SciELO - Scientific Electronic Library Online

 
vol.51 issue1The controversial Burkholderia cepacia complex, a group of plant growth promoting species and plant, animals and human pathogens author indexsubject indexarticles search
Home Pagealphabetic serial listing  

Services on Demand

Journal

Article

Indicators

  • Have no cited articlesCited by SciELO

Related links

Share


Revista argentina de microbiología

Print version ISSN 0325-7541On-line version ISSN 1851-7617

Rev. argent. microbiol. vol.51 no.1 Ciudad Autónoma de Buenos Aires Mar. 2019

http://dx.doi.org/10.1016/j.ram.2018.05.001 

SPECIAL ARTICLE

https://doi.org/10.1016/j.ram.2018.05.001

Current scenario of plasmid-mediated colistin resistance in Latin America

Escenario actual de la resistencia a la colistina mediada por plásmidos en América Latina

 

Cecilia Quiroga,c, Marcela Nastrob, José Di Conzab,c,*

a Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPAM), Facultad de Medicina, Paraguay 2155, C1121ABG, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
b Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junín 954, C1113AAD, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
c Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290 (1425), Ciudad Autónoma de Buenos Aires, Argentina

Received 22 December 2017; accepted 9 May 2018
Available online 23 June 2018

*Corresponding author.

E-mail address: jdiconza@gmail.com (J. Di Conza).


ABSTRACT

Colistin resistance can occur by chromosomal mutations and by acquisition of plasmid-carrying determinants, mainly mcr-1. In the recent years, we have observed the out-burst of this resistance gene in our region. Due to the risk of the rapid dissemination of mcr-1, this finding has worried and alerted different actors from the health field and has become one of the most prolific topics. Our review compiles available reports of well-documented mcr-1-positive strains of Enterobacteriaceae, obtained from different samples in Argentina and other countries of Latin America. Furthermore, it addresses the association of mcr-1 with ESBL resistance markers and outlines the platforms involved in their dissemination.

© 2018 Asociación Argentina de Microbiología. Published by Elsevier Espana, S.L.U. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

KEYWORDS

mcr-1; Colistin resistance; Enterobacteriaceae; Latin America

RESUMEN

La resistencia a la colistina puede ocurrir por mutaciones cromosómicas o por la adquisición de determinantes localizados en plásmidos, el principal es mcr-1. En los últimos años hemos observado la explosiva aparición de este gen de resistencia en nuestra región. Debido al riesgo que implica la rápida diseminación de mcr-1, este hallazgo ha preocupado y alertado a los diferentes actores del área de la salud, y se ha convertido en uno de los temas de investigación más importantes. La presente revisión compila los informes de aislamientos portadores de mcr-1 debidamente documentados en Enterobacteriaceae, obtenidos de diferentes muestras en Argentina y otros países de América Latina. Además, aborda la asociación de mcr-1 con otros marcadores de resistencia, como las BLEE, y describe las plataformas involucradas en su diseminación.

© 2018 Asociación Argentina de Microbiología. Publicado por Elsevier España, S.L.U. Este es un artículo Open Access bajo la licencia CC BY-NC-ND (http://creativecommons.org/licenses/by-nc-nd/4.0/).

PALABRAS CLAVE

mcr-1; Resistencia a colistina; Enterobacteriaceae; América Latina


 

Polymyxins (colistin and polymyxin B) are polycationic peptides, which act by targeting the negative charges in bacterial lipopolysaccharide (LPS), the complex lipoglycan that makes up most of the gram-negative outer membrane. Colistin (Polymyxin E) is one of the few cationic antimicrobial peptides commercialized in both human and veterinary medicine. Since its introduction in the 1950s, colistin (COL) has been used mainly as a topical treatment in human medicine owing to its nephrotoxicity and neurotoxicity when given systemically. There are two commercially available forms: colistin sulphate (CS) and colistin methanesulphonate sodium (CMS), the latter being less toxic than colistin when administered parenterally and consequently being the most used both in parenteral and inhalational formulations. In aqueous solutions, CMS undergoes in vivo conversion9. On the other hand, CS is the only form of COL approved in pig production in some countries for the control of Entero-bacteriaceae infections, particularly for those caused by Escherichia coli. However, COL has been massively used at sub-inhibitory concentrations as growth promoters for decades to improve weight gain and therefore to maximize feed conversion efficiency in animal production22. Noteworthy, the last survey conducted by the World Organization for Animal Health (OIE) in the year 2012 revealed that only 10 countries reported the use of COL as growth promoter42. Interestingly, in the late 2000s and after decades of COL use in swine, several studies began to report a significant COL resistance rate in Enterobacteriaceae1,4,23,33,49,55. The increasing incidence of infections caused by multidrug resistant gram-negative bacteria, especially the ones caused by carbapenemase-producing bacilli in the last years, has led to the use of polymyxins as one of the last resort treatments49.

