#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

RC1339/APRc from Is a Novel Aspartic Protease with Properties of Retropepsin-Like Enzymes


Several rickettsiae are pathogenic to humans by causing severe infections, including epidemic typhus (Rickettsia prowazekii), Rocky Mountain spotted fever (Rickettsia rickettsii), and Mediterranean spotted fever (Rickettsia conorii). Progress in correlating rickettsial genes and gene functions has been greatly hampered by the intrinsic difficulty in working with these obligate intracellular bacteria, despite the increasing insights into the mechanisms of pathogenesis of and the immune response to rickettsioses. Therefore, comparison of the multiple available genomes of Rickettsia is proving to be the most practical method to identify new factors that may play a role in pathogenicity. Here, we identified and characterized a novel retropepsin-like enzyme, APRc, that is expressed by at least two pathogenic rickettsial species, R. conorii and R. rickettsii. We have also established that APRc acts to process two major surface antigen/virulence determinants (OmpB/Sca5, OmpA/Sca0) in vitro and we suggest that this processing event is important for protein function. We demonstrate that APRc is specifically inhibited by drugs clinically used to treat HIV infections, providing the exciting possibility of targeting this enzyme for therapeutic intervention. With this work, we demonstrate that retropepsin-type aspartic proteases are indeed present in prokaryotes, suggesting that these enzymes may represent an ancestral form of these proteases.


Vyšlo v časopise: RC1339/APRc from Is a Novel Aspartic Protease with Properties of Retropepsin-Like Enzymes. PLoS Pathog 10(8): e32767. doi:10.1371/journal.ppat.1004324
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004324

Souhrn

Several rickettsiae are pathogenic to humans by causing severe infections, including epidemic typhus (Rickettsia prowazekii), Rocky Mountain spotted fever (Rickettsia rickettsii), and Mediterranean spotted fever (Rickettsia conorii). Progress in correlating rickettsial genes and gene functions has been greatly hampered by the intrinsic difficulty in working with these obligate intracellular bacteria, despite the increasing insights into the mechanisms of pathogenesis of and the immune response to rickettsioses. Therefore, comparison of the multiple available genomes of Rickettsia is proving to be the most practical method to identify new factors that may play a role in pathogenicity. Here, we identified and characterized a novel retropepsin-like enzyme, APRc, that is expressed by at least two pathogenic rickettsial species, R. conorii and R. rickettsii. We have also established that APRc acts to process two major surface antigen/virulence determinants (OmpB/Sca5, OmpA/Sca0) in vitro and we suggest that this processing event is important for protein function. We demonstrate that APRc is specifically inhibited by drugs clinically used to treat HIV infections, providing the exciting possibility of targeting this enzyme for therapeutic intervention. With this work, we demonstrate that retropepsin-type aspartic proteases are indeed present in prokaryotes, suggesting that these enzymes may represent an ancestral form of these proteases.


Zdroje

1. WeinertLA, WerrenJH, AebiA, StoneGN, JigginsFM (2009) Evolution and diversity of Rickettsia bacteria. BMC Biol 7: 6.

2. FournierPE, RaoultD (2009) Current knowledge on phylogeny and taxonomy of Rickettsia spp. Ann N Y Acad Sci 1166: 1–11.

3. GillespieJJ, WilliamsK, ShuklaM, SnyderEE, NordbergEK, et al. (2008) Rickettsia phylogenomics: unwinding the intricacies of obligate intracellular life. PLoS One 3: e2018.

4. GoddardJ (2009) Historical and recent evidence for close relationships among Rickettsia parkeri, R. conorii, R. africae, and R. sibirica: implications for rickettsial taxonomy. J Vector Ecol 34: 238–242.

5. ParolaP, PaddockCD, RaoultD (2005) Tick-borne rickettsioses around the world: emerging diseases challenging old concepts. Clin Microbiol Rev 18: 719–756.

6. WalkerDH (2007) Rickettsiae and rickettsial infections: the current state of knowledge. Clin Infect Dis 45 (Suppl 1) S39–S44.

7. WalkerDH, IsmailN (2008) Emerging and re-emerging rickettsioses: endothelial cell infection and early disease events. Nat Rev Microbiol 6: 375–386.

8. JonesKE, PatelNG, LevyMA, StoreygardA, BalkD, et al. (2008) Global trends in emerging infectious diseases. Nature 451: 990–993.

9. Stephen Dumler (2012) Clinical Disease: Current Treatment and New Challenges. In: Guy Hughes Palmer AFA, editor. Intracellular Pathogens II: Rickettsiales. Washington, D.C.: ASM Press. pp. 1–39.

