www.fgks.org   »   [go: up one dir, main page]

Academia.eduAcademia.edu
Corynebacterium ulcerans isolates from humans and dogs: fibrinogen, fibronectin and collagen-binding, antimicrobial and PFGE profiles Liliane Simpson-Louredo, Juliana Nunes Ramos, Renata Stavracakis Peixoto, Louisy Sanches Santos, Camila Azevedo Antunes, et al. Antonie van Leeuwenhoek Journal of Microbiology ISSN 0003-6072 Volume 105 Number 2 Antonie van Leeuwenhoek (2014) 105:343-352 DOI 10.1007/s10482-013-0080-5 1 23 Your article is protected by copyright and all rights are held exclusively by Springer Science +Business Media Dordrecht. This e-offprint is for personal use only and shall not be selfarchived in electronic repositories. If you wish to self-archive your article, please use the accepted manuscript version for posting on your own website. You may further deposit the accepted manuscript version in any repository, provided it is only made publicly available 12 months after official publication or later and provided acknowledgement is given to the original source of publication and a link is inserted to the published article on Springer's website. The link must be accompanied by the following text: "The final publication is available at link.springer.com”. 1 23 Author's personal copy Antonie van Leeuwenhoek (2014) 105:343–352 DOI 10.1007/s10482-013-0080-5 ORIGINAL PAPER Corynebacterium ulcerans isolates from humans and dogs: fibrinogen, fibronectin and collagen-binding, antimicrobial and PFGE profiles Liliane Simpson-Louredo • Juliana Nunes Ramos • Renata Stavracakis Peixoto • Louisy Sanches Santos • Camila Azevedo Antunes • Elisa Martins Ladeira • Cintia Silva Santos • Veronica Viana Vieira • Maria Helena Simões Villas Bôas • Raphael Hirata Jr. • Ana Luı́za Mattos-Guaraldi Received: 2 September 2013 / Accepted: 15 November 2013 / Published online: 27 November 2013 Ó Springer Science+Business Media Dordrecht 2013 Abstract Corynebacterium ulcerans has been increasingly isolated as an emerging zoonotic agent of diphtheria and other infections from companion animals. Since pets are able to act as symptomless carriers, it is also essential to identify virulence potential for humans of these isolates. In this work the ability of C. ulcerans to bind to fibrinogen (Fbg), fibronectin (Fn) and Type I collagen as well the genetic relationship among strains isolated from human and asymptomatic dogs in Rio de Janeiro (Brazil) were analyzed. Five pulsed-field gel electrophoresis (PFGE) profiles were demonstrated (I, II, III, IV and V). In addition, the IV and V profiles exhibiting C85 % similarity were expressed by the BR-AD41 and BR-AD61 strains from companion dogs living in L. Simpson-Louredo and J. N. Ramos contributed equally for the first authorship in this manuscript. L. Simpson-Louredo  J. N. Ramos  L. S. Santos  C. S. Santos  R. Hirata Jr.  A. L. Mattos-Guaraldi (&) Laboratory of Diphtheria and Corynebacteria of Clinical Relevance-LDCIC, Collaborating Center for Diphtheria of CGLAB/SVS/MS, School of Medical Sciences, Universidade do Estado do Rio de Janeiro—UERJ, Rio de Janeiro, RJ, Brazil e-mail: guaraldi@uerj.br the same neighborhood. Independent of the PFGEtypes, human and dog isolates showed affinity to Fbg, Fn and collagen. Heterogeneity of PFGE profiles indicated endemicity of C. ulcerans in the Rio de Janeiro metropolitan area. Differences in the expression of adhesins to the human extracellular matrix may contribute to variations in the virulence and zoonotic potential of C. ulcerans strains. Keywords Corynebacterium ulcerans  Dog  Fibrinogen  Fibronectin  PFGE Introduction Infection with Corynebacterium ulcerans is recognised as an emerging zoonotic disease and there is evidence that the number of cases and the severity of R. S. Peixoto Institute of Microbiology Professor Paulo de Góes, Universidade Federal do Rio de Janeiro (IMPPG/UFRJ), Rio de Janeiro, RJ, Brazil C. A. Antunes Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil L. Simpson-Louredo  J. N. Ramos  E. M. Ladeira  V. V. Vieira  M. H. S. V. Bôas National Institute for Quality Control in Health (INCQS), Fundação Oswaldo Cruz—FIOCRUZ, Rio de Janeiro, RJ, Brazil 123 Author's personal copy 344 clinical signs is increasing in industrialized and developing countries (Dewinter et al. 2005; De Zoysa et al. 2005; Mattos-Guaraldi et al. 2008a; Wagner et al. 2010; Kamada et al. 2012; Urakawa et al. 2013). Human infections by C. ulcerans may be fatal and mostly occur in adults with close animal contact (Wellinghausen et al. 2002; Lartigue et al. 2005). Lately, C. ulcerans has been increasingly isolated as emerging zoonotic agent from companion animals such as cats and dogs (Dias et al. 2011a; Zakikhany and Efstratiou 2012). There is, therefore, a potentially large reservoir of infection with little knowledge about the risks of zoonotic transmission. The majority of