More stories

  • in

    Selection constrains lottery assembly in the microbiomes of closely related diatom species

    1.Armbrust EV. The life of diatoms in the world’s oceans. Nature. 2009;459:185–92.CAS 
    PubMed 

    Google Scholar 
    2.Bowler C, Allen AE, Badger JH, Grimwood J, Jabbari K, Kuo A, et al. The Phaeo- dactylum genome reveals the evolutionary history of diatom genomes. Nature. 2008;456:239–44.CAS 
    PubMed 

    Google Scholar 
    3.Amin SA, Parker MS, Armbrust EF. Interactions between Diatoms and Bacteria. Microbiol Mol Biol Rev. 2012;76:667–84.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    4.Cirri E, Pohnert G. Algae- bacteria interactions that balance the planktonic microbiome. New Phytologist. 2019;223:100–6.
    Google Scholar 
    5.Mühlenbruch M, Grossart H, Eigemann F, Voss M. Mini‐review: Phytoplankton‐ derived polysaccharides in the marine environment and their interactions with heterotrophic bacteria. Environ Microbiol. 2018;20:2671–85.PubMed 

    Google Scholar 
    6.Koedooder C, Stock W, Willems A, Mangelinckx S, de Troch M, Vyverman W, et al. Diatom-bacteria interactions modulate the composition and productivity of benthic diatom biofilms. Front Microbiol. 2019;10:1255.PubMed 
    PubMed Central 

    Google Scholar 
    7.Teeling H, Fuchs BM, Bennke CM, Krüger K, Chafee M, Kappelmann L, et al. Recurring patterns in bacterioplankton dynamics during coastal spring algae blooms. eLife. 2016;5:e11888.PubMed 
    PubMed Central 

    Google Scholar 
    8.von Scheibner M, Sommer U, Jürgens K. Tight coupling of glaciecola spp. and diatoms during cold-water phytoplankton spring blooms. Front Microbiol. 2017;8:27.
    Google Scholar 
    9.Zhang H, Hou F, Xie W, Wang K, Zhou X, Zhang D, et al. Interaction and assembly processes of abundant and rare microbial communities during a diatom bloom process. Environ Microbiol. 2020;22:1707–19.CAS 
    PubMed 

    Google Scholar 
    10.Amin SA, Hmelo LR, van Tol HM, Durham BP, Carlson LT, Heal KR, et al. Interaction and signalling between a cosmopolitan phytoplankton and associated bacteria. Nature. 2015;522:98–101.CAS 
    PubMed 

    Google Scholar 
    11.Bigalke A, Pohnert G. Algicidal bacteria trigger contrasting responses in model diatom communities of different composition. MicrobiologyOpen. 2019;8:e00818.PubMed 
    PubMed Central 

    Google Scholar 
    12.Meyer N, Pohnert G. Isolate-specific resistance to the algicidal bacterium Kordia algicida in the diatom Chaetoceros genus. Botanica Marina. 2019;62:527–35.CAS 

    Google Scholar 
    13.Sison-Mangus MP, Jiang S, Tran KN, Kudela RM. Host-specific adaptation governs the interaction of the marine diatom, Pseudo-nitzschia and their microbiota. ISME J. 2014;8:63–76. https://doi.org/10.1038/ismej.2013.138CAS 
    Article 
    PubMed 

    Google Scholar 
    14.Stock W, Blommaert L, de Troch M, Mangelinckx S, Willems A, Vyverman W, et al. Host specificity in diatom–bacteria interactions alleviates antagonistic effects. FEMS Microbiol Ecol. 2019;95:fiz171.CAS 
    PubMed 

    Google Scholar 
    15.Nemergut DR, Schmidt SK, Fukami T, O’Neill SP, Bilinski TM, Stanish LF, et al. Patterns and processes of microbial community assembly. Microbiol Mol Biol Rev. 2013;77:342–56.PubMed 
    PubMed Central 

    Google Scholar 
    16.Ahern OM, Whittaker KA, Williams TC, Hunt DE, Rynearson TA. Host genotype structures the microbiome of a globally dispersed marine phytoplankton. Proc Natl Acad Sci. 2021;118:e2105207118.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    17.Vega NM, Gore J. Stochastic assembly produces heterogeneous communities in the Caenorhabditis elegans intestine. PLoS Biol. 2017;15:e2000633.PubMed 
    PubMed Central 

    Google Scholar 
    18.Lazzaro BP, Fox GM. Host–microbe interactions: winning the colonization lottery. Curr Biol. 2017;27:R642–R644.CAS 
    PubMed 

    Google Scholar 
    19.Burke C, Steinberg P, Rusch D, Kjelleberg S, Thomas T. Bacterial community assembly based on functional genes rather than species. Proc Natl Acad Sci. 2011;108:14288–93.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    20.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.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    21.Shibl AA, Isaac A, Ochsenkühn MA, Cárdenas A, Fei C, Behringer G, et al. Diatom modulation of select bacteria through use of two unique secondary metabolites. Proc Natl Acad Sci. 2020;117:27445–55.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    22.Zhou J, Ning D. Stochastic community assembly: does it matter in microbial ecology? Microbiol Mol Biol Rev. 2017;81:e00002–17.PubMed 
    PubMed Central 

    Google Scholar 
    23.Behringer G, Ochsenkühn MA, Fei C, Fanning J, Koester JA, Amin SA. Bacterial communities of diatoms display strong conservation across strains and time. Front Microbiol. 2018;9:1–15.
    Google Scholar 
    24.Crenn K, Duffieux D, Jeanthon C. Bacterial Epibiotic Communities of Ubiquitous and Abundant Marine Diatoms Are Distinct in Short- and Long-Term Associa- tions. Front Microbiol. 2018;9:1–12.
    Google Scholar 
    25.Grossart HP, Levold F, Allgaier M, Simon M, Brinkhoff T. Marine diatom species harbour distinct bacterial communities. Environ Microbiol. 2005;7:860–73.CAS 
    PubMed 

    Google Scholar 
    26.Guannel ML, Horner-Devine MC, Rocap G. Bacterial community composition differs with species and toxigenicity of the diatom Pseudo-nitzschia. Aquatic Microbial Ecol. 2011;64:117–33.
    Google Scholar 
    27.Ajani PA, Kahlke T, Siboni N, Carney R, Murray SA, Seymour JR. The microbiome of the cosmopolitan diatom Leptocylindrus reveals significant spatial and temporal variability. Front Microbiol. 2018;9:1–12.
    Google Scholar 
    28.Kaczmarska I, Ehrman JM, Bates SS, Green DH, Léger C, Harris J. Diversity and distribution of epibiotic bacteria on Pseudo-nitzschia multiseries (Bacillar- iophyceae) in culture, and comparison with those on diatoms in native seawater. Harmful Algae. 2005;4:725–41.
    Google Scholar 
    29.Sapp M, Wichels A, Gerdts G. Impacts of cultivation of marine diatoms on the associated bacterial community. Appl Environ Microbiol. 2007;73:3117–20.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    30.Mönnich J, Tebben J, Bergemann J, Case R, Wohlrab S, Harder T. Niche-based assembly of bacterial consortia on the diatom Thalassiosira rotula is stable and reproducible. ISME J. 2020;14:1614–25.PubMed 
    PubMed Central 

    Google Scholar 
    31.Baker LJ, Kemp PF. Exploring bacteria-diatom associations using single-cell whole genome amplification. Aquatic Microbial Ecol. 2014;72:73–88.
    Google Scholar 
    32.Candela M, Biagi E, Maccaferri S, Turroni S, Brigidi P. Intestinal microbiota is a plastic factor responding to environmental changes. Trends Microbiol. 2012;20:385–91.CAS 
    PubMed 