Until very recently, COL resistance occurred by chromosomal mutations only, and therefore, this resistance phenotype was spread exclusively via clonal transmission of resistant isolates. However, two years ago Liu et al. reported the emergence of a plasmid-mediated colistin resistance determinant, mcr-139. Due to the risk of its rapid dissemination, this finding has worried and alerted different actors from the health field, becoming one of the most prolific topics.

Acquired resistance to colistin

Acquired resistance to colistin in Enterobacteriaceae was described many years ago45,64, but in the last years colistin-resistant K. pneumoniae isolates were more frequently reported worldwide10-11-32-52, among others. In Latin America there are reports from Brazil, Ecuador, Venezuela and Colombia18,27,50,56,58.

Acquired resistance to COL in Enterobacteriaceae was first documented in Argentina in the last years together with the emergence of KPC-producing isolates, probably derived from the use of this antimicrobial agent in the therapy of infections by these multidrug-resistant K. pneumoniae isolates. This resistance is believed to have occurred by selection of resistant subpopulations in the epidemic clone belonging to ST258. However, it has also been observed in non KPC-producing K. pneumoniae isolates probably as a result of clonal spread43. The last report of the PAHO (Panamerican Health Organization) from the year 2015 informed a COL resistance rate of 10.3% in K. pneumoniae isolates from hospitalized patients in Argentina, whereas in Escherichia coli and Enterobacter cloacae the percentage only reached 2.1%51. Similar results were obtained at the University Hospital José de San Martín, where COL resistance in K. pneumoniae in the year 2010 was 2% and 3%, 3.6%, 3.8%, 7%, 12% and 8%, respectively, in the subsequent years (unpublished results). With regard to E. coli isolates, COL resistance emerged in the last two years, where an important increase from 0.2% to 1.3% was observed, mostly as a result of plasmid-mediated resistance44. On the other hand, acquired resistance to COL remains very low in Acinetobac-ter baumannii (0.2%) and Pseudomonas aeruginosa (2.5%) isolates51.

Chromosomal resistance: different pathways

Bacteria often use different strategies to protect themselves from exposure to cationic antimicrobial peptides such as colistin. Such strategies include modifications of their lipopolysaccharides (LPSs), which are involved in the initial step of interaction between polymyxins and bacteria8,47,48. These alterations can be achieved by several covalent changes in lipid A (also called endotoxin molecule) of LPS. Among them, the cationic substitution of the phosphate group by the addition of 4-amino-4-deoxy-i_-arabinose (L-Ara4N) and phosphoethanolamine (PEtN) are the most common LPS modifications. L-Ara4N modification is more effective than PEtN modification because the decrease in the net charge leading to colistin resistance is more effective. The synthesis and transfer of L-Ara4N and PEtN is mediated by an operon regulated by the activation of two-component systems (TCSs). PmrA/PmrB is the main system contributing to polymyxin resistance (by environmental stimuli, and specific mutations within the TCS), although in some gram-negative species is known to occur via PhoP/PhoQ8,48.

Other modifications associated to chromosomal polymyxin resistance have been described, such as the reduction of acyl groups mediated by an lpxR-like deacylation and hydroxylation of lipid A48. Moreover, it was reported that K. pneumoniae uses alternative strategies such as participation of an efflux pump or capsule formation48.

The plasmid-mediated resistance gene mcr-1

In 2015, the emergence of a plasmid-mediated colistin resistance marker, named mcr-1, was reported in China39. Since then, numerous reports of the mcr-1 gene have been documented in Enterobacteriaceae cultured from food, animals, environment and humans worldwide, most of them detected in over 20 countries within 3 months of its first report8.

The mcr-1 gene encodes a PEtN transferase leading to the addition of PEtN to the lipid A. An in silico analysis of the amino acid sequence of this protein revealed that it is strongly related to enzymes present in gramnegative bacteria, some of which are intrinsically resistant to polymyxin8,19,48,52. This gene was detected mainly in E. coli and to a lesser extent, in other bacteria such as K. pneumoniae. To date, several variants of mcr-1 have been reported in the literature (mcr-1.2 to mcr-1.8, and mcr-1.10)2,20,40,66,69 or are deposited in Genbank (variants mcr-1.9, mcr-1.11, mcr-1.12, and mcr-1.13). These variants have one amino acid change each and exhibit a similar effect on colistin resistance compared with mcr-1.

