1887

Abstract

Amphibian skin microbiomes can play a critical role in host survival against emerging diseases by protecting their host against pathogens. While a plethora of biotic and abiotic factors have been shown to influence the taxonomic diversity of amphibian skin microbiomes it remains unclear whether functional genomic diversity varies in response to temporal and environmental factors. Here we applied a metagenomic approach to evaluate whether seasonality, distinct elevations/sites, and pathogen presence influenced the functional genomic diversity of the skin microbiome. We obtained a gene catalogue of 92 107 nonredundant annotated genes and a set of 50 unique metagenome assembled genomes (MAGs). Our analysis showed that genes linked to general and potential antifungal traits significantly differed across seasons and sampling locations at different elevations. Moreover, we found that the functional genomic diversity of skin microbiome differed between . infected and not infected axolotls only during winter, suggesting an interaction between seasonality and pathogen infection. In addition, we identified the presence of genes and biosynthetic gene clusters (BGCs) linked to potential antifungal functions such as biofilm formation, quorum sensing, secretion systems, secondary metabolite biosynthesis, and chitin degradation. Interestingly genes linked to these potential antifungal traits were mainly identified in Burkholderiales and Chitinophagales MAGs. Overall, our results identified functional traits linked to potential antifungal functions in the skin microbiome regardless of variation in the functional diversity across seasons, elevations/sites, and pathogen presence. Our findings suggest that potential antifungal traits found in Burkholderiales and Chitinophagales taxa could be related to the capacity of to survive in the presence of Bd, although further experimental analyses are required to test this hypothesis.

Funding
This study was supported by the:
  • UNAM-PAPIIT (Award IA200921)
    • Principle Award Recipient: EriaA. Rebollar
  • This is an open-access article distributed under the terms of the Creative Commons Attribution License. This article was made open access via a Publish and Read agreement between the Microbiology Society and the corresponding author’s institution.
Loading

Article metrics loading...

/content/journal/mgen/10.1099/mgen.0.001181
2024-01-19
2024-04-28
Loading full text...

Full text loading...

/deliver/fulltext/mgen/10/1/mgen001181.html?itemId=/content/journal/mgen/10.1099/mgen.0.001181&mimeType=html&fmt=ahah

References

  1. Fisher MC, Henk DA, Briggs CJ, Brownstein JS, Madoff LC et al. Emerging fungal threats to animal, plant and ecosystem health. Nature 2012; 484:186–194 [View Article] [PubMed]
    [Google Scholar]
  2. Fisher MC, Gurr SJ, Cuomo CA, Blehert DS, Jin H et al. Threats posed by the fungal kingdom to humans, wildlife and agriculture. mBio 2020; 11:e00449-20 [View Article] [PubMed]
    [Google Scholar]
  3. Scheele BC, Pasmans F, Skerratt LF, Berger L, Martel A et al. Amphibian fungal panzootic causes catastrophic and ongoing loss of biodiversity. Science 2019; 363:1459–1463 [View Article] [PubMed]
    [Google Scholar]
  4. Allender MC, Ravesi MJ, Haynes E, Ospina E, Petersen C et al. Ophidiomycosis, an emerging fungal disease of snakes: targeted surveillance on military lands and detection in the western US and Puerto Rico. PLoS One 2020; 15:e0240415 [View Article] [PubMed]
    [Google Scholar]
  5. Hoyt JR, Kilpatrick AM, Langwig KE. Ecology and impacts of white-nose syndrome on bats. Nat Rev Microbiol 2021; 19:196–210 [View Article] [PubMed]
    [Google Scholar]
  6. Bahrndorff S, Alemu T, Alemneh T, Lund Nielsen J. The microbiome of animals: implications for conservation biology. Int J Genomics 2016; 2016:5304028 [View Article] [PubMed]
    [Google Scholar]
  7. Trevelline BK, Fontaine SS, Hartup BK, Kohl KD. Conservation biology needs a microbial renaissance: a call for the consideration of host-associated microbiota in wildlife management practices. Proc Biol Sci 2019; 286:20182448 [View Article] [PubMed]
    [Google Scholar]
  8. Wei F, Wu Q, Hu Y, Huang G, Nie Y et al. Conservation metagenomics: a new branch of conservation biology. Sci China Life Sci 2019; 62:168–178 [View Article] [PubMed]
    [Google Scholar]