    Google Scholar 
    33.Goh C, Vallejos DFV, Nicotra AB, Mathesius U. The Impact of Beneficial Plant-Associated Microbes on Plant Phenotypic Plasticity. J Chem Ecol. 2013;826–39.34.Vanormelingen P, Vanelslander B, Sato S, Gillard J, Trobajo R, Sabbe K, et al. Heterothallic sexual reproduction in the model diatom Cylindrotheca. Eur J Phycol. 2013;48:93–105.
    Google Scholar 
    35.Li H, Yang G, Sun Y, Wu S, Zhang X. Cylindrotheca closterium is a species complex as was evidenced by the variations of rbcL gene and SSU rDNA. J Ocean Univer China. 2007;6:167–74.
    Google Scholar 
    36.Stock W, Vanelslander B, Rüdiger F, Sabbe K, Vyverman W, Karsten U. Thermal niche differentiation in the benthic diatom Cylindrotheca closterium (Bacillar- iophyceae) complex. Front Microbiol. 2019;10:1395.PubMed 
    PubMed Central 

    Google Scholar 
    37.de Brouwer JFC, Wolfstein K, Ruddy GK, Jones TER, Stal LJ. Biogenic stabilization of intertidal sediments: the importance of extracellular polymeric substances produced by benthic diatoms. Micro Ecol. 2005;49:501–12.CAS 

    Google Scholar 
    38.Najdek M, Blažina M, Djakovac T, Kraus R. The role of the diatom Cylindrotheca closterium in a mucilage event in the northern Adriatic Sea: Coupling with high salinity water intrusions. J Plankton Res. 2005;27:851–62.
    Google Scholar 
    39.Eaton Jw, Moss B. The estimation of numbers and pigment content in epipelic algal populations. Limnol Oceanogr. 1966;11:584–95.
    Google Scholar 
    40.Anderson RA. (editor). Algal Culturing Techniques. Elsevier Academic Press; 2005.41.Muyzer G, de Waal EC, Uitterlinden AG. Profiling of complex microbial popula- tions by denaturing gradient gel electrophoresis analysis of polymerase chain reaction-amplified genes coding for 16S rRNA. Appl Environ Microbiol. 1993;59:695–700.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    42.Tytgat B, Verleyen E, Sweetlove M, D’hondt S, Clercx P, van Ranst E, et al. Bacterial community composition in relation to bedrock type and macrobiota in soils from the Sør Rondane Mountains, East Antarctica. FEMS Microbiol Ecol. 2016;92.43.D’Hondt AS, Stock W, Blommaert L, Moens T, Sabbe K. Nematodes stimulate biomass accumulation in a multispecies diatom biofilm. Marine Environ Res. 2018;140:78–89.
    Google Scholar 
    44.Zhang J, Kobert K, Flouri T, Stamatakis A. PEAR: A fast and accurate Illumina Paired-End reAd mergeR. Bioinformatics. 2014;30:614–20.CAS 
    PubMed 

    Google Scholar 
    45.Edgar RC. UPARSE: Highly accurate OTU sequences from microbial amplicon reads. Nat Methods. 2013;10:996–8.CAS 
    PubMed 

    Google Scholar 
    46.Murali A, Bhargava A, Wright ES. IDTAXA: a novel approach for accurate taxo- nomic classification of microbiome sequences. Microbiome. 2018;6:1–14.
    Google Scholar 
    47.Wright ES. Using DECIPHER v2. 0 to analyze big biological sequence data in R. R Journal. 2016;8.48.Quast C, Pruesse E, Yilmaz P, Gerken J, Schweer T, Yarza P, et al. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 2012;41:D590–D596.PubMed 
    PubMed Central 

    Google Scholar 
    49.Nawrocki EP. Structural RNA Homology Search and Alignment using Covariance Models. Washington University in St. Louis; 2009.50.Hoang DT, Chernomor O, von Haeseler A, Minh BQ, Vinh LS. UFBoot2: improving the ultrafast bootstrap approximation. Mol Biol Evol. 2018;35:518–22.CAS 
    PubMed 

    Google Scholar 
    51.Nguyen LT, Schmidt HA, von Haeseler A, Minh BQ. IQ-TREE: a fast and effective stochastic algorithm for estimating maximum-likelihood phylogenies. Mol Biol Evol. 2015;32:268–74.CAS 
    PubMed 

    Google Scholar 
    52.Vanelslander B, Créach V, Vanormelingen P, Ernst A, Chepurnov VA, Sahan E, et al. Ecological differentiation between sympatric pseudocryptic species in the estuarine benthic diatom Navicula phyllepta (Bacillariophyceae). J Phycol. 2009;45:1278–89.CAS 
    PubMed 

    Google Scholar 
    53.Borcard D, Legendre P. All-scale spatial analysis of ecological data by means of principal coordinates of neighbour matrices. Ecol Modelling. 2002;153:51–68.
    Google Scholar 
    54.Louca S, Parfrey LW, Doebeli M. Decoupling function and taxonomy in the global ocean microbiome. Science. 2016;353:1272–7.CAS 
    PubMed 

    Google Scholar 
    55.Schloss PD, Westcott SL, Ryabin T, Hall JR, Hartmann M, Hollister EB, et al. Introducing mothur: Open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl Environ Microbiol. 2009;75:7537–41.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    56.DeSantis TZ, Hugenholtz P, Larsen N, Rojas M, Brodie EL, Keller K, et al. Green- genes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Appl Environ Microbiol. 2006;72:5069–72.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    57.McDonald D, Price MN, Goodrich J, Nawrocki EP, Desantis TZ, Probst A, et al. An improved Greengenes taxonomy with explicit ranks for ecological and evolu- tionary analyses of bacteria and archaea. ISME J. 2012;6:610–8.CAS 
    PubMed 

    Google Scholar 
    58.Stone L, Roberts A. The checkerboard score and species distributions. Oecologia. 1990;85:74–9.PubMed 

    Google Scholar 
    59.Sloan WT, Lunn M, Woodcock S, Head IM, Nee S, Curtis TP. Quantifying the roles of immigration and chance in shaping prokaryote community structure. Environ Microbiol. 2006;8:732–40.PubMed 

    Google Scholar 
    60.Burns AR, Stephens WZ, Stagaman K, Wong S, Rawls JF, Guillemin K, et al. Con- tribution of neutral processes to the assembly of gut microbial communities in the zebrafish over host development. ISME J. 2016;10:655–64.CAS 
    PubMed 

    Google Scholar 
    61.Hill MO. Diversity and evenness: a unifying notation and its consequences. Ecology. 1973;54:427–32.
    Google Scholar 
    62.Chiarello M, Auguet JC, Bettarel Y, Bouvier C, Claverie T, Graham NAJ, et al. Skin microbiome of coral reef fish is highly variable and driven by host phylogeny and diet. Microbiome. 2018;6:1–14.
    Google Scholar 
    63.Easson CG, Thacker RW. Phylogenetic signal in the community structure of host- specific microbiomes of tropical marine sponges. Front Microbiol. 2014;5:1–11.
    Google Scholar 
    64.Swierts T, Cleary DFR, de Voogd NJ. Prokaryotic communities of Indo-Pacific giant barrel sponges are more strongly influenced by geography than host phylogeny. FEMS Microbiol Ecol. 2018;94:1–12.
    Google Scholar 
    65.Mazel F, Davis KM, Loudon A, Kwong WK. Is Host Filtering the Main Driver of Phylosymbiosis across the Tree of Life. Msystems. 2018;3:1–15.
    Google Scholar 
    66.Fu H, Uchimiya M, Gore J, Moran MA. Ecological drivers of bacterial community assembly in synthetic phycospheres. Proc Natl Acad Sci. 2020;117.7:3656–62.
    Google Scholar 
    67.Taylor JD, Cunliffe M. Coastal bacterioplankton community response to diatom- derived polysaccharide microgels. Environ Microbiol Rep. 2017;9:151–7.CAS 
    PubMed 

    Google Scholar 
    68.Becker JW, Berube PM, Follett CL, Waterbury JB, Chisholm SW, Delong EF, et al. Closely related phytoplankton species produce similar suites of dissolved organic matter. Front Microbiol. 2014;5:1–14.CAS 

