity 38: 209–223.

42. RadinJN, OrihuelaCJ, MurtiG, GuglielmoC, MurrayPJ, et al. (2005) beta-Arrestin 1 participates in platelet-activating factor receptor-mediated endocytosis of Streptococcus pneumoniae. Infect Immun 73: 7827–7835.

43. TettelinH, NelsonKE, PaulsenIT, EisenJA, ReadTD, et al. (2001) Complete genome sequence of a virulent isolate of Streptococcus pneumoniae. Science 293: 498–506.

44. LanieJA, NgWL, KazmierczakKM, AndrzejewskiTM, DavidsenTM, et al. (2007) Genome sequence of Avery's virulent serotype 2 strain D39 of Streptococcus pneumoniae and comparison with that of unencapsulated laboratory strain R6. J Bacteriol 189: 38–51.

45. LizcanoA, ChinT, SauerK, TuomanenEI, OrihuelaCJ (2010) Early biofilm formation on microtiter plates is not correlated with the invasive disease potential of Streptococcus pneumoniae. Microb Pathog 48: 124–130.

46. RoschJW, BoydAR, HinojosaE, PestinaT, HuY, et al. (2010) Statins protect against fulminant pneumococcal infection and cytolysin toxicity in a mouse model of sickle cell disease. J Clin Invest 120: 627–635.

47. KushidaH (1961) Propylene oxide as a dehydrating agent for emedding with epoxy resin. J Electron Microscopy 10: 203.

48. AkacheB, GrimmD, PandeyK, YantSR, XuH, et al. (2006) The 37/67-kilodalton laminin receptor is a receptor for adeno-associated virus serotypes 8, 2, 3, and 9. J Virol 80: 9831–9836.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 9
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#