the cases of diphtheria caused by C. ulcerans has occurred in adult patients who had been fully or partially immunized with diphtheria toxoid vaccine (Leek et al. 1990; Dias et al. 2011a). In Europe, C. ulcerans is currently isolated in more frequency from diphtheria cases than Corynebacterium diphtheriae (De Zoysa et al. 2005; Perkins et al. 2010; Taylor et al. 2010; Wagner et al. 2010). C. ulcerans does not cause large epidemics and human-to-human transmission remains uncertain (Bonnet and Begg 1999; Mattos-Guaraldi et al. 2008a). The aim of this study was to investigate the affinity of C. ulcerans to human plasma fibrinogen (Fbg), fibronectin (Fn) and Type I collagen as a virulence property. In addition, the antimicrobial susceptibility profiles and the genetic relationship among C. ulcerans strains isolated from asymptomatic dogs and a human patient in Rio de Janeiro metropolitan area, Brazil were also determined. Materials and methods Origin of bacterial strains and culture conditions Three C. ulcerans strains isolated from asymptomatic dogs and one from human patient in Rio de Janeiro metropolitan area, during the period of 2000–2012, were analyzed (Table 1): BR-AD41 and BR-AD61 strains currently isolated, respectively, from nares to skin wounds of two domestic dogs in Duque de Caxias city; BR-AD22 strain, previously isolated from nares of a shelter dog in Niterói city (Dias et al. 2010); 809 strain previously isolated from an elderly woman with a fatal pulmonary infection and history of leg skin ulcers in the Rio de Janeiro city (Mattos-Guaraldi et al. 123 Antonie van Leeuwenhoek (2014) 105:343–352 2008a). C. ulcerans CDC KC279 strain, Corynebacterium pseudotuberculosis 1002 strain; C. diphtheriae subsp. mitis ATCC 27010 (C7 s (-) tox- [NCTC 11397]) and ATCC 27012 strains were also analyzed for comparison. To rule out the presence of any infectious or debilitating diseases, including dermatoses and the more obvious physical and behavioral abnormalities, a general assessment of each animal’s condition and nutritional status was recorded as previously described (CCAC 2013). Swabs from skin lesions, nares and ears samples from companion to shelter dogs were collected with sterile swabs and inoculated onto the chocolate tellurite agar plates and incubated at 35–37 °C for 72 h as described by Dias et al. (2011b). Stock cultures in 10 % skim milk with 25 % added glycerol were maintained at -70 °C and recovered as required by cultivation in Trypticase Soy Agar (TSA; Difco Laboratories, Detroit, MI.) at 37 °C for 24 h under aerobic conditions (Dias et al. 2010). Phenotyping, molecular characterization and toxigenicity testing The identification of C. ulcerans strains was achieved using both phenotypic and genotypic methods. Positive bacterial cultures for irregular Gram-positive rods were preliminarily characterized by colonial morphology, pigmentation, hemolysis and DNAse activity. The suspect bacterial isolates were identified as C. ulcerans by conventional biochemical assays and the semi-automatized API-Coryne System V3.0 (bioMérrieux, Lyon, France) with the API web decoding system http://www.apiweb.biomerieux.com (Efstratiou and George 1999; Funke and Bernard 2007; Pimenta et al. 2008; Dias et al. 2010). The production of phospholipase D (Pld) was evaluated by the CAMP test (i.e., inhibition of hemolysis by Staphylococcus aureus). Molecular identification of the isolates was carried out by a multiplex PCR (mPCR) used to provide simultaneous identification and toxigenicity of these corynebacterial species with zoonotic potential in addition to C. diphtheriae, based on primers targeting the following genes: rpoB (Corynebacterium spp.), 16S rRNA (C. ulcerans and C. pseudotuberculosis), pld (specific for C. pseudotuberculosis), dtxR (C. diphtheriae) and tox gene (diphtheria toxin-DT) (Torres et al. 2013). Author's personal copy Antonie van Leeuwenhoek (2014) 105:343–352 345 Table 1 Corynebacterium ulcerans strains isolated from human and asymptomatic dogs in Rio de Janeiro metropolitan area, Brazil City/year Source Clinical detail API-Coryne code (probability, T = 0.99) References Rio de Janeiro/2000 Lung Fatal pulmonary infection 0111326 (99.7 %) Mattos-Guaraldi et al. (2008a), Trost et al. (2011) Niterói/2010 Nare Asymptomatic 0111324 (7.2 %) Dias et al. (2010), Trost et al. (2011) BR-AD41 Duque de Caxias/2012 Tick bite skin lesion Asymptomatic 0111326 (99.7 %) This study BR-AD61 Duque de Caxias/2012 Nare Asymptomatic 0111306 (96.6 %) This study C. ulcerans isolates Human 809 Dog shelter BR-AD22 Companion RJ Rio de Janeiro, DC Duque de Caxias, N Niterói Antimicrobial susceptibility profiles The sensitivity to antimicrobial agents (Oxoid, Hampshire, UK), penicillin G (10 U), ampicillin (10 lg), cefotaxime (30 lg), imipenem (10 lg), erythromycin (15 lg), clindamycin (2 lg), tetracycline (30 lg), ciprofloxacin (5 lg), gentamicin (10 lg), rifampicin (5 lg), linezolid (30 lg) and vancomycin (30 lg) was determined by the disk diffusion method using inoculum equivalent to a 0.5 McFarland standard, according to previously adopted by other authors (MartinezMartinez et al. 1995; CLSI 2007; Dias et al. 2010). Plates were incubated at 37 °C for 24 h and reconfirmed at 48 h using a cation-adjusted Mueller–Hinton agar with 5 % sheep blood. Breakpoints for the susceptible strains were used as suggested by the CLSI for bacteria excluded from table 2A to 2K. As there is not yet a defined standard for interpreting these results, the one proposed in the CLSI document M45A (ISBN 1-56238-607-7) was used (CLSI 2007). The breakpoints for S. aureus established by CLSI were considered in the cases of penicillin and ampicillin. Minimum inhibitory concentrations (MIC) of penicillin G and clindamycin were also evaluated by the Etest (AB Biodisk, Solna, Sweden), as previously described (Martinez-Martinez et al. 1995). ECM and plasma proteins binding assays Bacterial binding assays using biotinylated Fbg, Fn and Type I collagen (all from Sigma Chemical Co.) were performed in 96 wells Elisa Microtiter plates (Costar 96 Well EIA/RIA Plate; Corning, NY, USA). Bacterial cultures grown for 24 h at 37 °C in trypticase soy broth—TSB medium were washed 29 with phosphate buffered saline (PBS), and resuspended in 0.1 M NaHCO3, pH 9.6 to a suspension of OD 0.2 at k = 650 nm (equivalent to 5 9 109 CFU/mL). The wells were sensitised with 100 lL of bacterial suspensions for 1 h at 37 °C, and overnight at 8 °C. A standard curve was also performed by diluting the biotinylated protein solutions to concentrations varying from 5 to 0.05 lg (1 h/37 °C). After blocking with 2 % bovine serum albumin (BSA Type V, Sigma) in PBS added of 0.05 % Tween-20 (PBST) for 1 h at 37 °C, the wells were washed 39 with PBST. The bacterial strains were reacted with 20 lg/mL of biotinylated ECM/plasma proteins for 1 h at 37 °C. After washing 39 with PBST, the wells were reacted for 30 min at 37 °C with 0.001 lg/mL Extravidinperoxidase (Sigma) prepared in PBST 1 % BSA. After washing 39 with PBST, the reaction was verified by adding 3,30 ,5,50 -tetramethylbenzidine liquid substrate (TMB, Sigma) for 20 min and the reaction blocked with 50 lL of 1 M HCl. The reaction was read in k = 450 nm in a microtiter plate reader. The colour intensity of the wells sensitised with the microorganisms was compared to the standard curve by GraphPad Prism 6.0 version. The results were expressed in micrograms of adhered proteins, in a mean ± SD of three independent assays performed in triplicate. The mean of the binding properties were compared by 123 Author's personal copy 346 Tukey’s multiple comparison test (Harlow and Lane 1988; Sabbadini et al. 2010). Pulsed-field gel electrophoresis (PFGE) Genomic DNA was prepared following a method described previously (Baio et al. 2013). The DNA was cleaved with SfiI (New England BioLabs) according to the manufacturer’s instructions. PFGE was carried out in 0.59 TRIS–borate–EDTA–1.2 % agarose gels at 13 °C with a CHEF DRII system (Bio-Rad). The pulse times were 1–30 s over 20 h. A lambda DNA concatemer (New England BioLabs) was used as a molecular size marker. Similarities among macrorestriction patterns were identified according to the criteria established by Tenover et al. (1995). The BioNumerics Fingerprinting software (Version 4.0, Applied Math, Sint-Martins-Latem, Belgium) was used to confirm the findings provided by visual observation. The similarity index of the strains was calculated using the Dice correlation coefficient with a band position tolerance of 1 % and unweighted-pair group method using average linkages (UPGMA) was used to construct a dendrogram. Strains were considered to belong to the same PFGE group if the similarity index was C85 % percentage band-based similarity coefficients as cut-off values. Ethical procedures The study was performed in compliance with the guidelines outlined in the Canadian Council on Animal Care (CCAC) and with the Brazilian government’s ethical guidelines for research involving animals (Fiocruz Ethic Committee for Animal Experiments-CEUA/FIOCRUZ-LW-64/12). Antonie van Leeuwenhoek (2014) 105:343–352 glycogen fermentation were variable characteristics. Pld production was demonstrated by the positive reverse CAMP test. Amplification profiles by mPCR identified BR-AD22, BR-AD41 BR-AD61 and 809 Brazilian strains as C. ulcerans (Fig. 1) with negative results for tox gene. Antimicrobial susceptibility profiles C. ulcerans BR-AD22, BR-AD41, BR-AD61 and 809 strains showed moderate susceptibility to penicillin G and clindamycin (Table 2). All C. ulcerans strains tested showed susceptibility to other antimicrobial agents tested, including erythromycin and vancomycin. ECM/plasma proteins binding properties The ability