    Google Scholar 
    69.Jackrel SL, Yang JW, Schmidt KC, Denef VJ. Host specificity of microbiome assembly and its fitness effects in phytoplankton. ISME J. 2021;15:774–88.PubMed 

    Google Scholar 
    70.Eigemann F, Hilt S, Salka I, Grossart HP. Bacterial community composition asso- ciated with freshwater algae: Species specificity vs. dependency on environ- mental conditions and source community. FEMS Microbiol Ecol. 2013;83:650–63.CAS 
    PubMed 

    Google Scholar 
    71.Barreto Filho MM, Walker M, Ashworth MP, Morris JJ. Structure and Long-Term Stability of the Microbiome in Diverse Diatom Cultures. Microbiol Spectr. 2021;9:e00269–21.PubMed Central 

    Google Scholar 
    72.Horner-Devine MC, Bohannan BJM. Phylogenetic clustering and overdispersion in bacterial communities. Ecology. 2006;87:S100–8.PubMed 

    Google Scholar 
    73.Zelezniak A, Andrejev S, Ponomarova O, Mende DR, Bork P, Patil KR. Metabolic dependencies drive species co-occurrence in diverse microbial communities. Proc Natl Acad Sci. 2015;112:6449–54.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    74.Goldford JE, Lu N, Bajić D, Estrela S, Tikhonov M, Sanchez-Gorostiaga A, et al. Emergent simplicity in microbial community assembly. Science. 2018;361:469–74.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    75.Li Y, Shipley B, Price JN, Dantas V, de L, Tamme R, et al. Habitat filtering deter- mines the functional niche occupancy of plant communities worldwide. J Ecol. 2018;106:1001–9.
    Google Scholar 
    76.Louca S, Jacques SMS, Pires APF, Leal JS, Srivastava DS, Parfrey LW, et al. High taxonomic variability despite stable functional structure across microbial com- munities. Nat Ecol Evol. 2016;1:0015.
    Google Scholar 
    77.Seymour JR, Amin SA, Raina JB, Stocker R. Zooming in on the phycosphere: the ecological interface for phytoplankton-bacteria relationships. Nat Microbiol. 2017;2:1–12.
    Google Scholar 
    78.Geng H, Belas R. Molecular mechanisms underlying Roseobacter–phytoplankton symbioses. Curr Opinion Biotechnol. 2010;21:332–8.CAS 

    Google Scholar 
    79.Christie-Oleza JA, Sousoni D, Lloyd M, Armengaud J, Scanlan DJ. Nutrient recy- cling facilitates long-term stability of marine microbial phototroph-heterotroph interactions. Nat Microbiol. 2017;2:1–10.
    Google Scholar 
    80.Edmundson SJ, Huesemann MH. The dark side of algae cultivation: characterizing night biomass loss in three photosynthetic algae, Chlorella sorokiniana, Nanno- chloropsis salina and Picochlorum sp. Algal Res. 2015;12:470–6.
    Google Scholar 
    81.Grossart H-P. Interactions between marine bacteria and axenic various conditions in the lab. Aquatic Microbial Ecol. 1999;19:1–11.
    Google Scholar  More

  • in

    Viral tag and grow: a scalable approach to capture and characterize infectious virus–host pairs