Very recently, four additional mcr determinants have been reported, mcr-2, mcr-3, mcr-4 and mcr-5. The mcr-2 gene was identified in an E. coli isolated from pigs and cattle in Belgium68. This marker was also recovered from human samples in Switzerland37. Phylogenetic analysis of mcr-2, which was also described as a PEtN transferase, showed 80.6% amino acid identity with mcr-1 and provided strong evidence that this protein may have originated from Moraxella catarrhalis68. The mcr-3 gene was found in E. coli isolated from pigs in Malaysia, in a clinical sample of K. pneumoniae isolated from Thailand, and a clinical sample of Salmonella Typhimurium from the United States70. Sequence analysis showed that mcr-3 is closely related to PEtN from Aeromonas spp. The mcr-4 gene was found in Salmonella Typhimurium and E. coli from human and food-producing animals in Europe13. MCR-4 has 34.0%, 35.0% and 49.0% amino acid sequence identity to MCR-1, MCR-2 and MCR-3, respectively. It has been proposed that mcr-4 may have emerged from a Shewanella species, a bacterium commonly present in aquatic niches. Lastly, mcr-5 was initially found in 14 Salmonella Paratyphi B dTa+ isolates12. MCR-5 shares 36.11% protein identity with MCR-1, 35.29% with MCR-2, 34.72% with MCR-3 and 33.71% with MCR-4. Sequence analysis revealed that mcr-5 might have been transferred from the environmental rod Cupriavidus gilardii.

These genes have completely changed the scenario of colistin resistance since they have become a potential threat to public health. In Argentina and other countries of Latin

America, mcr-1 occurrence is currently low, however, it is possible to observe its rapid increase (Table 1). Moreover, mcr-5 has been recently found in a colistin-resistant E. coli isolate from Brazil (NZ.PDMU00000000) (Fig. 1).

Table 1 mcr-1 -positive isolates reported in Latin America


Figure 1 mcr distribution in Latin America.

Furthermore, there is a clear association between this determinant and the different genes encoding p-lactamases, particularly those from the blaCTX-M family. It is worth mentioning that mcr-1 can be found in isolates collected not only from human and animal clinical samples but also from different sources such as pigs, wild birds, penguins, poultry production and environmental niches (Table 1). Among these isolates, E. coli was the most common species reported in Brazil and Argentina.

The coexistence of mcr-1 and carbapenem resistance determinants (such as KPC-2, NDM and IMP-8 carbapene-mases) in human samples is of great concern (Table 1). These findings highlight that Enterobacteriaceae isolates carrying both mcr-1 and carbapenemase-encoding genes may emerge as a serious threat to antimicrobial therapy.

Genetic environment and dissemination of the mcr gene

Previous studies have shown that mcr genes are commonly located in self-transferable plasmids, which are responsible for inter-species dissemination worldwide. Plasmid incompatibility groups IncI2, HI1, HI2, P, X1/X2, X3/X4, X4, FI, FIP and FII have been reported as carriers of the different mcr variants12,14,18,23,26,34,39'41,52,58,68,70. Furthermore, a hybrid plasmid, identified as IncI2/IncFIB also harbored this resistance gene67. In South America only a few reports identified the plasmids responsible for mcr spread. To date, only plasmids from incompatibility groups X4 and I2 carry the mcr gene in our region3,21,41,66. An initial analysis of the plasmid backbone suggests that these elements are highly similar to those reported in China, Europe or North America.

A common feature of mcr-1 variants is their genetic environment. This gene is commonly adjacent to the pap2 gene. Both genes can be found either surrounded by two copies of insertion sequence ISApl1, yielding a composite trans-poson, downstream to a single copy of ISApl1 or in the absence of this element62. It has been proposed that the presence or absence of this IS correlates with the adaptation of mcr-1 to a new host. A composite transposon indicates a recent acquisition of this marker whereas the absence of ISApl1 or the presence of a single copy suggests that it has been already adapted. Since all three genetic environments have been found in different plasmids, it has been hypothesized that independent recombination events have led to the adaptation of the mcr-pap2 segment to specific incompatibility groups, which are in turn responsible for dissemination of polymyxin resistance. However, a recent report demonstrated that the composite transposon Tn6330, which has two ISApl1 surrounding mcr-1, can translocate to other plasmids35. Therefore, the possibility of a wider dissemination to plasmids from other incompatibility groups should be considered. It is worth noting that a study on ISApl1 function showed that when this sequence is active it is detrimental to the host, which leads to its loss63. Indirectly, this phenomenon might control the dissemination of mcr-1.

In 2016 Xavier et al. identified the first mcr-2 gene in an IncX4 plasmid from bovine and porcine E. coli68. Further analysis showed that mcr-2 is located at the same site than mcr-1 when found in the IncX4 plasmid. Despite this fact, mcr-2 was found surrounded by two copies of the insertion sequence IS1595 allowing this DNA to generate circular intermediates65. This finding reveals the potentiality of mcr-2 to disseminate to other plasmids and therefore, to other organisms. More recently, novel mcr determinants have been described. The mcr-3 gene has been found in plasmids from the IncHI2 group harbored in Enterobactericeae as well as in the chromosome of Aeromonas veronii38,70. When located in plasmids, mcr-3 is adjacent to the truncated transposon element TnAs270, a mobile element probably responsible for its spread. On the other hand, mcr-4 and mcr-5 genes were found in small non-self-transferable plasmids from the ColE family12,13. Furthermore, the mcr-5 gene is located in a Tn3-family transposon.

Concluding remarks

The data summarized herein show the rapid spread of mcr-1 in Latin America. To the best of our knowledge, mcr-2, mcr-3 and mcr-4 determinants have not been found in our region yet.