of C. ulcerans to bind to human Fbg, Fn and Type I collagen molecules was demonstrated at varied levels (Fig. 2; Table 2). The three dog isolates (BR-AD22, BR-AD41, BR-AD61 strains) were capable to binding over than 10 % of biotinylated proteins, when reacted with 100 lL of a solution containing 20 lg/mL for 1 h/37 °C. Data showed a higher affinity of the dog isolates BR-AD22, BR-AD41, BR-AD61 strains to Fbg while a higher affinity of BRAD22 and BR-AD61 strains to Fn; and higher affinity of BR-AD22 to Type I collagen was shown. ELISA results showed the highest affinity to Fbg, Fn and Type I collagen for BR-AD22 strain (P \ 0.001). Conversely, 809 strain (isolated from a human patient with lethal pneumonia) adhered to human ECM/plasma proteins in lower intensities (P \ 0.001). CDC KC279 strain adhered to collagen and Fn in similar intensities of BR-AD61 and BR-AD41 strains, respectively (P [ 0.05). BR-AD41 and BR-AD61 showed similar adherence levels to Fbg (P [ 0.05). Results PFGE analysis Corynebacterium ulcerans phenotypic and genotypic characterization C. ulcerans strains were positive for catalase, urease, alkaline phosphatase and a-glucosidase. Nitrate reduction, pyrazinamidase, gelatinase, and esculin hydrolysis tests gave negative results. Fermentation tests were positive for glucose, ribose; negative for xylose, mannose, sucrose and lactose. Maltose and 123 PFGE of the SfiI-digested DNA of 5 strains revealed distinct PFGE profiles which were designated I, II, III, IV and V (Fig. 3). The PFGE profile IV differed by more than three bands from PFGE profile V by visual inspection. According to the interpretation criteria by Tenover et al. (1995), the BR-AD41 strain belonging to profile IV and BR-AD61 strain, profile V were considered related. These strains were isolated from Author's personal copy Antonie van Leeuwenhoek (2014) 105:343–352 347 Fig. 1 Amplification profile by multiplex polymerase chain reaction of Corynebacterium ulcerans (lanes 1–5), Corynebacterium diphtheriae (lanes 6 and 7), Corynebacterium pseudotuberculosis (lane 8): lane 1 BR-AD22 (tox-); 2 BR-AD41 (tox-); 3 BR-AD61 (tox-); 4 809; 5 CDC KC279 (tox?); 6 ATCC 27010 (tox-); 7 ATCC 27012 (tox?); 8 1002 (tox-); 9 molecular weight (100-bp DNA ladder) Table 2 Phenotypic and genotypic characteristics of Corynebacterium ulcerans strains isolated from human and dogs in Rio de Janeiro metropolitan area, Brazil (2000–2012) Strain number Antimicrobial agents CDC KC 279a Binding (lg)c PFGE type tox gene/ Pld/ DNAse Fbg Fn Collagen Penicillin G (MIC 0.19 C mg/ L); clindamycin (MIC C 1.5 mg/L) 1/1/1 0.2000 ± 0.0005774 0.4067 ± 0.006642 0.4680 ± 0.01328 I 809 Penicillin G, clindamycin 2/1/1 0.1270 ± 0.003464 0.1367 ± 0.007219 0.1650 ± 0.02801 II BRAD22 BRAD41 Penicillin G, clindamycin 2/1/1 0.2690 ± 0.004041 0.7160 ± 0.004619 0.1650 ± 0.02801 III Penicillin G, clindamycin 2/1/1 0.2290 ± 0.008660 0.3937 ± 0.001453 0.3280 ± 0.004041 IVd BRAD61 Penicillin G, clindamycin 2/1/1 0.2370 ± 0.0005774 0.6200 ± 0.001386 0.4557 ± 0.005487 Vd Moderate susceptibility to b NI not informed, PLD phospholipase D, DNAse deoxyribonuclease, Fbg fibrinogen, Fn fibronectin, ? positive, - negative a Control strain b Evaluated by disk diffusion method and E-test c Results expressed as a mean of three independent assays performed in triplicate d PFGE-types epidemiologically related dogs in the same neighborhood and exhibited C85 % similarity, so they were considered as belonging to the same PFGE group. The other PFGE profiles exhibited more than six bands of difference, indicating that they were epidemiologically unrelated. Discussion Toxigenic C. ulcerans and C. diphtheriae can lead to life-threatening disease that requires urgent treatment with diphtheria antitoxin (DAT) without waiting for laboratory confirmation. Antibiotic treatment of diphtheria-like illness caused by C. ulcerans should follow clinical guidelines for patients infected with C. diphtheriae. An appropriate antibiotic (penicillin or erythromycin) should be used to eliminate the causative organisms, stop exotoxin production and reduce communicability (Pickering et al. 2009; CDC 2011, 2012; Sekizuka et al. 2012). Clindamycin was formerly considered as an alternative in the treatment of the carrier of diphtheria bacilli, especially in those who were sensitive to penicillin (Zamiri and McEntegar 1972). Similar PFGE profiles or ribotypes have been previously observed in C. ulcerans strains isolated from human and animal colonization or infectious processes (Katsukawa et al. 2009, 2012; Komiya et al. 123 Author's personal copy 348 Fig. 2 Binding to human fibrinogen (Fbg), fibronectin (Fn) and Type I collagen by Corynebacterium ulcerans evaluated by enzyme linked immunosorbent assay (ELISA): BR-AD22, BRAD41 and BR-AD61 strains isolated from dogs, 809 strain from human; control CDC KC279 strain. Data showed the