    Improving our understanding of “viral tagging” flow cytometric signalsVT is a deceptively simple idea whereby a mixture of natural viruses are labeled with a DNA-binding fluorescent dye and ‘bait’ hosts infected by these stained viruses can be detected with flow cytometry via the fluorescent shift of “viral-tagged cells” [38, 39] (Fig. 1A, B). These viral-tagged cells can then be sorted, and the viral DNA separated using isotopic fractionation (the DNA of the cultured host is pre-labeled with “heavy” DNA) to access the metagenomes of the viruses that were experimentally determined to have infected these cell types. However, in practice, VT has been only minimally adopted by the community [43], presumably because it requires costly equipment (a high-performance flow sorter) and diverse technical expertise (flow cytometry, phage biology, and bioinformatics), while lacking sufficient benchmarking. To the latter, we sought to use a cultured phage-host model system (Pseudoalteromonas strain H71, hereafter H71, and its specific myophage PSA-HM1, hereafter HM1) to systematically assess the impact of various multiplicities of infection (MOIs; the ratio of the number of virus particles to the number of target cells, [48]) on the resultant VT signals. Further, we sought to augment VT to add an “and grow” capability whereby scalable single-virus cultivation, characterization, and sequencing could be enabled (Fig. 1C).Fig. 1: Overview of viral tagging, and the variant developed here—viral tag and grow.A Viruses are labeled with a green fluorescent dye and then mixed with potential host bacteria. B Fluorescence detection of individual cells with fluorescently-labeled viruses (FLVs) by flow cytometer. The flow cytometry plot (side scatter or forward scatter versus green fluorescence) shows the expected locations of FLV-tagged (VTs) and nontagged cells (NTs), which are flow-cytometrically green positive and negative, respectively. C Single-cell sorting of VTs is followed by subsequent amplification of infectious viruses. Single VTs are sorted into a 96-well plate that contains host culture. Culture growth is monitored by measuring optical density (OD) over time. A decrease in the OD curve from VT-containing wells (relative to the phage-negative control) indicates cell lysis by progeny viruses produced from a single isolated VT cell.Full size imageTo gain a better understanding of the biology behind VT signatures, we examined how H71 interacts with HM1, a phage specific for this host, and HS8, a phage that does not adsorb to this host – both assayed via flow cytometry and microscopy (for details, see Methods and online protocol, https://www.protocols.io/view/viral-tagging-and-grow-a-scalable-approach-to-captbwutpewn?form=MY01SV&OCID=MY01SV). Briefly, phages were stained with SybrGold (fluoresces green upon blue-light excitation) and for microscopy, H71 cells were stained with DAPI (fluoresces blue upon blue-light excitation, 4′,6-diamidino-2-phenylindole), as previously described [39, 49]. Replicate cultures of stained cells were then mixed with fluorescently-labeled phages (either HM1 or HS8 in each treatment) at infective MOIs = 1, 2, and 4, then these infections were incubated for 10 min, and processed (centrifuged and resuspended; see Methods for details) three times to remove free phages (see Methods for details). For microscopy, the relative fraction of virus-tagged (VTs) and nontagged cells (NTs) was measured from the available cells up to ~500 cells for each sample. For flow cytometry, cell detection was optimized to minimize background noise [50], and negative controls consisted of stained and washed sheath buffer and filtered Q water samples, as previously described [39].Overall, the resulting VT experiments were robust and informative. First, our cell-only optimizations resulted in controls that were impeccably clean (see representative cytograms and gating counts in Fig. 2A–C and  Supplementary Fig. S1). Second, in “virus addition” treatments, the resultant VT signal was distinct for specific (HM1) versus nonspecific (HS8) phages. Specifically, adding HM1 at MOIs = 1, 2, and 4 corresponded to VT population shifts of an average of 25%, 50%, and 80%, respectively, while NT populations proportionally decreased (Fig. 2D, E, linear regression r2 = 0.98). In contrast, for all tested MOIs of the nonspecific HS8 phage, the shifted populations were negligible (range: ~1.0–1.9%) and uncorrelated (Supplementary Fig. S2A, B; r2 = 0.14).Fig. 2: Flow cytometric and microscopic analyses of Pseudoalteromonas-phage associations.A Hierarchical gating for detection of Pseudoalteromonas strain H71 (hereafter, H71) and its subpopulations of viral tagged (VTs) and nontagged cells (NTs). A parent gate was drawn on H71 cells using FSC vs. SSC (Fig. S1) and represented in two types of contour and dot plots (left and right in the top of the gray box, respectively). From this gate, green-positive (VT) and -negative (NT) populations were sub-gated in the green fluorescence vs. SSC (right, dot plot) and quantified as percentage fractions of a parent population (bar charts in the gray box). B, C Flow cytometric plots of sheath buffer only (B) and stained/washed sheath buffer without phages (C) (see Methods and Fig. S1). D Flow cytometric detections for H71 cells (~106/ml) that were incubated with fluorescently-labeled specific phage HM1 at MOIs of 1, 2, and 4, respectively (from left to right). E Linear regression relationships between the MOIs (x-axis) and the percentages (Y-axis) of flow cytometric VT (green) and NT (black) populations for phage HM1 at MOIs of 1, 2, and 4, respectively. R-square values are represented. F DAPI (4′,6-diamidino-2-phenylindole, blue)-stained H71 cells were mixed with fluorescent phages HM1 (SybrGold, green) at MOIs of 1, 2, and 4, respectively (Methods for details). Above, the merged images of phage-host mixtures (Additional images are shown in Figs. S4–7). Below, an enlarged view of four regions selected from the above images. Interpretations of virus-tagged cells, nontagged cells, and “free” viruses are represented in the results and discussion and methods, respectively. Arrows point to phages found on the margin of bacterial cells. Scale bar, 2 µm. Microscopic observations for nonspecific phage HS8-H71 are shown in Fig. S8. G Correlation between the MOI (x-axis) and the microscopic fractions (y-axis) of VTs (green) and NTs (black) for phage HM1 at MOIs of 1, 2, and 4, respectively. R-square value is shown. H Impact of cell physiology on viral tagging signals. H71 cells (~106/ml) in the early log, late log, and stationary phase were infected by phage HM1 at MOIs of 1 (Left) and 4 (Right), respectively. Percentages of tagged populations were measured at the time point after fluorescently-labeled HM1 were inoculated for 20 min at various MOIs followed by centrifugation and resuspension to remove free viruses (see Methods for details). Each test was done in duplicate (error bars show standard deviations).Full size imageDespite observing a strong linear correlation between MOI and %VT for HM1, it was surprising that even at high MOIs = 1, 2, and 4, the resultant population shifts were 1.2- to 2.5-fold less than expected from theory alone based on Poisson distribution (see Supplementary Fig. S3). To investigate this, we used microscopy to inspect for virus clumping, positioning relative to cell surfaces, and background noise. These results revealed spot-like green signals of various sizes outside of host cells, which we interpreted as free viruses, and this was true even (a) at these higher MOIs, and (b) despite centrifugation to remove free viruses following incubation (see Methods; Fig. 2F and  Supplementary Figs. S4–7). We suspect these unincorporated SYBR-stained particles are viral aggregates, possibly due to host cell parts and/or debris in the lysate [51,52,53] or tangling of phage tails [54]. Prior work has shown that these and other mechanisms that decrease the accessibility of viral particles to host receptors could reduce observed infectious particles [48].Our third key observation in these experiments rested with an improved understanding of the ‘signal shift’ between VT and NT populations in the flow cytogram across varied MOIs. Again, comfortably, increasing the MOI pushed VT signals toward higher fluorescence, with NTs decreasing proportionally (Fig. 2F). We posited that such increased “VT” signal could result from multiple phages adsorbing per cell. Indeed, microscopy visualization of ~500 single cells per treatment revealed that the number of detectable phages per infected cell increased proportionally to the MOI (Fig. 2F, G and  Supplementary Figs. S4–6). For example, of the tagged cells, few (~14%) cells exhibited multiple phages adsorbed at an MOI = 1, whereas those numbers increased drastically at MOIs = 2 and 4, where most (~55% and 67%) tagged cells exhibited multiple adsorbed phages per cell. As a negative control, we examined VT signals for a nonspecific phage, and this revealed that virtually all of the 545 single cells that were examined were nontagged (99.3%) even at an MOI = 10 (Supplementary Fig. S7). Presumably, the remaining ~0.7% of cells that appeared to have a phage adsorbed represent promiscuous, reversible binding to nonhost cells as is known to occur in other phage model systems [39]. Mechanistically, multiple phages can bind to a single host cell. For example, under very high-titer infection conditions (e.g., MOI = 100) phages can distribute over an entire cell surface [55], presumably accessing broadly distributed receptors [56]. Prior VT work has demonstrated strong VT signals under very high MOI (e.g., MOI = 1000) conditions [43], though no optimization experiments were presented to understand these patterns and the false positives that would result from free phages coincidently sorted (see further discussion later).Finally, we re-evaluated the impact of cell physiology (e.g., early, middle, and late log phase host growth) and adsorption time (e.g., 20 min intervals from 0 to 120 min) on Pseudoalteromonas VT signals—and did so at two MOIs = 1 and 4, respectively (Fig. 2H). At both MOIs tested, growth phase was seen to impact the VT signals, with late log phase cells showing the highest fluorescent shift for VT cells in contrast to signals that were reduced in early log phase cells and nearly absent from stationary phase cells (Fig. 2H). This finding is consistent with our prior optimizations with Pseudoalteromonas phage-host model systems [39]. However, we observed that VT signals were optimal at 20 min after adsorption (see Methods) and, rather than stay high as we had previously observed, these experiments revealed that the VT signals were reduced by nearly half at subsequent time points. Though conflicting with our prior work [39], these current experiments employ hierarchical gating (Supplementary Fig. S1; see Methods), which we feel more appropriately quantify these patterns. This is because we interpret the signal reduction to be due to the lysis of first-adsorbed tagged cells and/or the injection of fluorescent DNA of the adsorbed virus(es) into cells as the latent period of phage HM1 for H71 cells under these conditions dictates [24]. Indeed, it has been reported that for phage lambda—E.coli system, the injection of fluorescent phage DNA followed by signal diffusion inside the cells decreased ~40% of the overall signal intensities of individual virus–host pairs [57].Together, though an extensive set of experiments, these findings are largely confirmatory with our prior work characterizing Pseudoalteromonas phages [39]. However, and critically, our prior work failed to rigorously investigate these phenomena with respect to their (i) flow cytogram population signatures, (ii) single-cell microscopy imaging, and (iii) hierarchically gated tagged-cell timing estimates. We hope that these additional clarifications here provide a better mechanistic understanding of VT signals, and encourage wider adoption of this promising high-throughput method to identify viruses that infect a particular host.Introducing VT and grow: VT coupled to plate-based cultivation assaysGiven this improved understanding of the VT signal, we next sought to expand VT to include an “and grow” capability to scalably capture and characterize viruses linked to hosts (conceptually presented in Fig. 1C). Pragmatically, this should also help resolve long-standing questions of (i) what fraction of VT cells lead to productive infections (i.e., does adsorption equal infection?, [45]), and (ii) whether sample processing (e.g., laser detection, sheath fluid growth inhibition [37, 58]) or cell density effects resulting from single-cell sorts [59, 60] would prohibit downstream growth assays.To this end, we used the Pseudoalteromonas-virus HM1 model system to optimize sorting and growth conditions. Specifically, we wondered how many cells from sorted populations would be required to observe lysis (both dynamically, and terminally) under various MOI conditions. To test this, viral-tagged cells (the “VT” treatment) or nontagged cells (the “NT” treatment) were sorted into individual wells of a 96-well plate containing growth medium; fresh host cells were added, and growth-lysis curves were established by measuring optical density (OD) over time (see Methods). Treatment variables included the number of cells sorted (n = 1, 3, or 9) and infection conditions (MOI = 1 or 4), while controls included (i) NT cells to control for false-positive culture lyses by free viruses coincidently sorted with target cells, and (ii) sorting process controls against host cell lysis and growth in plates consisting of wells containing cultures with and without phage HM1, respectively. For all experiments, cells were infected during late-exponential phase for 10 min, followed by dilution to halt further infection, and centrifugation to remove free viruses (see Methods, [41]).We first analyzed the reduced-titer MOI = 1 infection. When only single cells were sorted, the growth curves from those wells as compared to those of phage-free controls, showed that more than half (56%; 20/36) of the VT wells with detectably reduced OD, whereas only a single NT well (8%; 1/12) showed such a decrease (Fig. 3A). This low rate of false-positive culture lysis in NT wells suggests that in most of the VT wells, progeny phages produced from an isolated parent VT—not free viruses―infect and lyse the host culture (For more details, see the burst size