  9. Hohenlohe PA, Funk WC, Rajora OP. Population genomics for wildlife conservation and management. Mol Ecol 2021; 30:62–82 [View Article] [PubMed]
    [Google Scholar]
  10. Siomko SA, Greenspan SE, Barnett KM, Neely WJ, Chtarbanova S et al. Selection of an anti-pathogen skin microbiome following prophylaxis treatment in an amphibian model system. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220126 [View Article] [PubMed]
    [Google Scholar]
  11. Hird SM. Evolutionary biology needs wild microbiomes. Front Microbiol 2017; 8:725 [View Article] [PubMed]
    [Google Scholar]
  12. Foster KR, Schluter J, Coyte KZ, Rakoff-Nahoum S. The evolution of the host microbiome as an ecosystem on a leash. Nature 2017; 548:43–51 [View Article] [PubMed]
    [Google Scholar]
  13. West AG, Waite DW, Deines P, Bourne DG, Digby A et al. The microbiome in threatened species conservation. Biol Conserv 2019; 229:85–98 [View Article]
    [Google Scholar]
  14. Zenner C, Hitch TCA, Riedel T, Wortmann E, Tiede S et al. Early-life immune system maturation in chickens using a synthetic community of cultured gut bacteria. mSystems 2021; 6:e01300-20 [View Article] [PubMed]
    [Google Scholar]
  15. Dogra SK, Kwong Chung C, Wang D, Sakwinska O, Colombo Mottaz S et al. Nurturing the early life gut microbiome and immune maturation for long term health. Microorganisms 2021; 9:2110 [View Article] [PubMed]
    [Google Scholar]
  16. McCoy KD, Burkhard R, Geuking MB. The microbiome and immune memory formation. Immunol Cell Biol 2019; 97:625–635 [View Article] [PubMed]
    [Google Scholar]
  17. Graham DB, Xavier RJ. Conditioning of the immune system by the microbiome. Trends Immunol 2023; 44:499–511 [View Article] [PubMed]
    [Google Scholar]
  18. Caballero-Pérez J, Espinal-Centeno A, Falcon F, García-Ortega LF, Curiel-Quesada E et al. Transcriptional landscapes of Axolotl (Ambystoma mexicanum). Dev Biol 2018; 433:227–239 [View Article] [PubMed]
    [Google Scholar]
  19. Libertucci J, Young VB. The role of the microbiota in infectious diseases. Nat Microbiol 2019; 4:35–45 [View Article] [PubMed]
    [Google Scholar]
  20. Boucias DG, Zhou Y, Huang S, Keyhani NO. Microbiota in insect fungal pathology. Appl Microbiol Biotechnol 2018; 102:5873–5888 [View Article] [PubMed]
    [Google Scholar]
  21. Brucker RM, Harris RN, Schwantes CR, Gallaher TN, Flaherty DC et al. Amphibian chemical defense: antifungal metabolites of the microsymbiont Janthinobacterium lividum on the salamander Plethodon cinereus. J Chem Ecol 2008; 34:1422–1429 [View Article] [PubMed]
    [Google Scholar]
  22. Bates KA, Sommer U, Hopkins KP, Shelton JMG, Wierzbicki C et al. Microbiome function predicts amphibian chytridiomycosis disease dynamics. Microbiome 2022; 10:44 [View Article] [PubMed]
    [Google Scholar]
  23. Rebollar EA, Martínez-Ugalde E, Orta AH. The amphibian skin microbiome and its protective role against chytridiomycosis. Herpetologica 2020; 76:167 [View Article]
    [Google Scholar]
  24. Hoyt JR, Cheng TL, Langwig KE, Hee MM, Frick WF et al. Bacteria isolated from bats inhibit the growth of Pseudogymnoascus destructans, the causative agent of white-nose syndrome. PLoS One 2015; 10:e0121329 [View Article] [PubMed]
    [Google Scholar]
  25. Campbell LJ, Pawlik AH, Harrison XA, Lesbarrères D. Amphibian ranaviruses in Europe: important directions for future research. FACETS 2020; 5:598–614 [View Article]
    [Google Scholar]
  26. Womack MC, Steigerwald E, Blackburn DC, Cannatella DC, Catenazzi A et al. State of the amphibia 2020: a review of five years of amphibian research and existing resources. Ichthyol Herpetol 2022; 110: [View Article]
    [Google Scholar]
  27. Dodd CK. eds Amphibian Ecology and Conservation: A Handbook of Techniques Oxford University Press; 2011
    [Google Scholar]
  28. Longcore JE, Pessier AP, Nichols DK. Batrachochytrium dendrobatidis gen. et sp. nov., a chytrid pathogenic to amphibians. Mycologia 1999; 91:219–227 [View Article]
    [Google Scholar]
  29. Martel A, Spitzen-van der Sluijs A, Blooi M, Bert W, Ducatelle R et al. Batrachochytrium salamandrivorans sp. nov. causes lethal chytridiomycosis in amphibians. Proc Natl Acad Sci U S A 2013; 110:15325–15329 [View Article] [PubMed]
    [Google Scholar]
  30. Rollins-Smith LA. The role of amphibian antimicrobial peptides in protection of amphibians from pathogens linked to global amphibian declines. Biochim Biophys Acta 2009; 1788:1593–1599 [View Article] [PubMed]