highest affinity to Fbg, Fn and Type I collagen for BR-AD22 strain (P \ 0.001). The results were expressed in mean ± SD of three independent assays performed in triplicate. The mean of the binding properties were compared by Tukey’s multiple comparison test 2010; Berger et al. 2011). In Japan, 45 C. ulcerans dog isolates including 39 strains of a predominant PFGE type (A2), showed resistance or decreased sensitivity to clindamycin. The authors suggested that transmission among asymptomatic dogs might have occurred (Katsukawa et al. 2012). In our study, heterogeneity of PFGE profiles observed in strains isolated from human and dogs indicated endemicity of C. ulcerans in Rio de Janeiro metropolitan area. C. ulcerans strains of five different PFGE types showed moderate susceptibility to penicillin G and clindamycin, as previously 123 Antonie van Leeuwenhoek (2014) 105:343–352 demonstrated for other Brazilian C. diphtheriae strains (Pereira et al. 2008). Interestingly, two epidemiologically related PFGE profiles (profiles IV and V) were found in the BR-AD41 and BR-AD61 dog isolates from Duque de Caxias city in 2012. Virulence mechanisms of C. ulcerans should become a matter of higher interest especially due to the increase in the number and severity of cases of infection in immunized or partially immunized individuals. C. ulcerans strains seem to be endowed with an array of virulence factors other than DT such as catalase, proteases, deoxyribonuclease (DNAse), neuraminidase H (NanH), endoglycosidase E (EndoE), Pld toxin and subunits of adhesive pili of the SpaDEF type (Trost et al. 2011). Corynebacterium ulcerans strains producing Pld, but not DT toxin, are able to cause severe disease in humans, such as lymphadenitis, dermatitis, subcutaneous abscess and acute pharyngitis and lower respiratory tract infections (pneumonia and granulomatous nodules in pulmonary tissues) (Desseau et al. 1995; Hommez et al. 1999; Hatanaka et al. 2003; Dias et al. 2011a). Consequently, C. ulcerans strains unable to produce DT toxin should not be underestimated. Pld may cause an increase in vascular permeability, has dermonecrotic properties, and reduces viability of neutrophils and macrophages (Schmiel and Miller 1999). In an attempt to further investigate possible mechanisms that promote C. ulcerans infection and hematogenic dissemination, a previous study revealed a strain-dependent arthritogenic potential independent of catalase, DNAse, Pld and DT production. Some C. ulcerans strains showed a higher arthritogenic and mortality potential when compared to C. diphtheriae strains during an in vivo experimental infection in mice. C. ulcerans arthritis also had a hematogenic spread and viable bacteria were recovered from joints, blood, kidneys, liver, and spleen but not from the heart and lungs of mice (Gaede and Heesemann 1995; Tissi et al. 1999; Puliti et al. 2006; Dias et al. 2011b). C. ulcerans 809 human isolate (tox gene negative) caused skin lesions with a large extent of yellowish-white fibrinous (fibrin) deposits in the lower limb of the patient (Mattos-Guaraldi et al. 2008a). Dermonecrotic activity was also observed for both human 809 and dog BR-AD22 isolates in experiments performed with guinea pigs (Mattos-Guaraldi et al. 2008b). Comparative analysis of the complete genomes of C. ulcerans 809 and BR-AD22 strains and detection of Author's personal copy Antonie van Leeuwenhoek (2014) 105:343–352 349 Fig. 3 Pulsed-field gel electrophoresis (PFGE) profiles of Brazilian Corynebacterium ulcerans strains isolated from human and dogs. a Dendrogram of results of PFGE data. The percent similarity scale is based on UPGMA clustering of Dice coefficients generated by BioNumerics software v 4.0; b PFGE assay. Lane 1 k DNA ladder PFGE marker; lane 2 CDC KC 279 control strain (USA), profile I; lane 3 809 (human), profile II; lane 4 BR-AD22 (dog), profile III; lane 5 BR- AD41 (dog), profile IV; lane 6 BR-AD61 (dog), profile V candidate virulence factors was recently performed (Trost et al. 2011). The detection and functional assignments of singletons confirmed that the repertoire of potential virulence factors of the sequenced C. ulcerans were different in the two selected isolates from a human and animal source (809 and BR-AD22 strains, respectively). Two genes encoding surfaceanchored proteins with LPXTG motif, including the SPAD gene for the major pilin subunit of an adhesive pilus structure, were also detected as singletons. SpaD protein of C. ulcerans BR-AD22 differs in its amino acid sequence when compared with the functional counterpart CULC809-01952 from C.ulcerans 809, demonstrated that pili of the two C. ulcerans strains varied significantly in the primary sequence of their major pilins that in principle constitute the shaft of the corynebacterial pilus structure. Data suggested differences in the adhesive properties of C. ulcerans