Submit a preprint

Latest recommendationsrsstwitter

IdTitle * Authors * Abstract * Picture * Thematic fields * RecommenderReviewersSubmission date
24 Feb 2023
article picture

MacSyFinder v2: Improved modelling and search engine to identify molecular systems in genomes

A unique and customizable approach for functionally annotating prokaryotic genomes

Recommended by ORCID_LOGO based on reviews by Kwee Boon Brandon Seah and Max Emil Schön

Macromolecular System Finder (MacSyFinder) v2 (Néron et al., 2023) is a newly updated approach for performing functional annotation of prokaryotic genomes (Abby et al., 2014). This tool parses an input file of protein sequences from a single genome (either ordered by genome location or unordered) and identifies the presence of specific cellular functions (referred to as “systems”). These systems are called based on two criteria: (1) that the "quorum" of a minimum set of core proteins involved is reached the “quorum” of a minimum set of core proteins being involved that are present, and (2) that the genes encoding these proteins are in the expected genomic organization (e.g., within the same order in an operon), when ordered data is provided. I believe the MacSyFinder approach represents an improvement over more commonly used methods exactly because it can incorporate such information on genomic organization, and also because it is more customizable.

Before properly appreciating these points, it is worth noting the norms and key challenges surrounding high-throughput functional annotation of prokaryotic genomes. Genome sequences are being added to online repositories at increasing rates, which has led to an enormous amount of bacterial genome diversity available to investigate (Altermann et al., 2022). A key aspect of understanding this diversity is the functional annotation step, which enables genes to be grouped into more biologically interpretable categories. For instance, gene calls can be mapped against existing Clusters of Orthologous Genes, which are themselves grouped into general categories such as ‘Transcription’ and ‘Lipid metabolism’ (Galperin et al., 2021).

This approach is valuable but is primarily used for global summaries of functional annotations within a genome: for example, it could be useful to know that a genome is particularly enriched for genes involved in lipid metabolism. However, knowing that a particular gene is involved in the general process of lipid metabolism is less likely to be actionable. In other words, the desired specificity of a gene’s functional annotation will depend on the exact question being investigated. There is no shortage of functional ontologies in genomics that can be applied for this purpose (Douglas and Langille, 2021), and researchers are often overwhelmed by the choice of which functional ontology to use. In this context, giving researchers the ability to precisely specify the gene families and operon structures they are interested in identifying across genomes provides useful control over what precise functions they are profiling. Of course, most researchers will lack the information and/or expertise to fully take advantage of MacSyFinder’s customizable features, but having this option for specialized purposes is valuable.

The other MacSyFinder feature that I find especially noteworthy is that it can incorporate genomic organization (e.g., of genes ordered in operons) when calling systems. This is a rare feature among commonly used tools for functional annotation and likely results in much higher specificity. As the authors note, this capability makes the co-occurrence of paralogs, and other divergent genes that share sequence similarity, to contribute less noise (i.e., they result in fewer false positive calls).

It is important to emphasize that these features are not new additions in MacSyFinder v2, but there are many other valuable changes. Most practically, this release is written in Python 3, rather than the obsolete Python 2.7, and was made more computationally efficient, which will enable MacSyFinder to be more widely used and more easily maintained moving forward. In addition, the search algorithm for analyzing individual proteins was fundamentally updated as well. The authors show that their improvements to the search algorithm result in an 8% and 20% increase in the number of identified calls for single and multi-locus secretion systems, respectively. Taken together, MacSyFinder v2 represents both practical and scientific improvements over the previous version, which will be of great value to the field. 

References

Abby SS, Néron B, Ménager H, Touchon M, Rocha EPC (2014) MacSyFinder: A Program to Mine Genomes for Molecular Systems with an Application to CRISPR-Cas Systems. PLOS ONE, 9, e110726. https://doi.org/10.1371/journal.pone.0110726

Altermann E, Tegetmeyer HE, Chanyi RM (2022) The evolution of bacterial genome assemblies - where do we need to go next? Microbiome Research Reports, 1, 15. https://doi.org/10.20517/mrr.2022.02

Douglas GM, Langille MGI (2021) A primer and discussion on DNA-based microbiome data and related bioinformatics analyses. Peer Community Journal, 1. https://doi.org/10.24072/pcjournal.2

Galperin MY, Wolf YI, Makarova KS, Vera Alvarez R, Landsman D, Koonin EV (2021) COG database update: focus on microbial diversity, model organisms, and widespread pathogens. Nucleic Acids Research, 49, D274–D281. https://doi.org/10.1093/nar/gkaa1018

Néron B, Denise R, Coluzzi C, Touchon M, Rocha EPC, Abby SS (2023) MacSyFinder v2: Improved modelling and search engine to identify molecular systems in genomes. bioRxiv, 2022.09.02.506364, ver. 2 peer-reviewed and recommended by Peer Community in Genomics. https://doi.org/10.1101/2022.09.02.506364

MacSyFinder v2: Improved modelling and search engine to identify molecular systems in genomesBertrand Néron, Rémi Denise, Charles Coluzzi, Marie Touchon, Eduardo P. C. Rocha, Sophie S. Abby<p style="text-align: justify;">Complex cellular functions are usually encoded by a set of genes in one or a few organized genetic loci in microbial genomes. Macromolecular System Finder (MacSyFinder) is a program that uses these properties to mod...Bacteria and archaea, Bioinformatics, Functional genomicsGavin Douglas Kwee Boon Brandon Seah, Max Emil Schön2022-09-09 10:30:31 View
24 Feb 2023
article picture

Performance and limitations of linkage-disequilibrium-based methods for inferring the genomic landscape of recombination and detecting hotspots: a simulation study

How to interpret the inference of recombination landscapes on methods based on linkage disequilibrium?

Recommended by ORCID_LOGO based on reviews by 2 anonymous reviewers

Data interpretation depends on previously established and validated tools, designed for a specific type of data. These methods, however, are usually based on simple models with validity subject to a set of theoretical parameterized conditions and data types. Accordingly, the tool developers provide the potential users with guidelines for data interpretations within the tools’ limitation. Nevertheless, once the methodology is accepted by the community, it is employed in a large variety of empirical studies outside of the method’s original scope or that typically depart from the standard models used for its design, thus potentially leading to the wrong interpretation of the results.

Numerous empirical studies inferred recombination rates across genomes, detecting hotspots of recombination and comparing related species (e.g., Shanfelter et al. 2019, Spence and Song 2019). These studies used indirect methodologies based on the signals that recombination left in the genome, such as linkage disequilibrium and the patterns of haplotype segregation (e.g.,Chan et al. 2012). The conclusions from these analyses have been used, for example, to interpret the evolution of the chromosomal structure or the evolution of recombination among closely related species.