    [Google Scholar]
  31. Rollins-Smith LA, Reinert LK, O’Leary CJ, Houston LE, Woodhams DC. Antimicrobial Peptide defenses in amphibian skin. Integr Comp Biol 2005; 45:137–142 [View Article] [PubMed]
    [Google Scholar]
  32. Rollins-Smith LA, Doersam JK, Longcore JE, Taylor SK, Shamblin JC et al. Antimicrobial peptide defenses against pathogens associated with global amphibian declines. Dev Comp Immunol 2002; 26:63–72 [View Article] [PubMed]
    [Google Scholar]
  33. Ramsey JP, Reinert LK, Harper LK, Woodhams DC, Rollins-Smith LA. Immune defenses against Batrachochytrium dendrobatidis, a fungus linked to global amphibian declines, in the South African clawed frog, Xenopus laevis. Infect Immun 2010; 78:3981–3992 [View Article] [PubMed]
    [Google Scholar]
  34. Ellison AR, Tunstall T, DiRenzo GV, Hughey MC, Rebollar EA et al. More than skin deep: functional genomic basis for resistance to amphibian chytridiomycosis. Genome Biol Evol 2015; 7:286–298 [View Article] [PubMed]
    [Google Scholar]
  35. Becker MH, Harris RN, Minbiole KPC, Schwantes CR, Rollins-Smith LA et al. Towards a better understanding of the use of probiotics for preventing chytridiomycosis in Panamanian golden frogs. Ecohealth 2011; 8:501–506 [View Article] [PubMed]
    [Google Scholar]
  36. Harris RN, James TY, Lauer A, Simon MA, Patel A. Amphibian pathogen Batrachochytrium dendrobatidis is inhibited by the cutaneous bacteria of amphibian species. EcoHealth 2006; 3:53–56 [View Article]
    [Google Scholar]
  37. Harris RN, Brucker RM, Walke JB, Becker MH, Schwantes CR et al. Skin microbes on frogs prevent morbidity and mortality caused by a lethal skin fungus. ISME J 2009; 3:818–824 [View Article] [PubMed]
    [Google Scholar]
  38. Muletz CR, Myers JM, Domangue RJ, Herrick JB, Harris RN. Soil bioaugmentation with amphibian cutaneous bacteria protects amphibian hosts from infection by Batrachochytrium dendrobatidis. Biol Conserv 2012; 152:119–126 [View Article]
    [Google Scholar]
  39. Muletz-Wolz CR, Almario JG, Barnett SE, DiRenzo GV, Martel A et al. Inhibition of fungal pathogens across genotypes and temperatures by amphibian skin bacteria. Front Microbiol 2017; 8:1551 [View Article] [PubMed]
    [Google Scholar]
  40. Chen MY, Alexiev A, McKenzie VJ. Bacterial biofilm thickness and fungal inhibitory bacterial richness both prevent establishment of the amphibian fungal pathogen Batrachochytrium dendrobatidis. Appl Environ Microbiol 2022; 88:e0160421 [View Article] [PubMed]
    [Google Scholar]
  41. Harjai K, Sabharwal N. Biofilm formation and quorum sensing in rhizosphere. Biofilms Plant Soil Health 2017 [View Article]
    [Google Scholar]
  42. Lazazzera BA. Quorum sensing and starvation: signals for entry into stationary phase. Curr Opin Microbiol 2000; 3:177–182 [View Article] [PubMed]
    [Google Scholar]
  43. Trunk K, Peltier J, Liu Y-C, Dill BD, Walker L et al. The type VI secretion system deploys antifungal effectors against microbial competitors. Nat Microbiol 2018; 3:920–931 [View Article] [PubMed]
    [Google Scholar]
  44. Pena RT, Blasco L, Ambroa A, González-Pedrajo B, Fernández-García L et al. Relationship between quorum sensing and secretion systems. Front Microbiol 2019; 10:1100 [View Article] [PubMed]
    [Google Scholar]
  45. Brucker RM, Baylor CM, Walters RL, Lauer A, Harris RN et al. The identification of 2,4-diacetylphloroglucinol as an antifungal metabolite produced by cutaneous bacteria of the salamander Plethodon cinereus. J Chem Ecol 2008; 34:39–43 [View Article] [PubMed]
    [Google Scholar]
  46. De Boer W, Klein Gunnewiek PJA, Lafeber P, Janse JD, Spit BE et al. Anti-fungal properties of chitinolytic dune soil bacteria. Soil Biol Biochem 1998; 30:193–203 [View Article]
    [Google Scholar]
  47. Bhattacharya D, Nagpure A, Gupta RK. Bacterial chitinases: properties and potential. Crit Rev Biotechnol 2007; 27:21–28 [View Article] [PubMed]
    [Google Scholar]
  48. Bergstrom KSB, Kissoon-Singh V, Gibson DL, Ma C, Montero M et al. Muc2 protects against lethal infectious colitis by disassociating pathogenic and commensal bacteria from the colonic mucosa. PLoS Pathog 2010; 6:e1000902 [View Article] [PubMed]
    [Google Scholar]