The mcr-1 marker has been found in a wide variety of ecological niches, which represents a troublesome scenario for public health. In the absence of new antimicrobial agents against multidrug-resistant gram-negative pathogens, the effect of plasmid-mediated colistin resistance on human health should not be underestimated.

There is an urgent need for surveillance and molecular epidemiological studies in both human and veterinary medicine addressing the dissemination of mcr-1 among bacterial isolates. The fact that most mcr genes were found in other regions raises the question of whether a unique variant is locally disseminated or if the other variants were accurately sought.

Furthermore, we suggest a re-evaluation of the antimicrobial agents used in animal feeding in order to reduce the potential risk to pandrug resistance.

Funding

This work was supported by ANPCyT PICT 2013-1978, UBACyT 20020130100167BA and ANPCyT PICT 2015-1925.

Conflict of interest

The authors declare that they have no conflicts of interest.

Acknowledgements

The authors would like to thank Famiglietti A, Rodriguez CH and Gutkind G for their invaluable cooperation.

References

1. Aarestrup FM, Oliver Duran C, Burch DGS. Antimicrobial resistance in swine production. Anim Heal Res Rev. 2008;9:135-482.         [ Links ]

2. AbuOun M, Stubberfield EJ, Duggett NA, Kirchner M, Dormer L, Nunez-Garcia J, Randall LP, Lemma F, Crook DW, Teale C, Smith RP, Anjum MF. mcr-1 and mcr-2 variant genes identified in Moraxella species isolated from pigs in Great Britain from 2014 to 2015. J Antimicrob Chemother. 2017;72:2745-9.         [ Links ]

3. Aires CAM, da Conceipao-Neto OC, Tavares e Oliveira TR, Dias CF, Montezzi LF, Picao RC, Albano RM, Asensi MD, Carvalho-Assef APD. Emergence of the plasmid-mediated mcr-1 gene in clinical KPC-2-producing Klebsiella pneumoniae sequence type 392 in Brazil. Antimicrob Agents Chemother. 2017;61:e00317-417.         [ Links ]

4. Anjum MF, Duggett NA, Abu Oun M, Randall L, Nunez-Garcia J, Ellis RJ, Rogers J, Horton R, Brena C, Williamson S, Martelli F, Davies R, Teale C. Colistin resistance in Salmonella and Escherichia coli isolates from a pig farm in Great Britain. J Antimicrob Chemother. 2016;71:2306-13.         [ Links ]

5. Badano A, Ernst A, Larini S, Sempio R, Rapoport M, Albornoz E, Anchart E. Reporte del primer aislamiento de Escherichia coli portadora del gen mcr-1 en el Servicio de Microbiología del Hospital de Niños "V. J. Vilela" de Rosario, Santa Fe. In: XXIII Congreso Latinoamericano de Microbiología y XIV Congreso Argentino de Microbiología ALAM-CAM. 2016.         [ Links ]

6. Barbieri NL, Pimenta R, Araujo de Melo D, Wannemuehler Y, Nolan LK, Souza M, Logue CM. First identification of mcr-1 associated resistance in E. coli isolated from healthy production poultry in Rio de Janeiro state, Brazil. ASM Microbe. 2017.         [ Links ]

7. Barletta J, Salusso D, Errecalde L, Sisto A, Pasteran F, Perez H, Kaufman S. Resistencia transferible a polimixina B/colistín en E. coli mediada por gen mcr-1: reporte de cinco aislamientos clínicos en un Hospital General de la CABA. In: XXIII Congreso Latinoamericano de Microbiología y XIV Congreso Argentino de Microbiología ALAM-CAM. 2016.         [ Links ]

8. Baron S, Hadjadj L, Rolain J-M, Olaitan AO. Molecular mechanisms of polymyxin resistance: knowns and unknowns. Int J Antimicro Agents. 2016;48:583-91.         [ Links ]

9. Bergen PJ, Landersdorfer CB, Lee HJ, Li J, Nation RL. "Old" antibiotics for emerging multidrug-resistant bacteria. Curr Opin Infect Dis. 2012;25:626-33.         [ Links ]

10. Biswas S, Brunel J-M, Dubus J-C, Reynaud-Gaubert M, Rolain J-M. Colistin: an update on the antibiotic of the 21st century. Expert Rev Anti Infect Ther. 2012;10:917-34.         [ Links ]

11. Bogdanovich T, Adams-Haduch JM, Tian G-B, Nguyen MH, Kwak EJ, Muto CA, Doi Y. Colistin-resistant Klebsiella pneumoniae car-bapenemase (KPC)-producing Klebsiella pneumoniae belonging to the international epidemic clone ST258. Clin Infect Dis. 2011;53:373-6.         [ Links ]