Indirect methods have the advantage of collecting a large quantity of recombination events, and thus have a better resolution than direct methods (which only detect the few recombination events occurring at that time). On the other hand, indirect methods are affected by many different evolutionary events, such as demographic changes and selection. Indeed, the inference of recombination levels across the genome has not been studied accurately in non-standard conditions. Linkage disequilibrium is affected by several factors that can modify the recombination inference, such as demographic history, events of selection, population size, and mutation rate, but is also related to the size of the studied sample, and other technical parameters defined for each specific methodology.

Raynaud et al (2023) analyzed the reliability of the recombination rate inference when considering the violation of several standard assumptions (evolutionary and methodological) in one of the most popular families of methods based on LDhat (McVean et al. 2004), specifically its improved version, LDhelmet (Chan et al. 2012). These methods cover around 70 % of the studies that infer recombination rates. The authors used recombination maps, obtained from empirical studies on humans, and included hotspots, to perform a detailed simulation study of the capacity of this methodology to correctly infer the pattern of recombination and the location of these hotspots. Correlations between the real, and inferred values from simulations were obtained, as well as several rates, such as the true positive and false discovery rate to detect hotspots.

The authors of this work send a message of caution to researchers that are applying this methodology to interpret data from the inference of recombination landscapes and the location of hotspots. The inference of recombination landscapes and hotspots can differ considerably even in standard model conditions. In addition, demographic processes, like bottleneck or admixture, but also the level of population size and mutation rates, can substantially affect the estimation accuracy of the level of recombination and the location of hotspots. Indeed, the inference of the location of hotspots in simulated data with the same landscape, can be very imprecise when standard assumptions are violated or not considered. These effects may lead to incorrect interpretations, for example about the conservation of recombination maps between closely related species. Finally, Raynaud et al (2023) included a useful guide with advice on how to obtain accurate recombination estimations with methods based on linkage disequilibrium, also emphasizing the limitations of such approaches.

REFERENCES

Chan AH, Jenkins PA, Song YS (2012) Genome-Wide Fine-Scale Recombination Rate Variation in Drosophila melanogaster. PLOS Genetics, 8, e1003090. https://doi.org/10.1371/journal.pgen.1003090

McVean GAT, Myers SR, Hunt S, Deloukas P, Bentley DR, Donnelly P (2004) The Fine-Scale Structure of Recombination Rate Variation in the Human Genome. Science, 304, 581–584. https://doi.org/10.1126/science.1092500

Raynaud M, Gagnaire P-A, Galtier N (2023) Performance and limitations of linkage-disequilibrium-based methods for inferring the genomic landscape of recombination and detecting hotspots: a simulation study. bioRxiv, 2022.03.30.486352, ver. 2 peer-reviewed and recommended by Peer Community in Genomics. https://doi.org/10.1101/2022.03.30.486352

Spence JP, Song YS (2019) Inference and analysis of population-specific fine-scale recombination maps across 26 diverse human populations. Science Advances, 5, eaaw9206. https://doi.org/10.1126/sciadv.aaw9206

Performance and limitations of linkage-disequilibrium-based methods for inferring the genomic landscape of recombination and detecting hotspots: a simulation studyMarie Raynaud, Pierre-Alexandre Gagnaire, Nicolas Galtier<p style="text-align: justify;">Knowledge of recombination rate variation along the genome provides important insights into genome and phenotypic evolution. Population genomic approaches offer an attractive way to infer the population-scaled recom...Bioinformatics, Evolutionary genomics, Population genomicsSebastian Ernesto Ramos-Onsins2022-04-05 14:59:14 View
07 Feb 2023
article picture

RAREFAN: A webservice to identify REPINs and RAYTs in bacterial genomes

A workflow for studying enigmatic non-autonomous transposable elements across bacteria

Recommended by ORCID_LOGO based on reviews by Sophie Abby and 1 anonymous reviewer

Repetitive extragenic palindromic sequences (REPs) are common repetitive elements in bacterial genomes (Gilson et al., 1984; Stern et al., 1984). In 2011, Bertels and Rainey identified that REPs are overrepresented in pairs of inverted repeats, which likely form hairpin structures, that they referred to as “REP doublets forming hairpins” (REPINs). Based on bioinformatics analyses, they argued that REPINs are likely selfish elements that evolved from REPs flanking particular transposes (Bertels and Rainey, 2011). These transposases, so-called REP-associated tyrosine transposases (RAYTs), were known to be highly associated with the REP content in a genome and to have characteristic upstream and downstream flanking REPs (Nunvar et al., 2010). The flanking REPs likely enable RAYT transposition, and their horizontal replication is physically linked to this process. In contrast, Bertels and Rainey hypothesized that REPINs are selfish elements that are highly replicated due to the similarity in arrangement to these RAYT-flanking REPs, but independent of RAYT transposition and generally with no impact on bacterial fitness (Bertels and Rainey, 2011).

This last point was especially contentious, as REPINs are highly conserved within species (Bertels and Rainey, 2023), which is unusual for non-beneficial bacterial DNA (Mira et al., 2001). Bertels and Rainey have since refined their argument to be that REPINs must provide benefits to host cells, but that there are nonetheless signatures of intragenomic conflict in genomes associated with these elements (Bertels and Rainey, 2023). These signatures reflect the divergent levels of selections driving REPIN distribution: selection at the level of each DNA element and selection on each individual bacterium. I found this observation particularly interesting as I and my colleague recently argued that these divergent levels of selection, and the interaction between them, is key to understanding bacterial pangenome diversity (Douglas and Shapiro, 2021). REPINs could be an excellent system for investigating these levels of selection across bacteria more generally.

The problem is that REPINs have not been widely characterized in bacterial genomes, partially because no bioinformatic workflow has been available for this purpose. To address this problem, Fortmann-Grote et al. (2023) developed RAREFAN, which is a web server for identifying RAYTs and associated REPINs in a set of input genomes. The authors showcase their tool by applying it to 49 Stenotrophomonas maltophilia genomes and providing examples of how to identify and assess RAYT-REPIN hits. The workflow requires several manual steps, but nonetheless represents a straightforward and standardized approach. Overall, this workflow should enable RAYTs and REPINs to be identified across diverse bacterial species, which will facilitate further investigation into the mechanisms driving their maintenance and spread.