  49. Zarepour M, Bhullar K, Montero M, Ma C, Huang T et al. The mucin Muc2 limits pathogen burdens and epithelial barrier dysfunction during Salmonella enterica serovar Typhimurium colitis. Infect Immun 2013; 81:3672–3683 [View Article] [PubMed]
    [Google Scholar]
  50. Woodhams DC, Rollins-Smith LA, Reinert LK, Lam BA, Harris RN et al. Probiotics modulate a novel amphibian skin defense peptide that is antifungal and facilitates growth of antifungal bacteria. Microb Ecol 2020; 79:192–202 [View Article] [PubMed]
    [Google Scholar]
  51. Piovia-Scott J, Rejmanek D, Woodhams DC, Worth SJ, Kenny H et al. Greater species richness of bacterial skin symbionts better suppresses the amphibian fungal pathogen Batrachochytrium dendrobatidis. Microb Ecol 2017; 74:217–226 [View Article] [PubMed]
    [Google Scholar]
  52. Woodhams DC, Bletz M, Kueneman J, McKenzie V. Managing amphibian disease with skin microbiota. Trends Microbiol 2016; 24:161–164 [View Article] [PubMed]
    [Google Scholar]
  53. Woodhams DC, LaBumbard BC, Barnhart KL, Becker MH, Bletz MC et al. Prodigiosin, violacein, and volatile organic compounds produced by widespread cutaneous bacteria of amphibians can inhibit two Batrachochytrium fungal pathogens. Microb Ecol 2017; 75:1049–1062 [View Article] [PubMed]
    [Google Scholar]
  54. Loudon AH, Holland JA, Umile TP, Burzynski EA, Minbiole KPC et al. Interactions between amphibians’ symbiotic bacteria cause the production of emergent anti-fungal metabolites. Front Microbiol 2014; 5:441 [View Article] [PubMed]
    [Google Scholar]
  55. Martin H C, Ibáñez R, Nothias L-F, Boya P CA, Reinert LK et al. Viscosin-like lipopeptides from frog skin bacteria inhibit Aspergillus fumigatus and Batrachochytrium dendrobatidis detected by imaging mass spectrometry and molecular networking. Sci Rep 2019; 9:3019 [View Article] [PubMed]
    [Google Scholar]
  56. Muletz-Wolz CR, DiRenzo GV, Yarwood SA, Campbell Grant EH, Fleischer RC et al. Antifungal bacteria on woodland salamander skin exhibit high taxonomic diversity and geographic variability. Appl Environ Microbiol 2017; 83:e00186-17 [View Article] [PubMed]
    [Google Scholar]
  57. Bletz MC, Myers J, Woodhams DC, Rabemananjara FCE, Rakotonirina A et al. Estimating herd immunity to amphibian chytridiomycosis in madagascar based on the defensive function of amphibian skin bacteria. Front Microbiol 2017; 8:1751 [View Article] [PubMed]
    [Google Scholar]
  58. Rebollar EA, Gutiérrez-Preciado A, Noecker C, Eng A, Hughey MC et al. The Skin microbiome of the neotropical frog Craugastor fitzingeri: inferring potential bacterial-host-pathogen interactions from metagenomic data. Front Microbiol 2018; 9:466 [View Article] [PubMed]
    [Google Scholar]
  59. Nava-González B, Suazo-Ortuño I, López PB, Maldonado-López Y, Lopez-Toledo L et al. Inhibition of Batrachochytrium dendrobatidis infection by skin bacterial communities in wild amphibian populations. Microb Ecol 2021; 82:666–676 [View Article] [PubMed]
    [Google Scholar]
  60. Rebollar EA, Hughey MC, Medina D, Harris RN, Ibáñez R et al. Skin bacterial diversity of Panamanian frogs is associated with host susceptibility and presence of Batrachochytrium dendrobatidis. ISME J 2016; 10:1682–1695 [View Article] [PubMed]
    [Google Scholar]
  61. McKenzie VJ, Bowers RM, Fierer N, Knight R, Lauber CL. Co-habiting amphibian species harbor unique skin bacterial communities in wild populations. ISME J 2012; 6:588–596 [View Article] [PubMed]
    [Google Scholar]
  62. Xu L, Xiang M, Zhu W, Zhang M, Chen H et al. The behavior of amphibians shapes their symbiotic microbiomes. mSystems 2020; 5:e00626-20 [View Article] [PubMed]
    [Google Scholar]
  63. Ellison S, Knapp RA, Sparagon W, Swei A, Vredenburg VT. Reduced skin bacterial diversity correlates with increased pathogen infection intensity in an endangered amphibian host. Mol Ecol 2019; 28:127–140 [View Article] [PubMed]
    [Google Scholar]
  64. Longo AV, Zamudio KR. Temperature variation, bacterial diversity and fungal infection dynamics in the amphibian skin. Mol Ecol 2017; 26:4787–4797 [View Article] [PubMed]
    [Google Scholar]
  65. Jani AJ, Bushell J, Arisdakessian CG, Belcaid M, Boiano DM et al. The amphibian microbiome exhibits poor resilience following pathogen-induced disturbance. ISME J 2021; 15:1628–1640 [View Article] [PubMed]
    [Google Scholar]