10. GalvaoMA, SilvaLJ, NascimentoEM, CalicSB, SousaR, et al. (2005) Rickettsial diseases in Brazil and Portugal: occurrence, distribution and diagnosis. Rev Saude Publica 39: 850–856.

11. AzadAF (2007) Pathogenic rickettsiae as bioterrorism agents. Clin Infect Dis 45 (Suppl 1) S52–S55.

12. AnderssonSG, KurlandCG (1998) Reductive evolution of resident genomes. Trends Microbiol 6: 263–268.

13. BlancG, OgataH, RobertC, AudicS, SuhreK, et al. (2007) Reductive genome evolution from the mother of Rickettsia. PLoS Genet 3: e14.

14. DarbyAC, ChoNH, FuxeliusHH, WestbergJ, AnderssonSG (2007) Intracellular pathogens go extreme: genome evolution in the Rickettsiales. Trends Genet 23: 511–520.

15. WixonJ (2001) Featured organism: reductive evolution in bacteria: Buchnera sp., Rickettsia prowazekii and Mycobacterium leprae. Comp Funct Genomics 2: 44–48.

16. AnderssonSG, ZomorodipourA, AnderssonJO, Sicheritz-PontenT, AlsmarkUC, et al. (1998) The genome sequence of Rickettsia prowazekii and the origin of mitochondria. Nature 396: 133–140.

17. BechahY, ElKK, MediannikovO, LeroyQ, PelletierN, et al. (2010) Genomic, proteomic, and transcriptomic analysis of virulent and avirulent Rickettsia prowazekii reveals its adaptive mutation capabilities. Genome Res 20: 655–663.

18. FelsheimRF, KurttiTJ, MunderlohUG (2009) Genome sequence of the endosymbiont Rickettsia peacockii and comparison with virulent Rickettsia rickettsii: identification of virulence factors. PLoS One 4: e8361.

19. LebrunI, Marques-PortoR, PereiraAS, PereiraA, PerpetuoEA (2009) Bacterial toxins: an overview on bacterial proteases and their action as virulence factors. Mini Rev Med Chem 9: 820–828.

20. Potempa J, Travis J (2000) Proteinases as Virulence Factors in Bacterial Diseases and as Potential Targets for Therapeutic Intervention with Proteinase Inhibitors. In: Helm K, Korant B, Cheronis J, editors. Proteases as Targets for Therapy. 140th edition. New York: Springer Berlin Heidelberg. pp. 159–188.

21. WladykaB, PustelnyK (2008) Regulation of bacterial protease activity. Cell Mol Biol Lett 13: 212–229.

22. AmmermanNC, GillespieJJ, NeuwaldAF, SobralBW, AzadAF (2009) A typhus group-specific protease defies reductive evolution in rickettsiae. J Bacteriol 191: 7609–7613.

23. TemenakJJ, AndersonBE, McDonaldGA (2001) Molecular cloning, sequence and characterization of cjsT, a putative protease from Rickettsia rickettsii. Microb Pathog 30: 221–228.

24. RatnerL, HaseltineW, PatarcaR, LivakKJ, StarcichB, et al. (1985) Complete nucleotide sequence of the AIDS virus, HTLV-III. Nature 313: 277–284.

25. DunnBM, GoodenowMM, GustchinaA, WlodawerA (2002) Retroviral proteases. Genome Biol 3: REVIEWS3006.

26. DunnBM (2002) Structure and mechanism of the pepsin-like family of aspartic peptidases. Chem Rev 102: 4431–4458.

27. WlodawerA, GustchinaA (2000) Structural and biochemical studies of retroviral proteases. Biochim Biophys Acta 1477: 16–34.

28. CascellaM, MichelettiC, RothlisbergerU, CarloniP (2005) Evolutionarily conserved functional mechanics across pepsin-like and retroviral aspartic proteases. J Am Chem Soc 127: 3734–3742.

29. RawlingsND, BatemanA (2009) Pepsin homologues in bacteria. BMC Genomics 10: 437.

30. TusnadyGE, SimonI (2001) The HMMTOP transmembrane topology prediction server. Bioinformatics 17: 849–850.

31. BernardD, MehulB, Thomas-CollignonA, DelattreC, DonovanM, et al. (2005) Identification and characterization of a novel retroviral-like aspartic protease specifically expressed in human epidermis. J Invest Dermatol 125: 278–287.

32. MatsuiT, Kinoshita-IdaY, Hayashi-KisumiF, HataM, MatsubaraK, et al. (2006) Mouse homologue of skin-specific retroviral-like aspartic protease involved in wrinkle formation. J Biol Chem 281: 27512–27525.

33. ImamuraD, ZhouR, FeigM, KroosL (2008) Evidence that the Bacillus subtilis SpoIIGA protein is a novel type of signal-transducing aspartic protease. J Biol Chem 283: 15287–15299.