References

Bertels F, Rainey PB (2023) Ancient Darwinian replicators nested within eubacterial genomes. BioEssays, 45, 2200085. https://doi.org/10.1002/bies.202200085

Bertels F, Rainey PB (2011) Within-Genome Evolution of REPINs: a New Family of Miniature Mobile DNA in Bacteria. PLOS Genetics, 7, e1002132. https://doi.org/10.1371/journal.pgen.1002132

Douglas GM, Shapiro BJ (2021) Genic Selection Within Prokaryotic Pangenomes. Genome Biology and Evolution, 13, evab234. https://doi.org/10.1093/gbe/evab234

Fortmann-Grote C, Irmer J von, Bertels F (2023) RAREFAN: A webservice to identify REPINs and RAYTs in bacterial genomes. bioRxiv, 2022.05.22.493013, ver. 4 peer-reviewed and recommended by Peer Community in Genomics. https://doi.org/10.1101/2022.05.22.493013

Gilson E, Clément J m., Brutlag D, Hofnung M (1984) A family of dispersed repetitive extragenic palindromic DNA sequences in E. coli. The EMBO Journal, 3, 1417–1421. https://doi.org/10.1002/j.1460-2075.1984.tb01986.x

Mira A, Ochman H, Moran NA (2001) Deletional bias and the evolution of bacterial genomes. Trends in Genetics, 17, 589–596. https://doi.org/10.1016/S0168-9525(01)02447-7

Nunvar J, Huckova T, Licha I (2010) Identification and characterization of repetitive extragenic palindromes (REP)-associated tyrosine transposases: implications for REP evolution and dynamics in bacterial genomes. BMC Genomics, 11, 44. https://doi.org/10.1186/1471-2164-11-44

Stern MJ, Ames GF-L, Smith NH, Clare Robinson E, Higgins CF (1984) Repetitive extragenic palindromic sequences: A major component of the bacterial genome. Cell, 37, 1015–1026. https://doi.org/10.1016/0092-8674(84)90436-7

RAREFAN: A webservice to identify REPINs and RAYTs in bacterial genomesFrederic Bertels, Julia von Irmer, Carsten Fortmann-Grote<p style="text-align: justify;">Compared to eukaryotes, repetitive sequences are rare in bacterial genomes and usually do not persist for long. Yet, there is at least one class of persistent prokaryotic mobile genetic elements: REPINs. REPINs are ...Bacteria and archaea, Bioinformatics, Evolutionary genomics, Viruses and transposable elementsGavin Douglas2022-06-07 08:21:34 View
16 Dec 2022
article picture

Toeholder: a Software for Automated Design and In Silico Validation of Toehold Riboswitches

A novel approach for engineering biological systems by interfacing computer science with synthetic biology

Recommended by based on reviews by Wim Wranken and 1 anonymous reviewer

Biological systems depend on finely tuned interactions of their components. Thus, regulating these components is critical for the system's functionality. In prokaryotic cells, riboswitches are regulatory elements controlling transcription or translation. Riboswitches are RNA molecules that are usually located in the 5′-untranslated region of protein-coding genes. They generate secondary structures leading to the regulation of the expression of the downstream protein-coding gene (Kavita and Breaker, 2022). Riboswitches are very versatile and can bind a wide range of small molecules; in many cases, these are metabolic byproducts from the gene’s enzymatic or signaling pathway. Their versatility and abundance in many species make them attractive for synthetic biological circuits. One class that has been drawing the attention of synthetic biologists is toehold switches (Ekdahl et al., 2022; Green et al., 2014). These are single-stranded RNA molecules harboring the necessary elements for translation initiation of the downstream gene: a ribosome-binding site and a start codon. Conformation change of toehold switches is triggered by an RNA molecule, which enables translation.

To exploit the most out of toehold switches, automation of their design would be highly advantageous. Cisneros and colleagues (Cisneros et al., 2022) developed a tool, “Toeholder”, that automates the design of toehold switches and performs in silico tests to select switch candidates for a target gene. Toeholder is an open-source tool that provides a comprehensive and automated workflow for the design of toehold switches. While web tools have been developed for designing toehold switches (To et al., 2018), Toeholder represents an intriguing approach to engineering biological systems by coupling synthetic biology with computational biology. Using molecular dynamics simulations, it identified the positions in the toehold switch where hydrogen bonds fluctuate the most. Identifying these regions holds great potential for modifications when refining the design of the riboswitches. To be effective, toehold switches should provide a strong ON signal and a weak OFF signal in the presence or the absence of a target, respectively. Toeholder nicely ranks the candidate toehold switches based on experimental evidence that correlates with toehold performance (based on good ON/OFF ratios).

Riboswitches are highly appealing for a broad range of applications, including pharmaceutical and medical purposes (Blount and Breaker, 2006; Giarimoglou et al., 2022; Tickner and Farzan, 2021), thanks to their adaptability and inexpensiveness. The Toeholder tool developed by Cisneros and colleagues is expected to promote the implementation of toehold switches into these various applications.

References

Blount KF, Breaker RR (2006) Riboswitches as antibacterial drug targets. Nature Biotechnology, 24, 1558–1564. https://doi.org/10.1038/nbt1268

Cisneros AF, Rouleau FD, Bautista C, Lemieux P, Dumont-Leblond N, ULaval 2019 T iGEM (2022) Toeholder: a Software for Automated Design and In Silico Validation of Toehold Riboswitches. bioRxiv, 2021.11.09.467922, ver. 3 peer-reviewed and recommended by Peer Community in Genomics. https://doi.org/10.1101/2021.11.09.467922

Ekdahl AM, Rojano-Nisimura AM, Contreras LM (2022) Engineering Toehold-Mediated Switches for Native RNA Detection and Regulation in Bacteria. Journal of Molecular Biology, 434, 167689. https://doi.org/10.1016/j.jmb.2022.167689

Giarimoglou N, Kouvela A, Maniatis A, Papakyriakou A, Zhang J, Stamatopoulou V, Stathopoulos C (2022) A Riboswitch-Driven Era of New Antibacterials. Antibiotics, 11, 1243. https://doi.org/10.3390/antibiotics11091243

Green AA, Silver PA, Collins JJ, Yin P (2014) Toehold Switches: De-Novo-Designed Regulators of Gene Expression. Cell, 159, 925–939. https://doi.org/10.1016/j.cell.2014.10.002

Kavita K, Breaker RR (2022) Discovering riboswitches: the past and the future. Trends in Biochemical Sciences. https://doi.org/10.1016/j.tibs.2022.08.009

Tickner ZJ, Farzan M (2021) Riboswitches for Controlled Expression of Therapeutic Transgenes Delivered by Adeno-Associated Viral Vectors. Pharmaceuticals, 14, 554. https://doi.org/10.3390/ph14060554

To AC-Y, Chu DH-T, Wang AR, Li FC-Y, Chiu AW-O, Gao DY, Choi CHJ, Kong S-K, Chan T-F, Chan K-M, Yip KY (2018) A comprehensive web tool for toehold switch design. Bioinformatics, 34, 2862–2864. https://doi.org/10.1093/bioinformatics/bty216