  66. Jani AJ, Briggs CJ. The pathogen Batrachochytrium dendrobatidis disturbs the frog skin microbiome during a natural epidemic and experimental infection. Proc Natl Acad Sci U S A 2014; 111:E5049–E5058 [View Article] [PubMed]
    [Google Scholar]
  67. Bletz MC, Archer H, Harris RN, McKenzie VJ, Rabemananjara FCE et al. Host ecology rather than host phylogeny drives amphibian skin microbial community structure in the biodiversity hotspot of Madagascar. Front Microbiol 2017; 8:1530 [View Article] [PubMed]
    [Google Scholar]
  68. Kueneman JG, Bletz MC, McKenzie VJ, Becker CG, Joseph MB et al. Community richness of amphibian skin bacteria correlates with bioclimate at the global scale. Nat Ecol Evol 2019; 3:381–389 [View Article] [PubMed]
    [Google Scholar]
  69. Bletz MC, Perl RGB, Bobowski BT, Japke LM, Tebbe CC et al. Amphibian skin microbiota exhibits temporal variation in community structure but stability of predicted Bd-inhibitory function. ISME J 2017; 11:1521–1534 [View Article] [PubMed]
    [Google Scholar]
  70. Familiar López M, Rebollar EA, Harris RN, Vredenburg VT, Hero J-M. Temporal variation of the skin bacterial community and Batrachochytrium dendrobatidis infection in the terrestrial cryptic frog Philoria loveridgei. Front Microbiol 2017; 8:2535 [View Article] [PubMed]
    [Google Scholar]
  71. Bresciano JC, Salvador CA, Paz-Y-Miño C, Parody-Merino AM, Bosch J et al. Variation in the presence of anti-Batrachochytrium dendrobatidis bacteria of amphibians across life stages and elevations in Ecuador. Ecohealth 2015; 12:310–319 [View Article] [PubMed]
    [Google Scholar]
  72. Barnes EM, Kutos S, Naghshineh N, Mesko M, You Q et al. Assembly of the amphibian microbiome is influenced by the effects of land-use change on environmental reservoirs. Environ Microbiol 2021; 23:4595–4611 [View Article] [PubMed]
    [Google Scholar]
  73. Kearns PJ, Fischer S, Fernández-Beaskoetxea S, Gabor CR, Bosch J et al. Fight fungi with fungi: antifungal properties of the amphibian mycobiome. Front Microbiol 2017; 8:2494 [View Article] [PubMed]
    [Google Scholar]
  74. Longo AV, Zamudio KR. Environmental fluctuations and host skin bacteria shift survival advantage between frogs and their fungal pathogen. ISME J 2017; 11:349–361 [View Article] [PubMed]
    [Google Scholar]
  75. Martínez-Ugalde E, Ávila-Akerberg V, González Martínez TM, Vázquez Trejo M, Zavala Hernández D et al. The skin microbiota of the axolotl Ambystoma altamirani is highly influenced by metamorphosis and seasonality but not by pathogen infection. Anim Microbiome 2022; 4:63 [View Article] [PubMed]
    [Google Scholar]
  76. Woodhams DC, Alford RA, Antwis RE, Archer H, Becker MH et al. Antifungal isolates database of amphibian skin‐associated bacteria and function against emerging fungal pathogens. Ecology 2015; 96:595 [View Article]
    [Google Scholar]
  77. Basanta MD, Anaya‐Morales SL, Martínez‐Ugalde E, González Martínez TM, Ávila‐Akerberg VD et al. Metamorphosis and seasonality are major determinants of chytrid infection in a paedomorphic salamander. Anim Conserv 2022; 26:340–354 [View Article]
    [Google Scholar]
  78. Krueger F. Trim Galore; 2023 https://github.com/FelixKrueger/TrimGalore
  79. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods 2012; 9:357–359 [View Article] [PubMed]
    [Google Scholar]
  80. Danecek P, Bonfield JK, Liddle J, Marshall J, Ohan V et al. Twelve years of SAMtools and BCFtools. Gigascience 2021; 10:giab008 [View Article] [PubMed]
    [Google Scholar]
  81. Li D, Liu C-M, Luo R, Sadakane K, Lam T-W. MEGAHIT: an ultra-fast single-node solution for large and complex metagenomics assembly via succinct de Bruijn graph. Bioinformatics 2015; 31:1674–1676 [View Article] [PubMed]
    [Google Scholar]
  82. Bushnell B. BBMap: A Fast, Accurate, Splice-Aware Aligner. Lawrence Berkeley National Laboratory; 2014 https://jgi.doe.gov/data-and-tools/software-tools/bbtools/bb-tools-user-guide/bbmap-guide/
  83. Hyatt D, Chen G-L, Locascio PF, Land ML, Larimer FW et al. Prodigal: prokaryotic gene recognition and translation initiation site identification. BMC Bioinformatics 2010; 11:119 [View Article] [PubMed]
    [Google Scholar]
  84. Fu L, Niu B, Zhu Z, Wu S, Li W. CD-HIT: accelerated for clustering the next-generation sequencing data. Bioinformatics 2012; 28:3150–3152 [View Article] [PubMed]
    [Google Scholar]