distribution of sorted single VTs below). Presumably, the 16 VT wells that did not lyse were due to one of the following: (i) reduced viability of isolated VTs through multiple steps of sample preparation or sorting with high sheath pressure [37, 58], (ii) possible reversible virus adsorption from the VT cell prior to well capture, and/or (iii) mis-diagnoses due to the weak fluorescent shift of singly-VT cells as is a known challenge in fluorescence-based cell sorting [58, 61].Fig. 3: Evaluation of viral growth assay under various infection conditions.Two liquid cultures of Pseudoalteromonas strain H71 (105/ml) in the late-logarithmic growth phase were infected by specific phage HM1 at MOIs of 1 and 4, respectively. From each infected culture, varying numbers of tagged (VT) and nontagged (NT) cells were sorted into individual wells of a 96-well plate containing growth medium followed by the addition of fresh host cells (104 cells per well). Positive and negative controls (host culture with HM1 at an MOI of 0.1 and without HM1, respectively) were included in each plate (see Methods for details). From top to bottom, left to right in panels (A) MOI = 1 and (B) MOI = 4, respectively, pie charts depict the percentages of lysed (yellow) and nonlysed (gray) wells from the total wells containing the given numbers (n = 1, 3, and 9) of isolated VTs and NTs. Culture lysis for VT- and NT-containing wells was determined by comparing their growth curves (next to each pie chart, black lines) to those of negative (red) and positive controls (blue). The X-axis indicates the OD590nm and the Y-axis, the time in hours.Full size imageTo assess the MOI = 1 infections further, we evaluated the data for wells containing more than 1 cell sorted to each well. This revealed that sorting 3 or 9 cells improved the fraction of wells lysed in the VT treatments to 88 and 100%, respectively, but this came at the cost of increased false positives in the NT treatment (pie charts in Fig. 3A). The latter is likely due to the same challenges described above of differentiating the NT from VT populations when signal intensity was relatively low. Given the 96-well plate format, these experiments demonstrate the ability to follow growth kinetics for each well (time course OD figures in Fig. 3A). This revealed that single VT cell sorts had delayed lysis relative to the multiple-cell sorts and hints at the power such kinetics data could provide for scalably characterizing new en masse captured phage isolates from field samples. Stepping back, however, it is promising that the number of sorted cells per well, for both VT and NT wells, was linearly proportional to the percentages of lysed wells (r2 = 0.73 and 0.99), respectively (Supplementary Fig. S8). This suggests a robustness and repeatability for these experiments.Beyond the fraction of the VT and NT wells displaying clear lysis, the kinetics of lysis—particularly for single-cell sorts—can be a valuable first read-out for variability in virus infection dynamics. To assess this in our dataset, we examined the kinetics of OD readings through 20 h (growth-lysis curves in Fig. 3A). Focusing on the 36 wells containing a single VT cell, 20 lysed (reported above), but their lysis kinetics drastically differed—some wells showed stepwise decreases after early increases in OD and the others a very low or no increase followed by the curve recovery. Similar lysis patterns have been observed in other phage-host systems, where host culture growth depended on phage concentration, with suppression of host cells increasing with higher phage titers and vice versa [62, 63]. Our observation of the well-to-well variation in culture lysis is likely due to different progeny production from isolated VT per well, relating to the stochasticity of viral infection [37, 64,65,66,67]. However, the stochastic infection alone cannot explain such diverse lysis patterns, given the random nature of diffusion and contact of progeny particles from infected cells to neighboring susceptible cells in the fluid (i.e., the host culture) [68, 69]. Either biological or physical infection process, or both, could impact varied lysis pattern. Further experiments are required to test this hypothesis (e.g., single-cell burst size assay, [37]; see below).Finally, given that flow cytometric population separation was critical for optimizing lysis success and that simply sorting more cells comes at the cost of increased false-positive lysis, we next explored the impact of increasing the per-cell fluorescent VT signal with MOI = 4 infections. Indeed, sorting from these better-resolved populations improved our per-well lysis results as all of the VT wells lysed, and this was the case whether sorting 1, 3, or 9 cells per well (pie charts in Fig. 3B). For the NT wells, false positives were less problematic, but they did remain a minor problem as some wells (4–8%) lysed, and this increased in the multiple-cell sorted wells. Though VT and NT populations are likely better resolved, thereby reducing false-positive lysis in the NT wells from the MOI = 1 infections, presumably the higher MOI infections lead to free viruses being coincidently co-sorted in the sort droplets. Notably, the kinetic read-outs (growth-lysis curves in Fig. 3B) were relatively invariable, possibly suggesting that the much higher number of viruses-per-cell in these infections obscured virus-to-virus variability in life history traits [66, 67, 70].Together, these experiments provide strong baseline data for assessing the impact of VT signal quality, MOIs, and growth data and hint that the approach may also open up new windows into variation in trait space across virus isolates.New biology enabled by viral tag and grow: a window into “viral individuality”?A major challenge in viral ecology is scaling from the handful of viruses that might be well characterized to the millions of virus types in an average seawater or field sample. While diversity surveys have come a long way (e.g., hundreds of thousands of viruses in a single study [23]), the pragmatic challenges of taking physiological measurements across many viral isolates leaves modeling efforts with very little empirical data on virus life history traits, severely bottlenecking the viruses brought into predictive models [71]. Further, microbiologists have revealed that even among “clonal” isolates, there can be remarkable phenotypic heterogeneity, or “microbial individuality” [72,73,74]; does the same exist for viruses? Hints that there is such “virus individuality” among DNA viruses, including phages, are emerging with data demonstrating variability in single-cell burst size (progeny per infected cell), with up to ~100-fold differences and these differences attributed to stochastic events such as variation in starting points in cell size, growth stage, and resources [37, 64,65,66].Of particular interest in understanding ‘virus individuality’ are recent single-cell analyses developed for a Synechococcus phage-host model system that revealed a wide range of burst sizes (from 2 to 200 infective viruses/cell) within a laboratory clonal isolate [37]. Methodologically, this approach sorts cells—infected or not—into wells (e.g., of a 96-well plate) and follows their infection dynamics. This has the benefit of assessing a single cell’s growth-lysis curve in each well. However, a drawback is that experiments are more conveniently done at high MOI conditions (e.g., an MOI = 3 was used) to get larger numbers of wells lysing among the randomly sorted cells (see Methods). Increasing MOI will lead to more virus-containing and, therefore, lysing wells, subsequently greatly increasing the number of cells with multiple viruses attached such that it will confound measurements of lysis dynamics since they will be a function of both virus-to-virus ‘individuality’ and an unknown, but variable per-cell MOI [70, 75].Inspired by this latter work, we sought to improve such single-cell growth-lysis assays in ways that might leverage the scalability of VT + Grow. For these experiments, we wanted to reduce the MOI (to MOI = 0.5) since theory predicts that most (77%) of the infected cells would be singly infected (Poisson distribution), but keep it high enough to have a reasonably separated VT cell population (see Methods). After cells and viruses were mixed, individual VT cells were sorted into different wells containing growth medium, plates were incubated to allow lysis of the single sorted VT cell, and the number of plaques per well were determined by pour plate plaque assays (Fig. 4A; see Methods for details). This operationally single-cell burst size assay showed a wide range of infective viruses per cell (2 to 397, X-axis) from a total of 72 individual cells assessed (Y-axis) (on average = 100; Fig. 4B), with similar average population burst sizes of 110 ± 15 [24]. Though a clonal virus isolate, these findings suggest, just as seen for cyanophages [37], that stochastic events must dictate the specific burst size for any given interaction. However, unlike the prior work, it is unlikely that cells with multiple viruses adsorbed any of this signal since such events should be much rarer at an MOI = 0.5 instead of MOI = 3. This suggests that these stochastic events are of a biological nature, which we posit might mechanistically result from the timing of initial virus–host interactions and/or cell-to-cell or virus-to-virus variation in nonheritable traits such as per-cell nutrient stores. If we interpret such infected cell variability as ecologically relevant variation in “virocells” (sensu [13, 76, 77]), then these findings open a window into “virus individuality” via a more scalable and controllable characterization approach than previously available.Fig. 4: Distribution of virus burst sizes per single viral-tagged cell.A Schematic overview of single-cell assay for viral burst size determination by viral tagging and grow. In the latent period of infection, single viral-tagged cells (VTs) were sorted by flow cytometer from Pseudoalteromonas sp. H71 cells infected by phage HM1 at an MOI of 0.5 (see Methods for details). Following sorting single VTs into different wells of the 96-well plate containing growth medium (MSM), the plate was incubated to allow for viral progenies to release from infected cells. The number of viruses produced per VT was then determined by the number of plaques per poured plate using the traditional plaque assay. B Distribution of viral burst size from individual tagged cells. The number of progeny viruses (X-axis) per cell (Y-axis) are represented in bins of 20, with the exception of the first bin excluding single plaques. The number (n) of individual tagged cells assessed is represented at the top right corner.Full size imageLimitations and future development opportunities for VT and GrowThough these efforts provide a more robust foundation for broadening the use of VT related methods, there remain challenges. First, researchers must be aware that VT is not a simple method, and its success depends on instrument calibration and ultraclean sample processing to establish maximally separated VT and NT populations (see the link below for details on flow cytometric setup and optimization). Second, sorting purity, particularly in field applications, will be challenged by suboptimal VT flow cytometric signatures, e.g., mis-identification of NT cells. Though this can be overcome with very high MOI infections (e.g., 1000 viruses per cell, [43]), two issues remain: (i) the effective MOIs cannot be measured in field samples (and thus, unknown), and (ii) at such high MOIs, the experiments will suffer from coincident sorting of free viruses that will increase false positives. Another factor that could affect sorting purity is nonviral DNA in the environmental sample, whether it is associated with bacterial cells or not, which could be coincidently sorted. It is thus necessary to ensure that prior to any VT work, environmental samples are properly processed or treated for the removal of nonviral genes and other materials (e.g., filtration and/or centrifugation). Fortunately, the “and grow” approach added to VT provides an additional screening step whereby false-negatives and false positives can be discerned via growth-lysis monitoring. Further, the “and grow” component, a plate-based assay, enables faster and more scalable lysis screening (e.g., 96-well format) than the time- and labor-intensive traditional plaque assay [62, 63]. Third, viral aggregates that alter the effective MOI infection conditions could lead to confounding results when comparing results across laboratories. Here, we invite efforts to find and optimize approaches to reduce viral aggregates (e.g., detergents, sonication, syringe pumping), and until viral aggregates are eliminated, to microscopically examine the state of free viruses in new sample types, particularly for outlier results. Fourth, the methods remain dependent upon a cultivable host, and though VT has been applied to multiple heterotroph and cyanobacterial phage-host pairs [39], two big unknowns remain: (i) how will the “and grow” processing impact growth of these strains, and (ii) will non-marine model systems be amenable to these approaches. The in-depth optimizations presented here for a Pseudoalteromonas phage-host model system serve a foundation for understanding other target virus–host pairs. To this end, we suggest deep investigation for any new model systems being studied, and as information becomes more broadly available, invite a community-standards and benchmarking approach to determine ideal setups for infectious conditions (e.g., growth curve, MOIs) and instrumental parameters. To facilitate this, we have established a VT forum on the Viral Ecology VERVE Net living protocols at protocols.io (below) as a way to empower and broadly engage researchers interested in these new methods and the many variants that could blossom from this base. Specifically, the details for viral and bacterial sample processing can be found at https://www.protocols.io/view/viral-tagging-and-grow-a-scalable-approach-to-capt-bwutpewn?form=MY01SV&OCID=MY01SV and for flow cytometric optimization at https://www.protocols.io/view/bd-influx-cell-sorter-start-up-and-shut-427down-for-v-bv8cn9sw. Both protocols provide additional notes for critical steps to improve methodological reproducibility and/or sensitivity, and particularly for the latter, it will be updated regularly to better optimize, calibrate, and standardize a flow cytometer. More