34. LouisJM, AnianaA, WeberIT, SayerJM (2011) Inhibition of autoprocessing of natural variants and multidrug resistant mutant precursors of HIV-1 protease by clinical inhibitors. Proc Natl Acad Sci U S A 108: 9072–9077.

35. LiM, DimaioF, ZhouD, GustchinaA, LubkowskiJ, et al. (2011) Crystal structure of XMRV protease differs from the structures of other retropepsins. Nat Struct Mol Biol 18: 227–229.

36. WanM, TakagiM, LohBN, XuXZ, ImanakaT (1996) Autoprocessing: an essential step for the activation of HIV-1 protease. Biochem J 316 (Pt 2) 569–573.

37. IdoE, HanHP, KezdyFJ, TangJ (1991) Kinetic studies of human immunodeficiency virus type 1 protease and its active-site hydrogen bond mutant A28S. J Biol Chem 266: 24359–24366.

38. FodorSK, VogtVM (2002) Characterization of the protease of a fish retrovirus, walleye dermal sarcoma virus. J Virol 76: 4341–4349.

39. SchillingO, OverallCM (2008) Proteome-derived, database-searchable peptide libraries for identifying protease cleavage sites. Nat Biotechnol 26: 685–694.

40. SchillingO, HuesgenPF, BarreO, auf demKU, OverallCM (2011) Characterization of the prime and non-prime active site specificities of proteases by proteome-derived peptide libraries and tandem mass spectrometry. Nat Protoc 6: 111–120.

41. SchechterI, BergerA (1967) On the size of the active site in proteases. I. Papain. Biochem Biophys Res Commun 27: 157–162.

42. MaraniP, WagnerS, BaarsL, GenevauxP, de GierJW, et al. (2006) New Escherichia coli outer membrane proteins identified through prediction and experimental verification. Protein Sci 15: 884–889.

43. ChanYG, CardwellMM, HermanasTM, UchiyamaT, MartinezJJ (2009) Rickettsial outer-membrane protein B (rOmpB) mediates bacterial invasion through Ku70 in an actin, c-Cbl, clathrin and caveolin 2-dependent manner. Cell Microbiol 11: 629–644.

44. ChanYG, RileySP, ChenE, MartinezJJ (2011) Molecular basis of immunity to rickettsial infection conferred through outer membrane protein B. . Infect Immun 79: 2303–2313.

45. ChanYG, RileySP, MartinezJJ (2010) Adherence to and invasion of host cells by spotted Fever group rickettsia species. Front Microbiol 1: 139.

46. HackstadtT, MesserR, CieplakW, PeacockMG (1992) Evidence for proteolytic cleavage of the 120-kilodalton outer membrane protein of rickettsiae: identification of an avirulent mutant deficient in processing. Infect Immun 60: 159–165.

47. UchiyamaT, KawanoH, KusuharaY (2006) The major outer membrane protein rOmpB of spotted fever group rickettsiae functions in the rickettsial adherence to and invasion of Vero cells. Microbes Infect 8: 801–809.

48. RileySP, GohKC, HermanasTM, CardwellMM, ChanYG, et al. (2010) The Rickettsia conorii autotransporter protein Sca1 promotes adherence to nonphagocytic mammalian cells. Infect Immun 78: 1895–1904.

49. CardwellMM, MartinezJJ (2009) The Sca2 autotransporter protein from Rickettsia conorii is sufficient to mediate adherence to and invasion of cultured mammalian cells. Infect Immun 77: 5272–5280.

50. ChenJC, HottesAK, McAdamsHH, McGrathPT, ViollierPH, et al. (2006) Cytokinesis signals truncation of the PodJ polarity factor by a cell cycle-regulated protease. EMBO J 25: 377–386.

51. LouisJM, WeberIT, TozserJ, CloreGM, GronenbornAM (2000) HIV-1 protease: maturation, enzyme specificity, and drug resistance. Adv Pharmacol 49: 111–146.

52. LouisJM, WondrakEM, KimmelAR, WingfieldPT, NashedNT (1999) Proteolytic processing of HIV-1 protease precursor, kinetics and mechanism. J Biol Chem 274: 23437–23442.

53. SibandaBL, BlundellT, HobartPM, FoglianoM, BindraJS, et al. (1984) Computer graphics modelling of human renin. Specificity, catalytic activity and intron-exon junctions. FEBS Lett 174: 102–111.

54. YamauchiT, NagahamaM, HoriH, MurakamiK (1988) Functional characterization of Asp-317 mutant of human renin expressed in COS cells. FEBS Lett 230: 205–208.