  85. Cantalapiedra CP, Hernández-Plaza A, Letunic I, Bork P, Huerta-Cepas J. eggNOG-mapper v2: functional annotation, orthology assignments, and domain prediction at the metagenomic scale. Mol Biol Evol 2021; 38:5825–5829 [View Article] [PubMed]
    [Google Scholar]
  86. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinforma Oxf Engl 2014; 30:923–930 [View Article] [PubMed]
    [Google Scholar]
  87. Xie F, Jin W, Si H, Yuan Y, Tao Y et al. An integrated gene catalog and over 10,000 metagenome-assembled genomes from the gastrointestinal microbiome of ruminants. Microbiome 2021; 9:137 [View Article] [PubMed]
    [Google Scholar]
  88. Oksanen J, Blanchet FG, Kindt R, Legendre P, Minchin P et al. Vegan: community ecology package. R package version 2.6-4; 2022 https://cran.rproject.org/web/packages/vegan/index.html
  89. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 2014; 15:550 [View Article] [PubMed]
    [Google Scholar]
  90. Bourgon R, Gentleman R, Huber W. Independent filtering increases detection power for high-throughput experiments. Proc Natl Acad Sci U S A 2010; 107:9546–9551 [View Article] [PubMed]
    [Google Scholar]
  91. Stephens M, Carbonetto P, Chaoxing D, Gerard D, Sun L et al. Methods for adaptive shrinkage, using empirical bayes. R package version 2.2-63; 2023 https://cran.r-project.org/web/packages/ashr/index.html
  92. Cevallos MA, Basanta MD, Bello-López E, Escobedo-Muñoz AS, González-Serrano FM et al. Genomic characterization of antifungal Acinetobacter bacteria isolated from the skin of the frogs Agalychnis callidryas and Craugastor fitzingeri. FEMS Microbiol Ecol 2022; 98:fiac126 [View Article] [PubMed]
    [Google Scholar]
  93. Bletz MC, Bunk B, Spröer C, Biwer P, Reiter S et al. Amphibian skin-associated Pigmentiphaga: genome sequence and occurrence across geography and hosts. PLoS One 2019; 14:e0223747 [View Article] [PubMed]
    [Google Scholar]
  94. Uritskiy GV, DiRuggiero J, Taylor J. MetaWRAP-a flexible pipeline for genome-resolved metagenomic data analysis. Microbiome 2018; 6:158 [View Article] [PubMed]
    [Google Scholar]
  95. Parks DH, Imelfort M, Skennerton CT, Hugenholtz P, Tyson GW. CheckM: assessing the quality of microbial genomes recovered from isolates, single cells, and metagenomes. Genome Res 2015; 25:1043–1055 [View Article] [PubMed]
    [Google Scholar]
  96. Olm MR, Brown CT, Brooks B, Banfield JF. dRep: a tool for fast and accurate genomic comparisons that enables improved genome recovery from metagenomes through de-replication. ISME J 2017; 11:2864–2868 [View Article] [PubMed]
    [Google Scholar]
  97. Chaumeil P-A, Mussig AJ, Hugenholtz P, Parks DH, Hancock J. GTDB-Tk: a toolkit to classify genomes with the Genome Taxonomy Database. Bioinformatics 2020; 36:1925–1927 [View Article] [PubMed]
    [Google Scholar]
  98. Nguyen L-T, Schmidt HA, von Haeseler A, Minh BQ. IQ-TREE: a fast and effective stochastic algorithm for estimating maximum-likelihood phylogenies. Mol Biol Evol 2015; 32:268–274 [View Article] [PubMed]
    [Google Scholar]
  99. Seemann T. Prokka: rapid prokaryotic genome annotation. Bioinforma Oxf Engl 2014; 30:2068–2069 [View Article] [PubMed]
    [Google Scholar]
  100. Medema MH, Blin K, Cimermancic P, de Jager V, Zakrzewski P et al. antiSMASH: rapid identification, annotation and analysis of secondary metabolite biosynthesis gene clusters in bacterial and fungal genome sequences. Nucleic Acids Res 2011; 39:W339–W346 [View Article] [PubMed]
    [Google Scholar]
  101. Kautsar SA, Blin K, Shaw S, Navarro-Muñoz JC, Terlouw BR et al. MIBiG 2.0: a repository for biosynthetic gene clusters of known function. Nucleic Acids Res 2020; 48:D454–D458 [View Article] [PubMed]
    [Google Scholar]
  102. Catenazzi A, Flechas SV, Burkart D, Hooven ND, Townsend J et al. Widespread elevational occurrence of antifungal bacteria in andean amphibians decimated by disease: a complex role for skin symbionts in defense against chytridiomycosis. Front Microbiol 2018; 9:465 [View Article] [PubMed]
    [Google Scholar]
  103. Kruger A. Frog skin microbiota vary with host species and environment but not chytrid infection. Front Microbiol 2020; 11:1330 [View Article] [PubMed]
    [Google Scholar]
  104. Ruthsatz K, Lyra ML, Lambertini C, Belasen AM, Jenkinson TS et al. Skin microbiome correlates with bioclimate and Batrachochytrium dendrobatidis infection intensity in Brazil’s Atlantic forest treefrogs. Sci Rep 2020; 10:22311 [View Article] [PubMed]