12. Borowiak M, Fischer J, Hammerl JA, Hendriksen RS, Szabo I, Malorny B. Identification of a novel transposon-associated phosphoethanolamine transferase gene, mcr-5, conferring colistin resistance in d-tartrate fermenting Salmonella enterica subsp. Enterica serovar Paratyphi B. J Antimicrob Chemother. 2017;72:3317-24.         [ Links ]

13. Carattoli A, Villa L, Feudi C, Curcio L, Orsini S, Luppi A, Pezzotti G, Magistrali CF. Novel plasmid-mediated colistin resistance mcr-4 gene in Salmonella and Escherichia coli, Italy 2013 Spain and Belgium, 2015 to 2016. Eurosurveillance. 2017;22:30589.         [ Links ]

14. Dabos ML, Nastro M, Famiglietti A, Bonnin R, Rodriguez H, Dortet L, Naas T. Escherichia coli clinical isolates harboring mcr-1 and mcr-1.3 genes recovered from a university hospital in Buenos Aires Argentina. In: 27th ECCMID. 2017.         [ Links ]

15. da Conceição-Neto OC, Aires CAM, Pereira NF, da Silva LH, Picâo RC, Siqueira BN, Albano RM, Asensi MD, Carvalho-Assef AP. Detection of the plasmid-mediated mcr-1 gene in clinical KPC-2-producing Escherichia coli isolates in Brazil. Int J Antimicrob Agents. 2017;50:282-4.         [ Links ]

16. Dalmolin TV, Castro L, Mayer FQ, Zavascki AP, Martins AF, de Lima-Morales D, Barth AL. Co-occurrence of mcr-1 and blaKPC-2 in a clinical isolate of Escherichia coli in Brazil. J Antimicrob Chemother. 2017;72:2404-5.         [ Links ]

17. Dalmolin TV, Martins AF, Zavascki AP, de Lima-Morales D, Barth AL. Acquisition of the mcr-1 gene by a high-risk clone of KPC-2-producing Klebsiella pneumoniae ST437/CC258 Brazil. Diagn Microbiol Infect Dis. 2018;90:132-3.         [ Links ]

18. Delgado-Blas JF, Ovejero CM, Abadia-Patiño L, Gonzalez-Zorn B. Coexistence of mcr-1 and blaNDM-1 in Escherichia coli from Venezuela. Antimicrob Agents Chemother. 2016;60:6356 -8.         [ Links ]

19. Di Conza J, Radice M, Gutkind G. MCR-1: rethinking the origin. Int J Antimicrob Agents. 2017;50:737.         [ Links ]

20. Di Pilato V, Arena F, Tascini C, Cannatelli A, Henrici De Angelis L, Fortunato S, Giani T, Menichetti F, Rossolini GM. mcr-1.2, a new mcr variant carried on a transferable plasmid from a colistin-resistant KPC carbapenemase-producing Klebsiella pneumoniae strain of sequence type 512. Antimicrob Agents Chemother. 2016;60:5612-5.         [ Links ]

21. Dominguez J. Estudio y evaluación de la dinámica de resistencia a cefalosporinas y fluoroquinolonas en Escherichia coli y Salmonella spp. en animales de producción. Tesis de doctorado de la Universidad de Bueno Aires 2017. Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires.         [ Links ]

22. Dominguez JE, Figueroa Espinosa RA, Redondo LM, Cejas D, Gutkind GO, Chacana PA, Di Conza JA, Fernández-Miyakawa ME. Plasmid-mediate colistin resistanc in Escherichia coli recovered from healthy poultry. Rev Argent Microbiol. 2017;49:297-8.         [ Links ]

23. Doumith M, Godbole G, Ashton P, Larkin L, Dallman T, Day M, Day M, Muller-Pebody B, Ellington MJ, de Pinna E, Johnson AP, Hopkins KL, Woodford N. Detection of the plasmid-mediated mcr-1 gene conferring colistin resistance in human and food isolates of Salmonella enterica and Escherichia coli in England and Wales. J Antimicrob Chemother. 2016;71:2300-5.         [ Links ]

24. Elena A, Cejas D, Magariños F, Jewtuchowicz V, Facente A, Gutkind G, Di Conza J, Radice M. Spread of clonally related Escherichia coli harboring an IncA/C1 plasmid encoding IMP-8 and its recruitment into an unrelated MCR-1-containing isolate. Antimicrob Agents Chemother. 2018;62:e02414-7, https://doi.org/10.1128/AAC.02414-17.