Toeholder: a Software for Automated Design and In Silico Validation of Toehold RiboswitchesAngel F. Cisneros, François D. Rouleau, Carla Bautista, Pascale Lemieux, Nathan Dumont-Leblond<p>Abstract:&nbsp;Synthetic biology aims to engineer biological circuits, which often involve gene expression. A particularly promising group of regulatory elements are riboswitches because of their versatility with respect to their targets, but e...BioinformaticsSahar Melamed2022-02-16 14:40:13 View
15 Dec 2022
article picture

Botrytis cinerea strains infecting grapevine and tomato display contrasted repertoires of accessory chromosomes, transposons and small RNAs

Exploring genomic determinants of host specialization in Botrytis cinerea

Recommended by based on reviews by Cecile Lorrain and Thorsten Langner

The genomics era has pushed forward our understanding of fungal biology. Much progress has been made in unraveling new gene functions and pathways, as well as the evolution or adaptation of fungi to their hosts or environments through population studies (Hartmann et al. 2019; Gladieux et al. 2018). Closing gaps more systematically in draft genomes using the most recent long-read technologies now seems the new standard, even with fungal species presenting complex genome structures (e.g. large and highly repetitive dikaryotic genomes; Duan et al. 2022). Understanding the genomic dynamics underlying host specialization in phytopathogenic fungi is of utmost importance as it may open new avenues to combat diseases. A strong host specialization is commonly observed for biotrophic and hemi-biotrophic fungal species or for necrotrophic fungi with a narrow host range, whereas necrotrophic fungi with broad host range are considered generalists (Liang and Rollins, 2018; Newman and Derbyshire, 2020). However, some degrees of specialization towards given hosts have been reported in generalist fungi and the underlying mechanisms remain to be determined.

Botrytis cinerea is a polyphagous necrotrophic phytopathogen with a particularly wide host range and it is notably responsible for grey mould disease on many fruits, such as tomato and grapevine. Because of its importance as a plant pathogen, its relatively small genome size and its taxonomical position, it has been targeted for early genome sequencing and a first reference genome was provided in 2011 (Amselem et al. 2011). Other genomes were subsequently sequenced for other strains, and most importantly a gapless assembled version of the initial reference genome B05.10 was provided to the community (van Kan et al. 2017). This genomic resource has supported advances in various aspects of the biology of B. cinerea such as the production of specialized metabolites, which plays an important role in host-plant colonization, or more recently in the production of small RNAs which interfere with the host immune system, representing a new class of non-proteinaceous virulence effectors (Dalmais et al. 2011; Weiberg et al. 2013).

In the present study, Simon et al. (2022) use PacBio long-read sequencing for Sl3 and Vv3 strains, which represent genetic clusters in B. cinerea populations found on tomato and grapevine. The authors combined these complete and high-quality genome assemblies with the B05.10 reference genome and population sequencing data to perform a comparative genomic analysis of specialization towards the two host plants. Transposable elements generate genomic diversity due to their mobile and repetitive nature and they are of utmost importance in the evolution of fungi as they deeply reshape the genomic landscape (Lorrain et al. 2021). Accessory chromosomes are also known drivers of adaptation in fungi (Möller and Stukenbrock, 2017). Here, the authors identify several genomic features such as the presence of different sets of accessory chromosomes, the presence of differentiated repertoires of transposable elements, as well as related small RNAs in the tomato and grapevine populations, all of which may be involved in host specialization. Whereas core chromosomes are highly syntenic between strains, an accessory chromosome validated by pulse-field electrophoresis is specific of the strains isolated from grapevine. Particularly, they show that two particular retrotransposons are discriminant between the strains and that they allow the production of small RNAs that may act as effectors. The discriminant accessory chromosome of the Vv3 strain harbors one of the unraveled retrotransposons as well as new genes of yet unidentified function.

I recommend this article because it perfectly illustrates how efforts put into generating reference genomic sequences of higher quality can lead to new discoveries and allow to build strong hypotheses about biology and evolution in fungi. Also, the study combines an up-to-date genomics approach with a classical methodology such as pulse-field electrophoresis to validate the presence of accessory chromosomes. A major input of this investigation of the genomic determinants of B. cinerea is that it provides solid hints for further analysis of host-specialization at the population level in a broad-scale phytopathogenic fungus.

References

Amselem J, Cuomo CA, Kan JAL van, Viaud M, Benito EP, Couloux A, Coutinho PM, Vries RP de, Dyer PS, Fillinger S, Fournier E, Gout L, Hahn M, Kohn L, Lapalu N, Plummer KM, Pradier J-M, Quévillon E, Sharon A, Simon A, Have A ten, Tudzynski B, Tudzynski P, Wincker P, Andrew M, Anthouard V, Beever RE, Beffa R, Benoit I, Bouzid O, Brault B, Chen Z, Choquer M, Collémare J, Cotton P, Danchin EG, Silva CD, Gautier A, Giraud C, Giraud T, Gonzalez C, Grossetete S, Güldener U, Henrissat B, Howlett BJ, Kodira C, Kretschmer M, Lappartient A, Leroch M, Levis C, Mauceli E, Neuvéglise C, Oeser B, Pearson M, Poulain J, Poussereau N, Quesneville H, Rascle C, Schumacher J, Ségurens B, Sexton A, Silva E, Sirven C, Soanes DM, Talbot NJ, Templeton M, Yandava C, Yarden O, Zeng Q, Rollins JA, Lebrun M-H, Dickman M (2011) Genomic Analysis of the Necrotrophic Fungal Pathogens Sclerotinia sclerotiorum and Botrytis cinerea. PLOS Genetics, 7, e1002230. https://doi.org/10.1371/journal.pgen.1002230

Dalmais B, Schumacher J, Moraga J, Le Pêcheur P, Tudzynski B, Collado IG, Viaud M (2011) The Botrytis cinerea phytotoxin botcinic acid requires two polyketide synthases for production and has a redundant role in virulence with botrydial. Molecular Plant Pathology, 12, 564–579. https://doi.org/10.1111/j.1364-3703.2010.00692.x

Duan H, Jones AW, Hewitt T, Mackenzie A, Hu Y, Sharp A, Lewis D, Mago R, Upadhyaya NM, Rathjen JP, Stone EA, Schwessinger B, Figueroa M, Dodds PN, Periyannan S, Sperschneider J (2022) Physical separation of haplotypes in dikaryons allows benchmarking of phasing accuracy in Nanopore and HiFi assemblies with Hi-C data. Genome Biology, 23, 84. https://doi.org/10.1186/s13059-022-02658-2