    [Google Scholar]
  105. Phillott AD, Grogan LF, Cashins SD, McDonald KR, Berger L et al. Chytridiomycosis and seasonal mortality of tropical stream-associated frogs 15 years after introduction of Batrachochytrium dendrobatidis. Conserv Biol 2013; 27:1058–1068 [View Article] [PubMed]
    [Google Scholar]
  106. Berger L, Speare R, Hines HB, Marantelli G, Hyatt AD et al. Effect of season and temperature on mortality in amphibians due to chytridiomycosis. Aust Vet J 2004; 82:434–439 [View Article] [PubMed]
    [Google Scholar]
  107. Savage AE, Sredl MJ, Zamudio KR. Disease dynamics vary spatially and temporally in a North American amphibian. Biol Conserv 2011; 144:1910–1915 [View Article]
    [Google Scholar]
  108. Daszak P, Cunningham AA, Hyatt AD. Infectious disease and amphibian population declines. Divers Distrib 2003; 9:141–150 [View Article]
    [Google Scholar]
  109. Sheets CN, Schmidt DR, Hurtado PJ, Byrne AQ, Rosenblum EB et al. Thermal performance curves of multiple isolates of Batrachochytrium dendrobatidis, a lethal pathogen of amphibians. Front Vet Sci 2021; 8:687084 [View Article] [PubMed]
    [Google Scholar]
  110. Sonn JM, Berman S, Richards-Zawacki CL. The Influence of temperature on Chytridiomycosis in vivo. Ecohealth 2017; 14:762–770 [View Article] [PubMed]
    [Google Scholar]
  111. Bradley PW, Brawner MD, Raffel TR, Rohr JR, Olson DH et al. Shifts in temperature influence how Batrachochytrium dendrobatidis infects amphibian larvae. PLoS One 2019; 14:e0222237 [View Article] [PubMed]
    [Google Scholar]
  112. Yasumiba K, Bell S, Alford R. Cell density effects of frog skin bacteria on their capacity to inhibit growth of the chytrid fungus, Batrachochytrium dendrobatidis. Microb Ecol 2016; 71:124–130 [View Article] [PubMed]
    [Google Scholar]
  113. Taga ME, Bassler BL. Chemical communication among bacteria. Proc Natl Acad Sci U S A 2003; 100:14549–14554 [View Article] [PubMed]
    [Google Scholar]
  114. Kueneman JG, Woodhams DC, Van Treuren W, Archer HM, Knight R et al. Inhibitory bacteria reduce fungi on early life stages of endangered Colorado boreal toads (Anaxyrus boreas). ISME J 2016; 10:934–944 [View Article] [PubMed]
    [Google Scholar]
  115. Brunetti AE, Bunk B, Lyra ML, Fuzo CA, Marani MM et al. Molecular basis of a bacterial-amphibian symbiosis revealed by comparative genomics, modeling, and functional testing. ISME J 2022; 16:788–800 [View Article] [PubMed]
    [Google Scholar]
  116. Lacombe-Harvey M-È, Brzezinski R, Beaulieu C. Chitinolytic functions in actinobacteria: ecology, enzymes, and evolution. Appl Microbiol Biotechnol 2018; 102:7219–7230 [View Article] [PubMed]
    [Google Scholar]
  117. Chet I, Ordentlich A, Shapira R, Oppenheim A. Mechanisms of biocontrol of soil-borne plant pathogens by Rhizobacteria. Plant Soil 1990; 129:85–92 [View Article]
    [Google Scholar]
  118. Dohnálek J, Dušková J, Tishchenko G, Kolenko P, Skálová T et al. Chitinase Chit62J4 essential for chitin processing by human microbiome bacterium Clostridium paraputrificum J4. Molecules 2021; 26:5978 [View Article] [PubMed]
    [Google Scholar]
  119. Adrangi S, Faramarzi MA. From bacteria to human: a journey into the world of chitinases. Biotechnol Adv 2013; 31:1786–1795 [View Article] [PubMed]
    [Google Scholar]
  120. Punja ZK, Zhang Y-Y. Plant chitinases and their roles in resistance to fungal diseases. J Nematol 1993; 25:526–540 [PubMed]
    [Google Scholar]
  121. Vaghela B, Vashi R, Rajput K, Joshi R. Plant chitinases and their role in plant defense: A comprehensive review. Enzyme Microb Technol 2022; 159:110055 [View Article] [PubMed]
    [Google Scholar]
  122. Zhao Y, Park R-D, Muzzarelli RAA. Chitin deacetylases: properties and applications. Mar Drugs 2010; 8:24–46 [View Article] [PubMed]
    [Google Scholar]
  123. Orlando M, Buchholz PCF, Lotti M, Pleiss J. The GH19 engineering database: sequence diversity, substrate scope, and evolution in glycoside hydrolase family 19. PLoS ONE 2021; 16:e0256817 [View Article] [PubMed]
    [Google Scholar]
  124. Chen Y, Zhou N, Chen X, Wei G, Zhang A et al. Characterization of a new multifunctional GH20 β-N-acetylglucosaminidase from Chitinibacter sp. GC72 and its application in converting chitin into N-acetyl glucosamine. Front Microbiol 2022; 13:874908 [View Article] [PubMed]