25. Faccone D, Rapoport M, Albornoz E, Ceriana P, Lucero C, Gagetti P, Petroni A, MCR-Group, Pasteran F, Corso A. Epidemiology of mcr-J-producing Enterobacteriaceae clinical isolates from Argentina. ASM Microbe. 2017.         [ Links ]

26. Fernandes MR, McCulloch JA, Vianello MA, Moura Q, Pérez-Chaparro PJ, Esposito F, Sartori L, Dropa M, Matté MH, Lira DPA, Mamizuka EM, Lincopan N. First report of the globally disseminated IncX4 plasmid carrying the mcr-1 gene in a colistin-resistant Escherichia coli sequence type 101 isolate from a human infection in Brazil. Antimicrob Agents Chemother. 2016;60:6415-7.         [ Links ]

27. Fernandes MR, Moura Q, Sartori L, Silva KC, Cunha MP, Esposito F, Lopes R, Otutumi LK, Gonpalves DD, Dropa M, Matté MH, Monte DF, Landgraf M, Francisco GR, Bueno MF, de Oliveira Garcia D, Knöbl T, Moreno AM, Lincopan N. Silent dissemination of colistin-resistant Escherichia coli in South America could contribute to the global spread of the mcr-1 gene. Eurosurveillance. 2016;21:30214.         [ Links ]

28. Fernandes MR, Cardoso NS, Moura Q, Sartori L, Esposito F, Silva KC, Otutumi LK, Goncalves DD, Cunha MP, Dropa M, Knobl T, Matte MH, Moreno AM, Lincopan N. Co-production of MCR-1, CTX-M and CMY-2 plasmid-mediated ß-lactamase in Escherichia coli from food-producing animals in Brazil. In: XXIII Congreso Latinoamericano de Microbiología y XIV Congreso Argentino de Microbiología ALAM-CAM. 2016.         [ Links ]

29. Fernandes MR, Cerdeira L, Sellera FP, Monte DFM, Silva KC, Pérez-Chaparro PJ, McCulloch JA, Lincopan N. IncX4 plasmids are key vectors responsible for the dissemination of the mcr-1 gene among Escherichia coli from humans, animals, food and environment in Brazil. ASM Microbe. 2017.         [ Links ]

30. Figueroa R, Argaraña F, Marchisio M, Chuard D, Dominguez J, Gutkind G, Di Conza J. Descripción de dos casos clínicos de infecciones urinarias causadas por E. coli con resistencia a col-istina mediada por plásmidos. In: 4to Congreso Bioquímico del Litoral. 2017.         [ Links ]

31. Girardello R, Cury AP, Teberges RSR, Setubal RS, Inoue AP, Duarte AJS, Rossi F. Non-clonal detection of mcr-1 -producer Escherichia coli clinical isolates over one year in a Brazilian hospital. ASM Microbe. 2017.         [ Links ]

32. Kontopidou F, Plachouras D, Papadomichelakis E, Koukos G, Galani I, Poulakou G, Dimopoulos G, Antoniadou A, Armaganidis A, Giamarellou H. Colonization and infection b colistin-resistant gram negative bacteria in a cohort of critically ill patients. Clin Microbiol Infect. 2011;17:9-11.         [ Links ]

33. Kuo SC, Huang WC, Wang HY, Shiau YR, Cheng MF, Lauderdale TL. Colistin resistance gene mcr-1 in Escherichia coli isolates from humans and retail meats, Taiwan. J Antimicrob Chemother. 2016;71:2327-9.         [ Links ]

34. Li A, Yang Y.Miao M, Chavda KD, Mediavilla JR, Xie X, Feng P, Tang YW, Kreiswirth BN, Chen L, Du H. Complete sequences of mcr-1 -harboring plasmids from extended spectrum ß-lactamase- and carbapenemase-producing Enterobacteriaceae. Antimicrob Agents Chemother. 2016;60: 4351 -4.         [ Links ]

35. Li R, Xie M, Zhang J, Yang Z, Liu L, Liu X, Zhen Z, Chan EWC, Chen S. Genetic characterization of mcr-1-bearing plasmids to depict molecular mechanisms underlying dissemination of the colistin resistance determinant. Antimicrob Chemother. 2017;72:393-401.         [ Links ]

36. Liakopoulos A, Mevius DJ, Olse B, Bonnedahl J. The colistin resistance mcr-1 gene is going wild. J Antimicrob Chemother. 2016;71:2335-6.         [ Links ]

37. Liassine N, Assouvie L, Descombes MC, Tendon VD, Kieffer N, Poirel L, Nordmann P. Very low prevalence of MCR-1/MCR-2 plasmid-mediated colistin resistance in urinary tract Enterobac-teriaceae in Switzerland. Int J Infect Dis. 2016;51:4-5.         [ Links ]

38. Ling Z, Yin W, Li H, Zhang Q, Wang X, Wang Z, Ke Y, Wang Y, Shen J. Chromosome-mediated mcr-3 variants in Aeromonas veronii from chicken meat. Antimicrob Agents Chemother. 2017: 61.         [ Links ]

39. Liu YY, Wang Y, Walsh TR, Yi LX, Zhang R, Spencer J, Doi Y, Tian G, Dong B, Huang X, Yu LF, Gu D, Ren H, Chen X, Lv L, He D, Zhou H, Liang Z, Liu JH, Shen J. Emergence of plasmid-mediated colistin resistance mechanism MCR-1 in animals and human beings in China: a microbiological and molecular biological study. Lancet Infect Dis. 2016;16:161-8.         [ Links ]