55. RawlingsND, WallerM, BarrettAJ, BatemanA (2013) MEROPS: the database of proteolytic enzymes, their substrates and inhibitors. Nucleic Acids Res 42: D503–509.

56. Dunn BM (2013) Chapter 54 - Feline Immunodeficiency Virus Retropepsin. In: Rawlings ND, Salvesen G, editors. Handbook of Proteolytic Enzymes. Oxford: Academic Press. pp. 230–234.

57. Goldfarb NE, Dunn BM (2013) Chapter 44 - Human Immunodeficiency Virus 1 Retropepsin. In: Rawlings ND, Salvesen G, editors. Handbook of Proteolytic Enzymes. Oxford: Academic Press. pp. 190–199.

58. SimoesI, FaroR, BurD, KayJ, FaroC (2011) Shewasin A, an active pepsin homolog from the bacterium Shewanella amazonensis. FEBS J 278: 3177–3186.

59. TangJ, JamesMN, HsuIN, JenkinsJA, BlundellTL (1978) Structural evidence for gene duplication in the evolution of the acid proteases. Nature 271: 618–621.

60. ChandranV, FronzesR, DuquerroyS, CroninN, NavazaJ, et al. (2009) Structure of the outer membrane complex of a type IV secretion system. Nature 462: 1011–1015.

61. DongC, BeisK, NesperJ, Brunkan-LamontagneAL, ClarkeBR, et al. (2006) Wza the translocon for E. coli capsular polysaccharides defines a new class of membrane protein. Nature 444: 226–229.

62. ZieglerK, BenzR, SchulzGE (2008) A putative alpha-helical porin from Corynebacterium glutamicum. J Mol Biol 379: 482–491.

63. KoebnikR, LocherKP, VanGP (2000) Structure and function of bacterial outer membrane proteins: barrels in a nutshell. Mol Microbiol 37: 239–253.

64. SchulzGE (2002) The structure of bacterial outer membrane proteins. Biochim Biophys Acta 1565: 308–317.

65. MansuetoP, VitaleG, CascioA, SeiditaA, PepeI, et al. (2012) New insight into immunity and immunopathology of Rickettsial diseases. Clin Dev Immunol 2012: 967852.

66. BlomAM, HallstromT, RiesbeckK (2009) Complement evasion strategies of pathogens-acquisition of inhibitors and beyond. Mol Immunol 46: 2808–2817.

67. BlancG, NgwamidibaM, OgataH, FournierPE, ClaverieJM, et al. (2005) Molecular evolution of rickettsia surface antigens: evidence of positive selection. Mol Biol Evol 22: 2073–2083.

68. DautinN (2010) Serine protease autotransporters of enterobacteriaceae (SPATEs): biogenesis and function. Toxins (Basel) 2: 1179–1206.

69. ThompsonJD, HigginsDG, GibsonTJ (1994) CLUSTAL W: improving the sensitivity of progressive multiple sequence alignment through sequence weighting, position-specific gap penalties and weight matrix choice. Nucleic Acids Res 22: 4673–4680.

70. MulderNJ, ApweilerR, AttwoodTK, BairochA, BarrellD, et al. (2003) The InterPro Database, 2003 brings increased coverage and new features. Nucleic Acids Res 31: 315–318.

71. PeiJ, KimBH, GrishinNV (2008) PROMALS3D: a tool for multiple protein sequence and structure alignments. Nucleic Acids Res 36: 2295–2300.

72. PerkinsDN, PappinDJ, CreasyDM, CottrellJS (1999) Probability-based protein identification by searching sequence databases using mass spectrometry data. Electrophoresis 20: 3551–3567.

73. CraigR, BeavisRC (2004) TANDEM: matching proteins with tandem mass spectra. Bioinformatics 20: 1466–1467.

74. KellerA, NesvizhskiiAI, KolkerE, AebersoldR (2002) Empirical statistical model to estimate the accuracy of peptide identifications made by MS/MS and database search. Anal Chem 74: 5383–5392.

75. ColaertN, HelsensK, MartensL, VandekerckhoveJ, GevaertK (2009) Improved visualization of protein consensus sequences by iceLogo. Nat Methods 6: 786–787.

76. RileySP, PattersonJL, NavaS, MartinezJJ (2013) Pathogenic Rickettsia species acquire vitronectin from human serum to promote resistance to complement-mediated killing. Cell Microbiol 16: 849–861.

77. NikaidoH (1994) Isolation of outer membranes. Methods Enzymol 235: 225–234.

78. HillmanRDJr, BaktashYM, MartinezJJ (2013) OmpA-mediated rickettsial adherence to and invasion of human endothelial cells is dependent upon interaction with alpha2beta1 integrin. Cell Microbiol 15: 727–741.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 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#