strains. The first step in the infectious process of extracellular pathogens like C. diphtheriae is generally considered to be attachment to and colonization of host tissue surfaces. Evidence from other Grampositive pathogens suggests that bacterial surface adhesins recognizing adhesive matrix molecules or plasma proteins may serve as potential antigenic candidates for the development of novel immunotherapies, including diphtheria-like illness and invasive infections caused by C. ulcerans (Rivera et al. 2007). Fbg is a major protein in human plasma that has its synthesis dramatically upregulated during inflammation or under exposure to stress such systemic infections. It is, therefore, not surprising that many bacterial pathogens can interact with Fbg and manipulate its biology (Rivera et al. 2007). Fbg is primarily involved in the coagulation cascade system through its conversion to insoluble fibrin. Both Fbg and fibrin play overlapping roles in blood clotting, fibrinolysis, inflammatory response, cellular and matrix interactions and wound healing. Many bacterial pathogens exploit mechanisms involved in coagulation systems to colonize exposed tissue matrix proteins or evade immune mechanisms of bacterial clearance (Doolittle 1984; Lantz et al. 1985; Mosesson 2005; Sun 2006). The presence of Fbg onto bacterial surfaces may be an efficient trait to avoid phagocytosis in human hosts, as previously described with other Gram-positive pathogens (Schubert et al. 2002; Rennermalm et al. 2004; Pierno et al. 2006), including C. diphtheriae (Gomes et al. 2009; Sabbadini et al. 2010). Fn is a complex glycoprotein found in a soluble form in many body fluids (blood, saliva) and in an insoluble form as a component of cell surfaces, basement membranes, and the extracellular matrices. Soluble plasma Fn interacts with various bacteria and cell surfaces. Fn may serve as a receptor in the adherence of bacteria to host epithelial cell and may play an important role in tissue tropism. Fn can simultaneously bind to Fbg, fibrin, collagen, human cells and bacteria (Mosher 1975; Ruoslahti and Vaheri 1975; Engvall and Ruoslahti 1977; Engvall et al. 1978; 123 Author's personal copy 350 Livornese and Korzeniowski 1992). Type I collagen is the most prevalent form of several distinct types of collagen observed in the arterial walls, bone, dentin, dermis, tendon, and uterine wall. All types of collagen are active in Fn binding, but to a different degrees (Engvall et al. 1978). The binding to collagen and Fbg is mediated by the same binding site in Fn. The ability of all five PFGE-types of C. ulcerans to bind to human Fbg, Fn and Type I collagen molecules was demonstrated, but at varied levels. Similar to C. diphtheriae, qualitative and quantitative differences in the expression of Fbg, Fn and Type I collagen-binding adhesins may contribute to variations in the virulence potential to the human host and pseudomembrane formation by C. ulcerans strains (Sabbadini et al. 2010). The fact that the animal isolates adhered to human ECM/plasma proteins in higher intensities indicated that some C. ulcerans strains have evolved mechanisms to target human defense, resulting in a microbial invasion of cells constitutive of host barriers, disruption of barrier integrity, and systemic dissemination and invasion of deeper tissues. Acknowledgments This work was supported by Coordenação de Aperfeiçoamento de Pessoal de Nı́vel Superior (CAPES), Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Conselho Nacional de Desenvolvimento Cientı́fico e Tecnológico (CNPq), Programa Estratégico de Apoio à Pesquisa em Saúde-Fundação Oswaldo Cruz (PAPES V-FIOCRUZ), Sub-Reitoria de Pós-graduação e Pesquisa da Universidade do Estado do Rio de Janeiro (SR-2/UERJ). Conflict of interest The authors declare that they no have conflict of interest. References Baio PV, Mota HF, Freitas AD et al (2013) Clonal multidrugresistant Corynebacterium striatum within a nosocomial environment, Rio de Janeiro, Brazil. Mem Inst Oswaldo Cruz 108:23–29 Berger A, Huber I, Merbecks SS et al (2011) Toxigenic Corynebacterium ulcerans in woman and cat. Emerg Infect Dis 17:1767–1768 Bonnet JM, Begg NT (1999) Control of diphtheria: guidance for consultants in communicable disease control. Commun Dis Public Health 2:242–249 Canadian Council on Animal Care (CCAC) (2013) Guidelines on: procurement of animals used in science. http://www. ccac.ca/Documents/Standards/Guidelines/Procurement.pdf. Accessed 10 Mar 2013 123 Antonie van Leeuwenhoek (2014) 105:343–352 Centers for Disease Control and Prevention (CDC) (2011) Respiratory diphtheria-like illness caused by toxigenic Corynebacterium ulcerans—Idaho, 2010. MMWR 60:77 Centers for Disease Control and Prevention (2012) Health information for international travel. Oxford University Press, New York Clinical Laboratory Standards Institute (CLSI) (2007) Methods