Gladieux P, Condon B, Ravel S, Soanes D, Maciel JLN, Nhani A, Chen L, Terauchi R, Lebrun M-H, Tharreau D, Mitchell T, Pedley KF, Valent B, Talbot NJ, Farman M, Fournier E (2018) Gene Flow between Divergent Cereal- and Grass-Specific Lineages of the Rice Blast Fungus Magnaporthe oryzae. mBio, 9, e01219-17. https://doi.org/10.1128/mBio.01219-17

Hartmann FE, Rodríguez de la Vega RC, Carpentier F, Gladieux P, Cornille A, Hood ME, Giraud T (2019) Understanding Adaptation, Coevolution, Host Specialization, and Mating System in Castrating Anther-Smut Fungi by Combining Population and Comparative Genomics. Annual Review of Phytopathology, 57, 431–457. https://doi.org/10.1146/annurev-phyto-082718-095947

Liang X, Rollins JA (2018) Mechanisms of Broad Host Range Necrotrophic Pathogenesis in Sclerotinia sclerotiorum. Phytopathology®, 108, 1128–1140. https://doi.org/10.1094/PHYTO-06-18-0197-RVW

Lorrain C, Oggenfuss U, Croll D, Duplessis S, Stukenbrock E (2021) Transposable Elements in Fungi: Coevolution With the Host Genome Shapes, Genome Architecture, Plasticity and Adaptation. In: Encyclopedia of Mycology (eds Zaragoza Ó, Casadevall A), pp. 142–155. Elsevier, Oxford. https://doi.org/10.1016/B978-0-12-819990-9.00042-1

Möller M, Stukenbrock EH (2017) Evolution and genome architecture in fungal plant pathogens. Nature Reviews Microbiology, 15, 756–771. https://doi.org/10.1038/nrmicro.2017.76

Newman TE, Derbyshire MC (2020) The Evolutionary and Molecular Features of Broad Host-Range Necrotrophy in Plant Pathogenic Fungi. Frontiers in Plant Science, 11. https://doi.org/10.3389/fpls.2020.591733

Simon A, Mercier A, Gladieux P, Poinssot B, Walker A-S, Viaud M (2022) Botrytis cinerea strains infecting grapevine and tomato display contrasted repertoires of accessory chromosomes, transposons and small RNAs. bioRxiv, 2022.03.07.483234, ver. 4 peer-reviewed and recommended by Peer Community in Genomics. https://doi.org/10.1101/2022.03.07.483234

Van Kan JAL, Stassen JHM, Mosbach A, Van Der Lee TAJ, Faino L, Farmer AD, Papasotiriou DG, Zhou S, Seidl MF, Cottam E, Edel D, Hahn M, Schwartz DC, Dietrich RA, Widdison S, Scalliet G (2017) A gapless genome sequence of the fungus Botrytis cinerea. Molecular Plant Pathology, 18, 75–89. https://doi.org/10.1111/mpp.12384

Weiberg A, Wang M, Lin F-M, Zhao H, Zhang Z, Kaloshian I, Huang H-D, Jin H (2013) Fungal Small RNAs Suppress Plant Immunity by Hijacking Host RNA Interference Pathways. Science, 342, 118–123. https://doi.org/10.1126/science.1239705

Botrytis cinerea strains infecting grapevine and tomato display contrasted repertoires of accessory chromosomes, transposons and small RNAsAdeline Simon, Alex Mercier, Pierre Gladieux, Benoit Poinssot, Anne-Sophie Walker, Muriel Viaud<p style="text-align: justify;">The fungus <em>Botrytis cinerea</em> is a polyphagous pathogen that encompasses multiple host-specialized lineages. While several secreted proteins, secondary metabolites and retrotransposons-derived small RNAs have...Fungi, Structural genomics, Viruses and transposable elementsSebastien Duplessis Cecile Lorrain, Thorsten Langner2022-03-15 11:15:48 View
25 Nov 2022
article picture

Phenotypic and transcriptomic analyses reveal major differences between apple and pear scab nonhost resistance

Apples and pears: two closely related species with differences in scab nonhost resistance

Recommended by based on reviews by 3 anonymous reviewers

Nonhost resistance is a common form of disease resistance exhibited by plants against microorganisms that are pathogenic to other plant species [1]. Apples and pears are two closely related species belonging to Rosaceae family, both affected by scab disease caused by fungal pathogens in the Venturia genus. These pathogens appear to be highly host-specific. While apples are nonhosts for Venturia pyrina, pears are nonhosts for Venturia inaequalis. To date, the molecular bases of scab nonhost resistance in apple and pear have not been elucidated.

This preprint by Vergne, et al (2022) [2] analyzed nonhost resistance symptoms in apple/V. pyrina and pear/V. inaequalis interactions as well as their transcriptomic responses. Interestingly, the author demonstrated that the nonhost apple/V. pyrina interaction was almost symptomless while hypersensitive reactions were observed for pear/V. inaequalis interaction. The transcriptomic analyses also revealed a number of differentially expressed genes (DEGs) that corresponded to the severity of the interactions, with very few DEGs observed during the apple/V. pyrina interaction and a much higher number of DEGs during the pear/V. inaequalis interaction.

This type of reciprocal host-pathogen interaction study is valuable in gaining new insights into how plants interact with microorganisms that are potential pathogens in related species. A few processes appeared to be involved in the pear resistance against the nonhost pathogen V. inaequalis at the transcriptomic level, such as stomata closure, modification of cell wall and production of secondary metabolites as well as phenylpropanoids. Based on the transcriptomics changes during the nonhost interaction, the author compared the responses to those of host-pathogen interactions and revealed some interesting findings. They proposed a series of cascading effects in pear induced by the presence of V. inaequalis, which I believe helps shed some light on the basic mechanism for nonhost resistance.

I am recommending this study because it provides valuable information that will strengthen our understanding of nonhost resistance in the Rosaceae family and other plant species. The knowledge gained here may be applied to genetically engineer plants for a broader resistance against a number of pathogens in the future.​

References

1. Senthil-Kumar M, Mysore KS (2013) Nonhost Resistance Against Bacterial Pathogens: Retrospectives and Prospects. Annual Review of Phytopathology, 51, 407–427. https://doi.org/10.1146/annurev-phyto-082712-102319

2. Vergne E, Chevreau E, Ravon E, Gaillard S, Pelletier S, Bahut M, Perchepied L (2022) Phenotypic and transcriptomic analyses reveal major differences between apple and pear scab nonhost resistance. bioRxiv, 2021.06.01.446506, ver. 4 peer-reviewed and recommended by Peer Community in Genomics. https://doi.org/10.1101/2021.06.01.446506

Phenotypic and transcriptomic analyses reveal major differences between apple and pear scab nonhost resistanceE. Vergne, E. Chevreau, E. Ravon, S. Gaillard, S. Pelletier, M. Bahut, L. Perchepied<p style="text-align: justify;"><strong>Background. </strong>Nonhost resistance is the outcome of most plant/pathogen interactions, but it has rarely been described in Rosaceous fruit species. Apple (<em>Malus x domestica</em> Borkh.) have a nonho...Functional genomics, PlantsWirulda Pootakham Jessica Soyer, Anonymous2022-05-13 15:06:08 View
08 Nov 2022
article picture

Somatic mutation detection: a critical evaluation through simulations and reanalyses in oaks

How to best call the somatic mosaic tree?