40. Lu X, Hu Y, Luo M, Zhou H, Wang X, Du Y, Li Z, Xu J, Zhu B, Xu X, Kan B. MCR-1.6, a new MCR variant carried by an IncP plasmid in a colistin-resistant Salmonella enterica serovar Typhimurium isolate from a healthy individual. Antimicrob Agents Chemother. 2017;61:e02632-2716.         [ Links ]

41. Monte DF, Mem A, Fernandes MR, Cerdeira L, Esposito F, Galváo JA, Franco BDGM, Lincopan N, Landgraf M. Chicken meat as a reservoir of colistin-resistant Escherichia coli strains carrying mcr-1 genes in South America. Antimicrob Agents Chemother. 2017;61:e02718-2816.         [ Links ]

42. Moulin G, Góchez D, Diaz F, Szabo M, Lasley J. Outcomes from the OIE's questionnaire on antimicrobial use in animals in 2015. Bulletin de l'OIE. 2016:82-5, doi:10.20506/bull.2016.3.2563.         [ Links ]

43. Nastro M, Carranza N, Aprigliano F, Saposnik E, Barberis C, Garc a S, Vay C, Rodr guez CH, Famiglietti A. Emergence of colistin-resistant Klebsiell pneumoniae. Microbiological and epidemiological characterization of the isolates producing and non-producing KPC-type carbapenemase. Rev Argent Microbiol. 2013;45:185-90.         [ Links ]

44. Nastro M, Rodriguez CH, de Gregorio S, Foccoli M, Vay C, Famiglietti A. Resistencia adquirida a colistina en Klebsiella pneumoniae y E. coli en un Hospital Universitario. In: XXIII Congreso Latinoamericano de Microbiolog a y XIV Congreso Argentino de Microbiología ALAM-CAM. 2016.         [ Links ]

45. Nation RL, Li J. Colistin in the 21st century. Curr Opin Infect Dis. 2009;22:535-43.         [ Links ]

46. Nievas J, Torres D, Nicola F, Bonvehí P, Scocozza L, Alcalá W, Herrera F, Relloso S, Smayevsky J. Emergencia de aislamientos de Escherichia coli con resistencia a colistina mediada por plásmidos. Aspectos clínicos y microbiológicos. In: XXIII Congreso Latinoamericano de Microbiología y XIV Congreso Argentino de Microbiología ALAM-CAM. 2016.         [ Links ]

47. Nikaido H. Molecular basis of bacterial outer membrane permeability revisited. Microbiol Mol Biol Rev. 2003;67:593-656.         [ Links ]

48. Olaitan AO, Morand S, Rolain JM. Mechanisms of polymyxin resistance: acquired and intrinsic resistance in bacteria. Front Microbiol. 2014;5:643.         [ Links ]

49. Olaitan AO, Morand S, Rolain JM. Emergence of colistin-resistant bacteria in humans without colistin usage: a new worry and cause for vigilance. Int J Antimicrob Agents. 2016;47:1 -3.         [ Links ]

50. Ortega-Paredes D, Barba P, Zurita J. Colistin-resistant Escherichia coli clinical isolate harbouring the mcr-1 gene in Ecuador. Epidemiol Infect. 2016;144:2967-70.         [ Links ]

51. Pan American Health Organization: Home. Washington, DC, USA. Available from: www.paho.org [cited 2017].

52. Poirel L, Jayol A, Nordmann P. Polymyxins:. Antibacterial activity, susceptibility testing, and resistance mechanisms encoded by plasmids or chromosomes. Clin Microbiol Rev. 2017;30:557-96.         [ Links ]

53. Rapoport M, Faccone D, Pasteran F, Ceriana P, Albornoz E, Petroni A, Group MCR, Corso AA. First description of mcr-1-mediated colistin resistance in human infections caused by Escherichia coli i Latin America. Antimicrob Agents Chemother. 2016;60:4412-3.         [ Links ]

54. Rapoport M, Faccone D, Albornoz E, Ceriana P, Lucero C, Petroni A, MCR Group, Pasteran F, Corso A. Diseminación de aislamientos clínicos con resistencia plasmídica a colistina mediada por mcr-1 en Argentina. In: XXIII Congreso Latinoamericano de Microbiología y XIV Congreso Argentino de Microbiología ALAM-CAM. 2016.         [ Links ]

55. Rhouma M, Beaudry F, Thériault W, Letellier A. Colistin in pig production: chemistry, mechanism of antibacterial action, microbial resistance emergence and one health perspectives. Front Microbiol. 2016;7:1789.         [ Links ]

56. Rossi F, Girardello R, Cury AP, Di Gioia TSR, de Almeida JN, Duarte AJ da S. Emergence of colistin resistance in the largest university hospital complex of Sao Paulo, Brazil, over five years. Braz J Infect Dis. 2017;21:98-101.         [ Links ]

57. Rumi MV, Di Conza J, Pereyra A, Gentilini E, Gutkind G. Detection of mcr-1 gene in Escherichia coli from companion animals in Argentina. ASM Microbe. 2017.         [ Links ]