for antimicrobial dilution and disk susceptibility testing of infrequently isolated or fastidious bacteria, approved guideline. CLSI document M45-A (ISBN 1-56238-6077:4–6) De Zoysa A, Hawkey PM, Engler K, George R et al (2005) Characterization of toxigenic Corynebacterium ulcerans strains isolated from humans and domestic cats in the United Kingdom. J Clin Microbiol 43:4377–4381 Desseau RB, Brandt-Christensen M, Jensen OJ, Tonnesen P (1995) Pulmonary nodules due to Corynebacterium ulcerans. Eur Respir J 8(4):651–653 Dewinter LM, Bernard KA, Romney MG (2005) Human clinical isolates of Corynebacterium diphtheriae and Corynebacterium ulcerans collected in Canada from 1999 to 2003 but not fitting reporting criteria for cases of diphtheria. J Clin Microbiol 43:3447–3449 Dias AASO, Silva FC Jr, Pereira GA, Souza MC et al (2010) Corynebacterium ulcerans isolated from an asymptomatic dog kept in an animal shelter in the metropolitan area of Rio de Janeiro, Brazil. Vector Borne Zoonotic Dis 10: 743–748 Dias AASO, Santos LS, Sabbadini PS, Santos CS et al (2011a) Corynebacterium ulcerans diphtheria: an emerging zoonosis in Brazil and worldwide. Rev Saúde Pública 45:1176–1191 Dias AASO, Silva FC Jr, Santos LS, Ribeiro-Carvalho MM et al (2011b) Strain-dependent arthritogenic potential of the zoonotic pathogen Corynebacterium ulcerans. Vet Microbiol 53:323–331 Doolittle RF (1984) Fibrinogen and fibrin. Annu Rev Biochem 53:195–229 Efstratiou A, George RC (1999) Laboratory guidelines for the diagnosis of infections caused by Corynebacterium diphtheriae and Corynebacterium ulcerans. Commun Dis Public Health 2:250–257 Engvall E, Ruoslahti E (1977) Binding of soluble form of fibroblast surface protein, fibronectin, to collagen. Int J Cancer 20:1–5 Engvall E, Ruoslahti E, Miller EJ (1978) Affinity of fibronectin to collagens of different genetic types and to fibrinogen. J Exp Med 147:1584–1595 Funke G, Bernard AK (2007) Coryneform Gram-positive rods. In: Murray PR, Baron EJ, Jorgensen JH, Landry ML, Pfaller MA (eds) Manual of clinical microbiology, 9th edn. ASM Press, Washington, DC, pp 485–514 Gaede KI, Heesemann J (1995) Arthritogenicity of genetically manipulated Yersinia enterocolitica serotype O8 for Lewis rats. Infect Immun 63:714–719 Gomes DL, Martins CA, Faria LM, Santos LS et al (2009) Corynebacterium diphtheriae as an emerging pathogen in nephrostomy catheter-related infection: evaluation of traits associated with bacterial virulence. J Med Microbiol 58: 1419–1427 Author's personal copy Antonie van Leeuwenhoek (2014) 105:343–352 Harlow E, Lane D (1988) Antibodies: a laboratory manual. Cold Spring Harbor Laboratory Press, New York Hatanaka A, Tsunoda A, Okamoto M, Ooe K et al (2003) Corynebacterium ulcerans diphtheria in Japan. Emerg Infect Dis 9:752–753 Hommez J, Devriese LA, Vaneechoutte M, Riegel P et al (1999) Identification of nonlipophilic corynebacteria isolated form dairy cows with mastitis. J Clin Microbiol 37:954–957 Kamada T, Hatanaka A, Tasaki A, Honda K et al (2012) Case of caused by Corynebacterium ulcerans in Ibaraki Prefecture. Nihon Jibiinkoka Gakkai Kaiho 115:682–686 Katsukawa C, Kawahara R, Inoue K, Ishii A et al (2009) Toxigenic Corynebacterium ulcerans isolated from the domestic dog for the first time in Japan. Jpn J Infect Dis 62:171–172 Katsukawa C, Komiya T, Yamagishi H, Ishii A et al (2012) Prevalence of Corynebacterium ulcerans in dogs in Osaka. J Med Microbiol 61:266–273 Komiya T, Yukiji C, De Zoysa A, Masaaki I et al (2010) Two Japanese Corynebacterium ulcerans isolates from the same hospital: ribotype, toxigenicity and serum antitoxin titre. J Med Microbiol 59:1497–1504 Lantz MS, Switalski LM, Kornmam KS, Höök M (1985) Bacteroides intermedius binds fibrinogen. J Bacteriol 163:623–628 Lartigue MF, Monnet X, Lê Fleche A, Grimont PA et al (2005) Corynebacterium ulcerans in an immunocompromised patient with diphtheria and her dog. J Clin Microbiol 43:999–1001 Leek MD, Sivaloganathan S, Devaraj SK, Zamiri I et al (1990) Diphtheria with a rare difference Corynebacterium fatality with associated apoptotic cell death. Histopathology 6(2):187–189 Livornese LL, Korzeniowski OM (1992) Pathogenesis of infective endocarditis. In: Kaye D (ed) Infective endocarditis, 2nd edn. Raven Press, New York, pp 19–35 Martınez-Martınez L, Ortega MC, Suarez AL (1995) Comparison of E-test with broth microdilution and disk diffusion for susceptibility testing of coryneform bacteria. J Clin Microbiol 33:1318–1321 Mattos-Guaraldi AL, Sampaio JLM, Santos CS, Pimenta FP et al (2008a) First detection of Corynebacterium ulcerans producing diphtheria-like toxin in a case of human with pulmonary infection in the Rio de Janeiro metropolitan area, Brazil. Mem Inst Oswaldo Cruz 103:396–400 Mattos-Guaraldi AL, Villas-Boas MHS, Dias AASO, Silva Jr FC et al (2008b) Corynebacterium ulcerans infecting humans and dogs in