Recommended by based on reviews by 2 anonymous reviewers

Any multicellular organism is a molecular mosaic with some somatic mutations accumulated between cell lineages. Big long-lived trees have nourished this imaginary of a somatic mosaic tree, from the observation of spectacular phenotypic mosaics and also because somatic mutations are expected to potentially be passed on to gametes in plants (review in Schoen and Schultz 2019). The lower cost of genome sequencing now offers the opportunity to tackle the issue and identify somatic mutations in trees.

However, when it comes to characterizing this somatic mosaic from genome sequences, things become much more difficult than one would think in the first place. What separates cell lineages ontogenetically, in cell division number, or in time? How to sample clonal cell populations? How do somatic mutations distribute in a population of cells in an organ or an organ sample? Should they be fixed heterozygotes in the sample of cells sequenced or be polymorphic? Do we indeed expect somatic mutations to be fixed? How should we identify and count somatic mutations?

To date, the detection of somatic mutations has mostly been done with a single variant caller in a given study, and we have little perspective on how different callers provide similar or different results. Some studies have used standard SNP callers that assumed a somatic mutation is fixed at the heterozygous state in the sample of cells, with an expected allele coverage ratio of 0.5, and less have used cancer callers, designed to detect mutations in a fraction of the cells in the sample. However, standard SNP callers detect mutations that deviate from a balanced allelic coverage, and different cancer callers can have different characteristics that should affect their outcomes.

In order to tackle these issues, Schmitt et al. (2022) conducted an extensive simulation analysis to compare different variant callers. Then, they reanalyzed two large published datasets on pedunculate oak, Quercus robur.  The analysis of in silico somatic mutations allowed the authors to evaluate the performance of different variant callers as a function of the allelic fraction of somatic mutations and the sequencing depth. They found one of the seven callers to provide better and more robust calls for a broad set of allelic fractions and sequencing depths. The reanalysis of published datasets in oaks with the most effective cancer caller of the in silico analysis allowed them to identify numerous low-frequency mutations that were missed in the original studies.

I recommend the study of Schmitt et al. (2022) first because it shows the benefit of using cancer callers in the study of somatic mutations, whatever the allelic fraction you are interested in at the end. You can select fixed heterozygotes if this is your ultimate target, but cancer callers allow you to have in addition a valuable overview of the allelic fractions of somatic mutations in your sample, and most do as well as SNP callers for fixed heterozygous mutations. In addition, Schmitt et al. (2022) provide the pipelines that allow investigating in silico data that should correspond to a given study design, encouraging to compare different variant callers rather than arbitrarily going with only one. We can anticipate that the study of somatic mutations in non-model species will increasingly attract attention now that multiple tissues of the same individual can be sequenced at low cost, and the study of Schmitt et al. (2022) paves the way for questioning and choosing the best variant caller for the question one wants to address.

References

Schoen DJ, Schultz ST (2019) Somatic Mutation and Evolution in Plants. Annual Review of Ecology, Evolution, and Systematics, 50, 49–73. https://doi.org/10.1146/annurev-ecolsys-110218-024955

Schmitt S, Leroy T, Heuertz M, Tysklind N (2022) Somatic mutation detection: a critical evaluation through simulations and reanalyses in oaks. bioRxiv, 2021.10.11.462798. ver. 4 peer-reviewed and recommended by Peer Community in Genomics. https://doi.org/10.1101/2021.10.11.462798

Somatic mutation detection: a critical evaluation through simulations and reanalyses in oaksSylvain Schmitt, Thibault Leroy, Myriam Heuertz, Niklas Tysklind<p style="text-align: justify;">1. Mutation, the source of genetic diversity, is the raw material of evolution; however, the mutation process remains understudied, especially in plants. Using both a simulation and reanalysis framework, we set out ...Bioinformatics, PlantsNicolas BierneAnonymous, Anonymous2022-04-28 13:24:19 View
23 Sep 2022
article picture

MATEdb, a data repository of high-quality metazoan transcriptome assemblies to accelerate phylogenomic studies

MATEdb: a new phylogenomic-driven database for Metazoa

Recommended by ORCID_LOGO based on reviews by 2 anonymous reviewers

The development (and standardization) of high-throughput sequencing techniques has revolutionized evolutionary biology, to the point that we almost see as normal fine-detail studies of genome architecture evolution (Robert et al., 2022), adaptation to new habitats (Rahi et al., 2019), or the development of key evolutionary novelties (Hilgers et al., 2018), to name three examples. One of the fields that has benefited the most is phylogenomics, i.e. the use of genome-wide data for inferring the evolutionary relationships among organisms. Dealing with such amount of data, however, has come with important analytical and computational challenges. Likewise, although the steady generation of genomic data from virtually any organism opens exciting opportunities for comparative analyses, it also creates a sort of “information fog”, where it is hard to find the most appropriate and/or the higher quality data. I have personally experienced this not so long ago, when I had to spend several weeks selecting the most complete transcriptomes from several phyla, moving back and forth between the NCBI SRA repository and the relevant literature.

In an attempt to deal with this issue, some research labs have committed their time and resources to the generation of taxa- and topic-specific databases (Lathe et al., 2008), such as MolluscDB (Liu et al., 2021), focused on mollusk genomics, or EukProt (Richter et al., 2022), a protein repository representing the diversity of eukaryotes. A new database that promises to become an important resource in the near future is MATEdb (Fernández et al., 2022), a repository of high-quality genomic data from Metazoa. MATEdb has been developed from publicly available and newly generated transcriptomes and genomes, prioritizing quality over quantity. Upon download, the user has access to both raw data and the related datasets: assemblies, several quality metrics, the set of inferred protein-coding genes, and their annotation. Although it is clear to me that this repository has been created with phylogenomic analyses in mind, I see how it could be generalized to other related problems such as analyses of gene content or evolution of specific gene families. In my opinion, the main strengths of MATEdb are threefold:

  1. Rosa Fernández and her team have carefully scrutinized the genomic data available in several repositories to retrieve only the most complete transcriptomes and genomes, saving a lot of time in data mining to the user.
  2. These data have been analyzed to provide both the assembly and the set of protein-coding genes, easing the computational burden that usually accompanies these pipelines. Interestingly, all the data have been analyzed with the same software and parameters, facilitating comparisons among taxa.
  3. Genomic analysis can be intimidating, and even more for inexperienced users. That is particularly important when it comes to transcriptome and genome assembly because it has an effect in all downstream analyses. I believe that having access to already analyzed data softens this transition. The users can move forward on their research while they learn how to generate and analyze their data at their own pace.