  • in

    Exploring rhizo-microbiome transplants as a tool for protective plant-microbiome manipulation

    1.Mendes R, Raaijmakers JM. Cross-kingdom similarities in microbiome functions. ISME J. 2015;9:1905–7.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    2.Berg G, Rybakova D, Fischer D, Cernava T, Vergès M-CC, Charles T, et al. Microbiome definition re-visited: old concepts and new challenges. Microbiome. 2020;8:103.PubMed 
    PubMed Central 

    Google Scholar 
    3.Hall AB, Tolonen AC, Xavier RJ. Human genetic variation and the gut microbiome in disease. Nat Rev Genet. 2017;18:690–9.CAS 
    PubMed 

    Google Scholar 
    4.Trivedi P, Leach JE, Tringe SG, Sa T, Singh BK. Plant-microbiome interactions: from community assembly to plant health. Nat Rev Microbiol. 2020;18:607–21.CAS 
    PubMed 

    Google Scholar 
    5.Ramírez-Puebla ST, Servín-Garcidueñas LE, Jiménez-Marín B, Bolaños LM, Rosenblueth M, Martínez J, et al. Gut and root microbiota commonalities. Appl Environ Microbiol. 2013;79:2–9.PubMed 
    PubMed Central 

    Google Scholar 
    6.Lu T, Ke M, Lavoie M, Jin Y, Fan X, Zhang Z, et al. Rhizosphere microorganisms can influence the timing of plant flowering. Microbiome. 2018;6:231.PubMed 
    PubMed Central 

    Google Scholar 
    7.Zhang J, Liu Y-X, Zhang N, Hu B, Jin T, Xu H, et al. NRT1.1B is associated with root microbiota composition and nitrogen use in field-grown rice. Nat Biotechnol. 2019;37:676–84.CAS 
    PubMed 

    Google Scholar 
    8.Kwak MJ, Kong HG, Choi K, Kwon SK, Song JY, Lee J, et al. Rhizosphere microbiome structure alters to enable wilt resistance in tomato. Nat Biotechnol. 2018;36:1100–9.CAS 

    Google Scholar 
    9.Li H, La S, Zhang X, Gao L, Tian Y. Salt-induced recruitment of specific root-associated bacterial consortium capable of enhancing plant adaptability to salt stress. ISME J. 2021;15:2865–82.CAS 
    PubMed 

    Google Scholar 
    10.Xu L, Dong Z, Chiniquy D, Pierroz G, Deng S, Gao C, et al. Genome-resolved metagenomics reveals role of iron metabolism in drought-induced rhizosphere microbiome dynamics. Nat Commun. 2021;12:3209.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    11.Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol. 2017;17:219–32.CAS 
    PubMed 

    Google Scholar 
    12.Lee SM, Kong HG, Song GC, Ryu CM. Disruption of Firmicutes and Actinobacteria abundance in tomato rhizosphere causes the incidence of bacterial wilt disease. ISME J. 2021;15:330–47.CAS 
    PubMed 

    Google Scholar 
    13.Stacy A, Andrade-Oliveira V, McCulloch JA, Hild B, Oh JH, Perez-Chaparro PJ, et al. Infection trains the host for microbiota-enhanced resistance to pathogens. Cell. 2021;184:615–.e17.CAS 
    PubMed 

    Google Scholar 
    14.Sanders ME, Merenstein DJ, Reid G, Gibson GR, Rastall RA. Probiotics and prebiotics in intestinal health and disease: from biology to the clinic. Nat Rev Gastroenterol Hepatol. 2019;16:605–16.PubMed 

    Google Scholar 
    15.Bhattacharyya PN, Jha DK. Plant growth-promoting rhizobacteria (PGPR): emergence in agriculture. World J Microbiol Biotechnol. 2012;28:1327–50.CAS 
    PubMed 