58. Saavedra SY, Diaz L, Wiesner M, Correa A, Arévalo SA, Reyes J, Hidalgo AM, de la Cadena E, Perenguez M, Montano LA, Ardila J, Ríos R, Ovalle MV, Díaz P, Porras P, Villegas MV, Arias CA, Beltrán M, Duarte C. Genomic and molecular characterization of clinical isolates of Enterobacteriaceae harboring mcr-1 in Colombia, 2002-2016. Antimicrob Agents Chemother. 2017;61:e00841-917.         [ Links ]

59. Saavedra SY, Arévalo SA, Duarte C, Hidalgo AM, Montano LA, Ovalle MV. Emergencia por detección del gen de resistencia a colistina mcr-1 en aislamientos clínicos de Salmonella enterica serovar Typhimurium y E. coli en Colombia. In: XXIII Congreso Latinoamericano de Microbiología y XIV Congreso Argentino de Microbiología ALAM-CAM. 2016.         [ Links ]

60. Saavedra SY, Diaz L, Wiesner M, Correa A, Arevalo SA, Reyes J, Hidalgo AN, De la Cadena E, Perenguez M, Montaño LA, Ardila J, Rios R, Ovalle MV, Diaz PL, Porras P, Villegas MV, Arias C, Beltrán M, Duarte C. Whole genome characterization of clinical isolates of Enterobacteriaceae harboring mcr-1 in Colombia, 2002 to 2016. ASM Microbe. 2017.         [ Links ]

61. Sellera FP, Fernandes MR, Sartori L, Carvalho MP, Esposito F, Nascimento CL, Dutra GH, Mamizuka EM, Pérez-Chaparro PJ, McCulloch JA, Lincopan N. Escherichia coli carrying IncX4 plasmid-mediated mcr-1 and blaCTX-M genes in infected migratory Magellanic penguins (Spheniscus magellanicus). J Antimicrob Chemother. 2017;72:1255-6.         [ Links ]

62. Snesrud E, He S, Chandler M, Dekker JP, Hickman AB, McGann P, Dyda F. A model for transposition of the colistin resistance gene mcr-1 by ISApl1. Antimicrob Agents Chemother. 2016;60:6973-6.         [ Links ]

63. Snesrud E, Ong AC, Corey B, Kwak YI, Clifford R, Gleeson T, Wood S, Whitman TJ, Lesho EP, Hinkle M, McGann P. Analysis of serial isolates of mcr-1-positive Escherichia coli reveals a highly active ISApl1 transposon. Antimicrob Agents Chemother. 2017:61.         [ Links ]

64. Srinivas P, Rivard K. Polymyxin resistance in Gram-negative pathogens. Curr Infect Dis Rep. 2017;19:38.         [ Links ]

65. Sun J, Xu Y, Gao R, Lin J, Wei W, Srinivas S, Li D, Yang RS, Li XP, Liao XP, Liu YH, Feng Y. Deciphering MCR-2 colistin resistance. MBio. 2017;8:e00625-717.         [ Links ]

66. Tijet N, Faccone D, Rapoport M, Seah C, Pasterán F, Ceriana P, Albornoz E, Corso A, Petroni A, Melano RG. Molecular characteristics of mcr-1 -carrying plasmids and new mcr-1 variant recovered from polyclonal clinical Escherichia coli from Argentina and Canada. PLOS ONE. 2017;12:e0180347.         [ Links ]

67. Wang Q, Sun J, Li J, Ding Y, Li X-P, Lin J, Hassan B, Feng Y. Expanding landscapes of the diversified mcr-1 -bearing plasmid reservoirs. Microbiome. 2017;5:70.         [ Links ]

68. Xavier BB, Lammens C, Ruhal R, Kumar-Singh S, Butaye P, Goossens H, Malhotra-Kumar S. Identification of a novel plasmid-mediated colistin-resistance gene, mcr-2, in Escherichia coli, Belgium, June 2016. Eurosurveillance. 2016;21:30280.         [ Links ]

69. Yang Y-Q, Li Y-X, Song T, Yang Y-X, Jiang W, Zhang A-Y, Guo X-Y, Liu B-H, Wang Y-X, Lei C-W, Xiang R, Wang H-N. Colistin resistance gene mcr-1 and its variant in Escherichia coli isolates from chickens in China. Antimicrob Agents Chemother. 2017;61:e01204-1216.         [ Links ]

70. Yin W, Li H, Shen Y, Liu Z, Wang S, Shen Z, Zhang R, Walsh TR, Shen J, Wang Y. Novel plasmid-mediated colistin resistance gene mcr-3 in Escherichia coli. MBio. 2017;8:e00543-617.         [ Links ]

0325-7541/© 2018 Asociación Argentina de Microbiología. Published by Elsevier Espana, S.L.U. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Creative Commons License All the contents of this journal, except where otherwise noted, is licensed under a Creative Commons Attribution License