On a negative note, I see two main drawbacks. First, as of today (September 16th, 2022) this database is in an early stage and it still needs to incorporate a lot of animal groups. This has been discussed during the revision process and the authors are already working on it, so it is only a matter of time until all major taxa are represented. Second, there is a scalability issue. In its current format it is not possible to select the taxa of interest and the full database has to be downloaded, which will become more and more difficult as it grows. Nonetheless, with the appropriate resources it would be easy to find a better solution. There are plenty of examples that could serve as inspiration, so I hope this does not become a big problem in the future.

Altogether, I and the researchers that participated in the revision process believe that MATEdb has the potential to become an important and valuable addition to the metazoan phylogenomics community. Personally, I wish it was available just a few months ago, it would have saved me so much time.

References

Fernández R, Tonzo V, Guerrero CS, Lozano-Fernandez J, Martínez-Redondo GI, Balart-García P, Aristide L, Eleftheriadi K, Vargas-Chávez C (2022) MATEdb, a data repository of high-quality metazoan transcriptome assemblies to accelerate phylogenomic studies. bioRxiv, 2022.07.18.500182, ver. 4 peer-reviewed and recommended by Peer Community in Genomics. https://doi.org/10.1101/2022.07.18.500182

Hilgers L, Hartmann S, Hofreiter M, von Rintelen T (2018) Novel Genes, Ancient Genes, and Gene Co-Option Contributed to the Genetic Basis of the Radula, a Molluscan Innovation. Molecular Biology and Evolution, 35, 1638–1652. https://doi.org/10.1093/molbev/msy052

Lathe W, Williams J, Mangan M, Karolchik, D (2008). Genomic data resources: challenges and promises. Nature Education, 1(3), 2.

Liu F, Li Y, Yu H, Zhang L, Hu J, Bao Z, Wang S (2021) MolluscDB: an integrated functional and evolutionary genomics database for the hyper-diverse animal phylum Mollusca. Nucleic Acids Research, 49, D988–D997. https://doi.org/10.1093/nar/gkaa918

Rahi ML, Mather PB, Ezaz T, Hurwood DA (2019) The Molecular Basis of Freshwater Adaptation in Prawns: Insights from Comparative Transcriptomics of Three Macrobrachium Species. Genome Biology and Evolution, 11, 1002–1018. https://doi.org/10.1093/gbe/evz045

Richter DJ, Berney C, Strassert JFH, Poh Y-P, Herman EK, Muñoz-Gómez SA, Wideman JG, Burki F, Vargas C de (2022) EukProt: A database of genome-scale predicted proteins across the diversity of eukaryotes. bioRxiv, 2020.06.30.180687, ver. 5 peer-reviewed and recommended by Peer Community in Genomics. https://doi.org/10.1101/2020.06.30.180687

Robert NSM, Sarigol F, Zimmermann B, Meyer A, Voolstra CR, Simakov O (2022) Emergence of distinct syntenic density regimes is associated with early metazoan genomic transitions. BMC Genomics, 23, 143. https://doi.org/10.1186/s12864-022-08304-2

MATEdb, a data repository of high-quality metazoan transcriptome assemblies to accelerate phylogenomic studiesRosa Fernandez, Vanina Tonzo, Carolina Simon Guerrero, Jesus Lozano-Fernandez, Gemma I Martinez-Redondo, Pau Balart-Garcia, Leandro Aristide, Klara Eleftheriadi, Carlos Vargas-Chavez<p style="text-align: justify;">With the advent of high throughput sequencing, the amount of genomic data available for animals (Metazoa) species has bloomed over the last decade, especially from transcriptomes due to lower sequencing costs and ea...Bioinformatics, Evolutionary genomics, Functional genomicsSamuel Abalde2022-07-20 07:30:39 View
15 Sep 2022
article picture

EukProt: A database of genome-scale predicted proteins across the diversity of eukaryotes

EukProt enables reproducible Eukaryota-wide protein sequence analyses

Recommended by ORCID_LOGO based on reviews by 2 anonymous reviewers

 Comparative genomics is a general approach for understanding how genomes differ, which can be considered from many angles. For instance, this approach can delineate how gene content varies across organisms, which can lead to novel hypotheses regarding what those organisms do. It also enables investigations into the sequence-level divergence of orthologous DNA, which can provide insight into how evolutionary forces differentially shape genome content and structure across lineages. 
 
Such comparisons are often restricted to protein-coding genes, as these are sensible units for assessing putative function and for identifying homologous matches in divergent genomes. Although information is lost by focusing only on the protein-coding portion of genomes, this simplifies analyses and has led to crucial findings in recent years. Perhaps most dramatically, analyses based on hundreds of orthologous proteins across microbial eukaryotes are fundamentally changing our understanding of the eukaryotic tree of life (Burki et al. 2020).
 
These and other topics are highlighted in a new pre-print from Dr. Daniel Richter and colleagues, which describes EukProt (Richter et al. 2022): a database containing protein sets from 993 eukaryotic species. The authors provide a BLAST portal for matching custom sequences against this database (https://evocellbio.com/eukprot/) and the entire database is available for download (https://doi.org/10.6084/m9.figshare.12417881.v3). They also provide a subset of their overall dataset, ‘The Comparative Set’, which contains only high-quality proteomes and is meant to maximize phylogenetic diversity.
 
There are two major advantages of EukProt:
 
   1. It will enable researchers to quickly compare proteomes and perform phylogenomic analyses, without needing the skills or the time commitment to aggregate and process these data. The authors make it clear that acquiring the raw protein sets was non-trivial, as they were distributed across a wide variety of online repositories (some of which are no longer accessible!).
 
    2. Analyses based on this database will be more reproducible and easily compared across studies than those based on custom-made databases for individual studies. This is because the EukProt authors followed FAIR principles (Wilkinson et al. 2016) when building their database, which is a set of guidelines for enhancing data reusability. So, for instance, each proteome has a unique identifier in EukProt, and all species are annotated in a unified taxonomic framework, which will aid in standardizing comparisons across studies.
 
The authors make it clear that there is still work to be done. For example, there is an uneven representation of proteomes across different eukaryotic lineages, which can only be addressed by further characterization of poorly studied lineages. In addition, the authors note that it would ultimately be best for the EukProt database to be integrated into an existing large-scale repository, like NCBI, which would help ensure that important eukaryotic diversity was not ignored. Nonetheless, EukProt represents an excellent example of how reproducible bioinformatics resources should be designed and should prove to be an extremely useful resource for the field.
 