the metropolitan area of Rio de Janeiro, Brazil. Presented at the proceedings of the second annual meeting of DIPNET & tenth international meeting of the European Laboratory Working Group on Diphtheria, Larnaca, Cyprus, 61 Mosesson MW (2005) Fibrinogen and fibrin structure and functions. J Thromb Haemost 3:1894–1904 Mosher DF (1975) Cross-linking of cold-insoluble globulin by fibrin-stabilizing factor. J Biol Chem 250:6614–6621 Pereira A, Pimenta FP, Santos FRW, Damasco PV et al (2008) Antimicrobial resistance among Brazilian Corynebacterium diphtheriae strains. Mem Inst Oswaldo Cruz 103: 507–510 351 Perkins SL, Cordery R, Nixon G, Abrahams A et al (2010) Investigations and control measures following a non-travel-associated case of toxigenic Corynebacterium diphtheriae, London, United Kingdom, December 2009– January 2010. Euro Surveill 15(16),19544 Pickering LK, Baker CJ, Kimberlin DW, Long SS (2009) Diphtheria. Red book: 2009 report of the committee on infectious diseases, 28th edn. American Academy of Pediatrics, Elk Grove Village, pp 280–283 Pierno M, Maravigna L, Piazza R, Visai L, Speziale P (2006) FbsAdriven fibrinogen polymerization: a bacterial ‘‘deceiving strategy’’. Phys Rev Lett 96(2):028108. doi:10.1103/ PhysRevLett.96.028108 Pimenta FP, Souza MC, Pereira GA, Hirata R Jr et al (2008) DNase test as a novel approach for the routine screening of Corynebacterium diphtheriae. Lett Appl Microbiol 46:307–311 Puliti M, Hunolstein CV, Marangi M, Bistoni F et al (2006) Experimental model of infection with nontoxigenic strains of Corynebacterium diphtheriae and development of septic arthritis. J Med Microbiol 55:229–235 Rennermalm A, Nilsson M, Flock JI (2004) The fibrinogen binding protein of Staphylococcus epidermidis is a target for opsonic antibodies. Infect Immun 72:3081 Rivera J, Vannakambadi G, Höök M, Speziale P (2007) Fibrinogen-binding proteins of Gram-positive bacteria. Thromb Haemost 98:503–511 Ruoslahti E, Vaheri A (1975) Interaction of soluble fibroblast surface antigen with fibrinogen and fibrin. Identity with cold insoluble globulin of human plasma. J Exp Med 141:497–501 Sabbadini PS, Genovez MR, Silva CF, Adelino TL et al (2010) Fibrinogen binds to nontoxigenic and toxigenic Corynebacterium diphtheriae strains. Mem Inst Oswaldo Cruz 105:706–711 Schmiel DH, Miller VJ (1999) Bacterial phospholipases and pathogenesis. Microbes Infect 1:1103–1112 Schubert A, Zakikhany K, Schreiner M, Frank R et al (2002) A fibrinogen receptor from group B Streptococcus interacts with fibrinogen by repetitive units novel ligand binding sites. Mol Microbiol 46:557–569 Sekizuka T, Yamamoto A, Komiya T, Kenri T et al (2012) Corynebacterium ulcerans 0102 carries the gene encoding diphtheria toxin on a prophage different from the C. diphtheriae NCTC 13129 prophage. BMC Microbiol 12:72. doi:10.1186/1471-2180-12-72 Sun H (2006) The interaction between pathogens and the host coagulation system. Physiology (Bethesda) 21:281–288 Taylor J, Saveedra-Campos M, Harwood D, Pritchard G et al (2010) Toxigenic Corynebacterium ulcerans infection in a veterinary student in London, United Kingdon. Euro Surveill 15:1–3 Tenover FC, Arbeit RD, Goering RV, Mickelsen PA et al (1995) Interpreting chromosomal DNA restriction patterns produced by pulsed-field gel electrophoresis: criteria for bacterial strain typing. J Clin Microbiol 33:2233–2239 Tissi L, Puliti M, Barluzzi R, Orefici G et al (1999) Role of tumor necrosis factor alpha, interleukin-1b, and interleukin-6 in a mouse model of group B streptococcal arthritis. Infect Immun 67:4545–4550 Torres LFC, Ribeiro D, Hirata R Jr, Pacheco LG et al (2013) Multiplex PCR for identification and toxigenicity of 123 Author's personal copy 352 Corynebacterium group of zoonotic potential and an overview of human and animal infections. Mem Inst Oswaldo Cruz 108(3):272–279 Trost E, Al-Dilaimi A, Papavasiliou P, Schneider J et al (2011) Comparative analysis of two complete Corynebacterium ulcerans genomes and detection of candidate virulence factors. BMC Genomics 12:383. doi:10.1186/1471-2164-12-383 Urakawa T, Seto J, Yamamoto A, Nakajima T et al (2013) (2013) Subcutaneous abscess formation in the upper extremity caused by toxigenic Corynebacterium ulcerans. J Med Microbiol 62:489–493 123 Antonie van Leeuwenhoek (2014) 105:343–352 Wagner KS, White JM, Neal S, Crowcroft NS et al (2010) Diphtheria in the United Kingdom, 1986–2008: the increasing role of Corynebacterium ulcerans. Epidemiol Infect 138:1519–1530 Wellinghausen N, Sing A, Kern WV, Perner S et al (2002) A fatal case of necrotizing sinusitis due to toxigenic Corynebacterium ulcerans. Int J Med Microbiol 292:59–63 Zakikhany K, Efstratiou A (2012) Diphtheria in Europe: current problems and new challenges. Future Microbiol 7:595–607 Zamiri I, McEntegar MGT (1972) The sensitivity of diphtheria bacilli to eight antibiotics. J Clin Path 25:716–717