    Google Scholar 
    16.Bashan Y, de-Bashan LE, Prabhu SR, Hernandez J-P. Advances in plant growth-promoting bacterial inoculant technology: formulations and practical perspectives (1998–2013). Plant Soil. 2014;378:1–33.CAS 

    Google Scholar 
    17.Zmora N, Zilberman-Schapira G, Suez J, Mor U, Dori-Bachash M, Bashiardes S, et al. Personalized gut mucosal colonization resistance to empiric probiotics is associated with unique host and microbiome features. Cell. 2018;174:1388–.e21.CAS 

    Google Scholar 
    18.Kaakoush NO. Fecal transplants as a microbiome-based therapeutic. Curr Opin Microbiol. 2020;56:16–23.CAS 
    PubMed 

    Google Scholar 
    19.Kassam Z, Lee CH, Yuan Y, Hunt RH. Fecal microbiota transplantation for Clostridium difficile infection: systematic review and meta-analysis. Am J Gastroenterol. 2013;108:500–8.PubMed 

    Google Scholar 
    20.Baruch EN, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L, et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science. 2021;371:602–9.CAS 

    Google Scholar 
    21.Weller DM, Raaijmakers JM, Gardener BBM, Thomashow LS. Microbial populations responsible for specific soil suppressiveness to plant pathogens. Annu Rev Phytopathol. 2002;40:309–48.CAS 
    PubMed 

    Google Scholar 
    22.Gopal M, Gupta A, Thomas GV. Bespoke microbiome therapy to manage plant diseases. Front Microbiol. 2013;4:355.PubMed 
    PubMed Central 

    Google Scholar 
    23.Raaijmakers JM, Bonsall RF, Weller DM. Effect of population density of Pseudomonas fluorescens on production of 2,4-diacetylphloroglucinol in the rhizosphere of wheat. Phytopathology. 1999;89:470–5.CAS 
    PubMed 

    Google Scholar 
    24.Mazurier S, Corberand T, Lemanceau P, Raaijmakers JM. Phenazine antibiotics produced by fluorescent pseudomonads contribute to natural soil suppressiveness to Fusarium wilt. ISME J. 2009;3:977–91.CAS 
    PubMed 

    Google Scholar 
    25.Siddiqui ZA, Shakeel U. Screening of Bacillus isolates for potential biocontrol of the wilt disease complex of pigeon pea (Cajanus cajan) under greenhouse and small-scale field conditions. J Plant Pathol. 2007;89:179–83.
    Google Scholar 
    26.Yadav K, Damodaran T, Dutt K, Singh A, Muthukumar M, Rajan S, et al. Effective biocontrol of banana fusarium wilt tropical race 4 by a bacillus rhizobacteria strain with antagonistic secondary metabolites. Rhizosphere. 2021;18:100341.
    Google Scholar 
    27.Meng Q, Yin J, Rosenzweig N, Douches D, Hao JJ. Culture-based assessment of microbial communities in soil suppressive to potato common scab. Plant Dis. 2012;96:712–7.PubMed 

    Google Scholar 
    28.Carrión VJ, Cordovez V, Tyc O, Etalo DW, de Bruijn I, de Jager VCL, et al. Involvement of Burkholderiaceae and sulfurous volatiles in disease-suppressive soils. ISME J. 2018;12:2307–21.PubMed 
    PubMed Central 

    Google Scholar 
    29.Gómez Expósito R, de Bruijn I, Postma J, Raaijmakers JM. Current insights into the role of rhizosphere bacteria in disease suppressive soils. Front Microbiol. 2017;8:2529.PubMed 
    PubMed Central 

    Google Scholar 
    30.Raaijmakers JM, Mazzola M. Soil immune responses. Science. 2016;352:1392–3.CAS 

    Google Scholar 
    31.Bakker PAHM, Pieterse CMJ, de Jonge R, Berendsen RL. The soil-borne legacy. Cell. 2018;172:1178–80.CAS 
    PubMed 

    Google Scholar 
    32.Schlatter D, Kinkel L, Thomashow L, Weller D, Paulitz T. Disease suppressive soils: new insights from the soil microbiome. Phytopathology. 2017;107:1284–97.PubMed 

    Google Scholar 
    33.Kyselková M, Kopecký J, Frapolli M, Défago G, Ságová-Marecková M, Grundmann GL, et al. Comparison of rhizobacterial community composition in soil suppressive or conducive to tobacco black root rot disease. ISME J. 2009;3:1127–38.PubMed 

    Google Scholar 
    34.Rosenzweig N, Tiedje JM, Quensen JF, Meng Q, Hao JJ. Microbial communities associated with potato common scab-suppressive soil determined by pyrosequencing analyses. Plant Dis. 2012;96:718–25.PubMed 

    Google Scholar 
    35.Cha JY, Han S, Hong H-J, Cho H, Kim D, Kwon Y, et al. Microbial and biochemical basis of a Fusarium wilt-suppressive soil. ISME J. 2016;10:119–29.CAS 
    PubMed 

    Google Scholar 
    36.Liu X, Zhang S, Jiang Q, Bai Y, Shen G, Li S, et al. Using community analysis to explore bacterial indicators for disease suppression of tobacco bacterial wilt. Sci Rep. 2016;6:36773.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    37.Mendes R, Kruijt M, de Bruijn I, Dekkers E, van der Voort M, Schneider JHM, et al. Deciphering the rhizosphere microbiome for disease-suppressive bacteria. Science. 2011;332:1097–100.CAS 

    Google Scholar 
    38.Wei Z, Gu Y, Friman VP, Kowalchuk GA, Xu Y, Shen Q, et al. Initial soil microbiome composition and functioning predetermine future plant health. Sci Adv. 2019;5:eaaw0759.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    39.Rillig MC, Antonovics J, Caruso T, Lehmann A, Powell JR, Veresoglou SD, et al. Interchange of entire communities: microbial community coalescence. Trends Ecol Evol. 2015;30:470–6.PubMed 

    Google Scholar 
    40.Deng S, Caddell DF, Xu G, Dahlen L, Washington L, Yang J, et al. Genome wide association study reveals plant loci controlling heritability of the rhizosphere microbiome. ISME J. 2021;15:3181–94.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    41.Mendes LW, Mendes R, Raaijmakers JM, Tsai SM. Breeding for soil-borne pathogen resistance impacts active rhizosphere microbiome of common bean. ISME J. 2018;12:3038–42.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    42.Hu J, Wei Z, Kowalchuk GA, Xu Y, Shen Q, Jousset A. Rhizosphere microbiome functional diversity and pathogen invasion resistance build up during plant development. Environ Microbiol. 2020;22:5005–18.PubMed 

    Google Scholar 
    43.Schreiter S, Ding G-C, Heuer H, Neumann G, Sandmann M, Grosch R, et al. Effect of the soil type on the microbiome in the rhizosphere of field-grown lettuce. Front Microbiol. 2014;5:144.PubMed 
    PubMed Central 

    Google Scholar 
    44.Wei Z, Hu J, Gu Y, Yin S, Xu Y, Jousset A, et al. Ralstonia solanacearum pathogen disrupts bacterial rhizosphere microbiome during an invasion. Soil Biol Biochem. 2018;118:8–17.CAS 

    Google Scholar 
    45.Jiang G, Wei Z, Xu J, Chen H, Zhang Y, She X, et al. Bacterial wilt in China: History, current status, and future perspectives. Front Plant Sci. 2017;8:1549.PubMed 
    PubMed Central 

    Google Scholar 
    46.Manda RR, Addanki VA, Srivastava S. Bacterial wilt of solanaceous crops. Int J Chem Stud. 2020;8:1048–57.CAS 

    Google Scholar 
    47.Barik S, Reddy AC, Ponnam N, Kumari M, C AG, Reddy DCL, et al. Breeding for bacterial wilt resistance in eggplant (Solanum melongena L.): progress and prospects. Crop Prot. 2020;137:105270.CAS 