References

Burki F, Roger AJ, Brown MW, Simpson AGB (2020) The New Tree of Eukaryotes. Trends in Ecology & Evolution, 35, 43–55. https://doi.org/10.1016/j.tree.2019.08.008

Richter DJ, Berney C, Strassert JFH, Poh Y-P, Herman EK, Muñoz-Gómez SA, Wideman JG, Burki F, Vargas C de (2022) EukProt: A database of genome-scale predicted proteins across the diversity of eukaryotes. bioRxiv, 2020.06.30.180687, ver. 5 peer-reviewed and recommended by Peer Community in Genomics. https://doi.org/10.1101/2020.06.30.180687

Wilkinson MD, Dumontier M, Aalbersberg IjJ, Appleton G, Axton M, Baak A, Blomberg N, Boiten J-W, da Silva Santos LB, Bourne PE, Bouwman J, Brookes AJ, Clark T, Crosas M, Dillo I, Dumon O, Edmunds S, Evelo CT, Finkers R, Gonzalez-Beltran A, Gray AJG, Groth P, Goble C, Grethe JS, Heringa J, ’t Hoen PAC, Hooft R, Kuhn T, Kok R, Kok J, Lusher SJ, Martone ME, Mons A, Packer AL, Persson B, Rocca-Serra P, Roos M, van Schaik R, Sansone S-A, Schultes E, Sengstag T, Slater T, Strawn G, Swertz MA, Thompson M, van der Lei J, van Mulligen E, Velterop J, Waagmeester A, Wittenburg P, Wolstencroft K, Zhao J, Mons B (2016) The FAIR Guiding Principles for scientific data management and stewardship. Scientific Data, 3, 160018. https://doi.org/10.1038/sdata.2016.18

EukProt: A database of genome-scale predicted proteins across the diversity of eukaryotesDaniel J. Richter, Cédric Berney, Jürgen F. H. Strassert, Yu-Ping Poh, Emily K. Herman, Sergio A. Muñoz-Gómez, Jeremy G. Wideman, Fabien Burki, Colomban de Vargas<p style="text-align: justify;">EukProt is a database of published and publicly available predicted protein sets selected to represent the breadth of eukaryotic diversity, currently including 993 species from all major supergroups as well as orpha...Bioinformatics, Evolutionary genomicsGavin Douglas2022-06-08 14:19:28 View
23 Aug 2022
article picture

A novel lineage of the Capra genus discovered in the Taurus Mountains of Turkey using ancient genomics

Goat ancient DNA analysis unveils a new lineage that may have hybridized with domestic goats

Recommended by based on reviews by Torsten Günther and 1 anonymous reviewer

The genomic analysis of ancient remains has revolutionized the study of the past over the last decade. On top of the discoveries related to human evolution, plant and animal archaeogenomics has been used to gain new insights into the domestication process and the dispersal of domestic forms.

In this study, Daly and colleagues analyse the genomic data from seven goat specimens from the Epipalaeolithic recovered from the Direkli Cave in the Taurus Mountains in southern Turkey. They also generate new genomic data from Capra lineages across the phylogeny, contributing to the availability of genomic resources for this genus. Analysis of the ancient remains is compared to modern genomic variability and sheds light on the complexity of the Tur wild Capra lineages and their relationship with domestic goats and their wild ancestors.

Authors find that during the Late Pleistocene in the Taurus Mountains wild goats from the Tur lineage, today restricted to the Caucasus region, were not rare and cohabited with Bezoar, the wild goats that are the ancestors of domestic goats. They identify the Direkli Cave specimens as a lineage separate from the 
West and East Caucasus Tur modern lineages. Also, analysis of the genomic data and mitochondrial haplotypes reveals hybridization between the Tur and the Bezoar wild lineages. Interestingly, authors also find an uneven amount of Tur ancestry among Neolithic domestic goats, with European domestic goats showing evidence of this ancient Tur ancestry, whereas Neolithic Iranian domestic goats do not, a pattern that is also observed in some modern European domestic goats.

A modified D statistic, Dex, is developed to examine the contribution of the ancient Tur lineage in domestic goats through time and space. Dex measures the relative degree of allele sharing, derived specifically in a selected genome or group of genomes, and may have some utility in genera with complex admixture histories or admixture from ghost lineages. Results confirm that Neolithic European goat had an excess of allele sharing with this ancient Tur lineage, something that is absent in contemporary goats eastwards or in modern goats.

Interspecific gene flow is not uncommon among mammals, but the case of Capra has the additional motivation of understanding the origins of the domestic species. This work uncovers an ancient Tur lineage that is different from the modern ones and is additionally found in another geographic area. Furthermore, evidence shows that this ancient lineage exhibits substantial amounts of allele sharing with the wild ancestor of the domestic goat, but also with the Neolithic Eurasian domestic goats, highlighting the complexity of the domestication process.

This work has also important implications in understanding the effect of over-hunting and habitat disruption during the Anthropocene on the evolution of the Capra genus. The availability of more ancient specimens and better coverage of the modern genomic variability can help quantifying the lineages that went lost and identify the causes of their extinction.

This work is limited by the current availability of whole genomes from modern Capra specimens, but pieces of evidence as well that an effort is needed to obtain more genomic data from ancient goats from different geographic ranges to determine to what extent these lineages contributed to goat domestication.

References

Daly KG, Arbuckle BS, Rossi C, Mattiangeli V, Lawlor PA, Mashkour M, Sauer E, Lesur J, Atici L, Cevdet CM and Bradley DG (2022) A novel lineage of the Capra genus discovered in the Taurus Mountains of Turkey using ancient genomics. bioRxiv, 2022.04.08.487619, ver. 5 peer-reviewed and recommended by Peer Community in Genomics. https://doi.org/10.1101/2022.04.08.487619

A novel lineage of the Capra genus discovered in the Taurus Mountains of Turkey using ancient genomicsKevin G. Daly, Benjamin S. Arbuckle, Conor Rossi, Valeria Mattiangeli, Phoebe A. Lawlor, Marjan Mashkour, Eberhard Sauer, Joséphine Lesur, Levent Atici, Cevdet Merih Erek, Daniel G. Bradley<p>Direkli Cave, located in the Taurus Mountains of southern Turkey, was occupied by Late Epipaleolithic hunters-gatherers for the seasonal hunting and processing of game including large numbers of wild goats. We report genomic data from new and p...Evolutionary genomics, Population genomics, VertebratesLaura Botigué2022-04-15 12:05:47 View