    [Google Scholar]
  125. Ren X-B, Dang Y-R, Liu S-S, Huang K-X, Qin Q-L et al. Identification and characterization of three chitinases with potential in direct conversion of crystalline chitin into N,N’-diacetylchitobiose. Mar Drugs 2022; 20:165 [View Article] [PubMed]
    [Google Scholar]
  126. Liao W, Liu P, Liao W, Miao L. Complete genome of the chitin-degrading bacterium, Paenibacillus xylanilyticus W4. Genome Biol Evol 2019; 11:3252–3255 [View Article] [PubMed]
    [Google Scholar]
  127. Arimori T, Kawamoto N, Shinya S, Okazaki N, Nakazawa M et al. Crystal structures of the catalytic domain of a novel glycohydrolase family 23 chitinase from Ralstonia sp. A-471 reveals A unique arrangement of the catalytic residues for inverting chitin hydrolysis. J Biol Chem 2013; 288:18696–18706 [View Article] [PubMed]
    [Google Scholar]
  128. Yamada Y, Kuzuyama T, Komatsu M, Shin-Ya K, Omura S et al. Terpene synthases are widely distributed in bacteria. Proc Natl Acad Sci U S A 2015; 112:857–862 [View Article] [PubMed]
    [Google Scholar]
  129. Gershenzon J, Dudareva N. The function of terpene natural products in the natural world. Nat Chem Biol 2007; 3:408–414 [View Article] [PubMed]
    [Google Scholar]
  130. Cane DE, Ikeda H. Exploration and mining of the bacterial terpenome. Acc Chem Res 2012; 45:463–472 [View Article] [PubMed]
    [Google Scholar]
  131. Toffolatti SL, Maddalena G, Passera A, Casati P, Bianco PA et al. 16 - Role of terpenes in plant defense to biotic stress. In Biocontrol Agents and Secondary Metabolites 2021
    [Google Scholar]
  132. Huang AC, Osbourn A. Plant terpenes that mediate below-ground interactions: prospects for bioengineering terpenoids for plant protection. Pest Manag Sci 2019; 75:2368–2377 [View Article] [PubMed]
    [Google Scholar]
  133. Pang Z, Chen J, Wang T, Gao C, Li Z et al. Linking plant secondary metabolites and plant microbiomes: a review. Front Plant Sci 2021; 12:621276 [View Article] [PubMed]
    [Google Scholar]
  134. Rudolf JD, Alsup TA, Xu B, Li Z. Bacterial terpenome. Nat Prod Rep 2021; 38:905–980 [View Article] [PubMed]
    [Google Scholar]
  135. Su R, Zhang S, Zhang X, Wang S, Zhang W. Neglected skin-associated microbial communities: a unique immune defense strategy of Bufo raddei under environmental heavy metal pollution. Environ Sci Pollut Res Int 2023; 30:22330–22342 [View Article] [PubMed]
    [Google Scholar]
  136. Wax N, Walke JB, Haak DC, Belden LK. Comparative genomics of bacteria from amphibian skin associated with inhibition of an amphibian fungal pathogen, Batrachochytrium dendrobatidis. PeerJ 2023; 11:e15714 [View Article] [PubMed]
    [Google Scholar]
  137. Brunetti AE, Lyra ML, Melo WGP, Andrade LE, Palacios-Rodríguez P et al. Symbiotic skin bacteria as a source for sex-specific scents in frogs. Proc Natl Acad Sci U S A 2019; 116:2124–2129 [View Article] [PubMed]
    [Google Scholar]
  138. Cao L, Do T, Link AJ. Mechanisms of action of ribosomally synthesized and posttranslationally modified peptides (RiPPs). J Ind Microbiol Biotechnol 2021; 48:kuab005 [View Article] [PubMed]
    [Google Scholar]
  139. Arnison PG, Bibb MJ, Bierbaum G, Bowers AA, Bugni TS et al. Ribosomally synthesized and post-translationally modified peptide natural products: overview and recommendations for a universal nomenclature. Nat Prod Rep 2013; 30:108–160 [View Article] [PubMed]
    [Google Scholar]
  140. Chikindas ML, Weeks R, Drider D, Chistyakov VA, Dicks LMT. Functions and emerging applications of bacteriocins. Curr Opin Biotechnol 2018; 49:23–28 [View Article] [PubMed]
    [Google Scholar]
  141. Niehus R, Oliveira NM, Li A, Fletcher AG, Foster KR. The evolution of strategy in bacterial warfare via the regulation of bacteriocins and antibiotics. eLife 2021; 10:e69756 [View Article] [PubMed]
    [Google Scholar]
  142. Heilbronner S, Krismer B, Brötz-Oesterhelt H, Peschel A. The microbiome-shaping roles of bacteriocins. Nat Rev Microbiol 2021; 19:726–739 [View Article] [PubMed]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/mgen/10.1099/mgen.0.001181
Loading
/content/journal/mgen/10.1099/mgen.0.001181
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF

Supplementary material 2

EXCEL

Supplementary material 3

EXCEL
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error