    Google Scholar 
    48.Wei Z, Yang X, Yin S, Shen Q, Ran W, Xu Y. Efficacy of Bacillus-fortified organic fertiliser in controlling bacterial wilt of tomato in the field. Appl Soil Ecol. 2011;48:152–9.
    Google Scholar 
    49.Park E-J, Lee S-D, Chung E-J, Lee M-H, Um H-Y, Murugaiyan S, et al. MicroTom – A model plant system to study bacterial wilt by Ralstonia solanacearum. Plant Pathol J. 2007;23:239–44.
    Google Scholar 
    50.Schandry N. A practical guide to visualization and statistical analysis of R. solanacearum infection data using R. Front Plant Sci. 2017;8:623.PubMed 
    PubMed Central 

    Google Scholar 
    51.Gu S, Wei Z, Shao Z, Friman VP, Cao K, Yang T, et al. Competition for iron drives phytopathogen control by natural rhizosphere microbiomes. Nat Microbiol. 2020;5:1002–10.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    52.Klindworth A, Pruesse E, Schweer T, Peplies J, Quast C, Horn M, et al. Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies. Nucleic Acids Res. 2013;41:e1.CAS 
    PubMed 

    Google Scholar 
    53.Cole JR, Wang Q, Fish JA, Chai B, McGarrell DM, Sun Y, et al. Ribosomal Database Project: data and tools for high throughput rRNA analysis. Nucleic Acids Res. 2014;42:D633–642.CAS 

    Google Scholar 
    54.Katoh K, Misawa K, Kuma K, Miyata T. MAFFT: a novel method for rapid multiple sequence alignment based on fast Fourier transform. Nucleic Acids Res. 2002;30:3059–66.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    55.Price MN, Dehal PS, Arkin AP. FastTree 2-approximately maximum-likelihood trees for large alignments. PLoS ONE. 2010;5:e9490.PubMed 
    PubMed Central 

    Google Scholar 
    56.Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, et al. QIIME allows analysis of high-throughput community sequencing data. Nat Methods. 2010;7:335–6.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    57.Edgar RC. Search and clustering orders of magnitude faster than BLAST. Bioinformatics. 2010;26:2460–1.CAS 

    Google Scholar 
    58.Dixon P. VEGAN, a package of R functions for community ecology. J Veg Sci. 2003;14:927–30.
    Google Scholar 
    59.Shenhav L, Thompson M, Joseph TA, Briscoe L, Furman O, Bogumil D, et al. FEAST: fast expectation-maximization for microbial source tracking. Nat Methods. 2019;16:627–32.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    60.Mendiburu F de. agricolae: Statistical procedures for agricultural research. R package version 1.3–5. 2021.61.Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550.PubMed 
    PubMed Central 

    Google Scholar 
    62.Robinson MD, McCarthy DJ, Smyth GK. edgeR: a bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–40.CAS 
    PubMed 

    Google Scholar 
    63.Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, et al. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12:R60.PubMed 
    PubMed Central 

    Google Scholar 
    64.Deng Y, Jiang Y-H, Yang Y, He Z, Luo F, Zhou J. Molecular ecological network analyses. BMC Bioinformatics. 2012;13:113.PubMed 
    PubMed Central 

    Google Scholar 
    65.Zhou J, Deng Y, Luo F, He Z, Tu Q, Zhi X. Functional molecular ecological networks. mBio. 2010;1:e00169–10.PubMed 
    PubMed Central 

    Google Scholar 
    66.Ma B, Wang Y, Ye S, Liu S, Stirling E, Gilbert JA, et al. Earth microbial co-occurrence network reveals interconnection pattern across microbiomes. Microbiome. 2020;8:82.PubMed 
    PubMed Central 

    Google Scholar 
    67.Kuntal BK, Chandrakar P, Sadhu S, Mande SS. ‘NetShift’: a methodology for understanding ‘driver microbes’ from healthy and disease microbiome datasets. ISME J. 2019;13:442–54.PubMed 

    Google Scholar 
    68.Douglas GM, Maffei VJ, Zaneveld JR, Yurgel SN, Brown JR, Taylor CM, et al. PICRUSt2 for prediction of metagenome functions. Nat Biotechnol. 2020;38:685–8.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    69.Choi K, Choi J, Lee PA, Roy N, Khan R, Lee HJ, et al. Alteration of bacterial wilt resistance in tomato plant by microbiota transplant. Front Plant Sci. 2020;11:1186.PubMed 
    PubMed Central 

    Google Scholar 
    70.Davar D, Dzutsev AK, McCulloch JA, Rodrigues RR, Chauvin J-M, Morrison RM, et al. Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science. 2021;371:595–602.CAS 

    Google Scholar 
    71.D’Haens GR, Jobin C. Fecal microbial transplantation for diseases beyond recurrent Clostridium difficile infection. Gastroenterology. 2019;157:624–36.PubMed 

    Google Scholar 
    72.Gough E, Shaikh H, Manges AR. Systematic review of intestinal microbiota transplantation (fecal bacteriotherapy) for recurrent Clostridium difficile infection. Clin Infect Dis. 2011;53:994–1002.PubMed 

    Google Scholar 
    73.Durack J, Lynch SV. The gut microbiome: relationships with disease and opportunities for therapy. J Exp Med. 2019;216:20–40.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    74.Danne C, Rolhion N, Sokol H. Recipient factors in faecal microbiota transplantation: one stool does not fit all. Nat Rev Gastroenterol Hepatol. 2021;18:503–13.PubMed 

    Google Scholar 
    75.Jiang G, Wang N, Zhang Y, Zhang Y, Yu J, Zhang Y, et al. The relative importance of soil moisture in predicting bacterial wilt disease occurrence. Soil Ecol Lett. 2021;3:356–66.76.Wei Z, Friman VP, Pommier T, Geisen S, Jousset A, Shen Q. Rhizosphere immunity: targeting the underground for sustainable plant health management. Front Agric Sci Eng. 2020;7:317–28.
    Google Scholar 
    77.Hu J, Wei Z, Friman VP, Gu SH, Wang X-F, Eisenhauer N, et al. Probiotic diversity enhances rhizosphere microbiome function and plant disease suppression. mBio. 2016;7:e01790–16.CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    78.Bakker PAHM, Doornbos RF, Zamioudis C, Berendsen RL, Pieterse CMJ. Induced systemic resistance and the rhizosphere microbiome. Plant Pathol J. 2013;29:136–43.PubMed 
    PubMed Central 

    Google Scholar 
    79.Wei Z, Yang T, Friman VP, Xu Y, Shen Q, Jousset A. Trophic network architecture of root-associated bacterial communities determines pathogen invasion and plant health. Nat Commun. 2015;6:8413.CAS 
    PubMed 

    Google Scholar 
    80.Li M, Wei Z, Wang J, Jousset A, Friman V-P, Xu Y, et al. Facilitation promotes invasions in plant-associated microbial communities. Ecol Lett. 2019;22:149–58.PubMed 

    Google Scholar 
    81.Mendes LW, Raaijmakers JM, de Hollander M, Mendes R, Tsai SM. Influence of resistance breeding in common bean on rhizosphere microbiome composition and function. ISME J. 2018;12:212–24.PubMed 

    Google Scholar 
    82.Rosales PF, Bordin GS, Gower AE, Moura S. Indole alkaloids: 2012 until now, highlighting the new chemical structures and biological activities. Fitoterapia. 2020;143:104558.CAS 
    PubMed 

    Google Scholar 
    83.Sarbu LG, Bahrin LG, Babii C, Stefan M, Birsa ML. Synthetic flavonoids with antimicrobial activity: a review. J Appl Microbiol. 2019;127:1282–90.CAS 
    PubMed 

    Google Scholar 
    84.Madadi E, Mazloum-Ravasan S, Yu JS, Ha JW, Hamishehkar H, Kim KH. Therapeutic application of betalains: a review. Plants. 2020;9:E1219.PubMed 

    Google Scholar 
    85.Ryan RP, Monchy S, Cardinale M, Taghavi S, Crossman L, Avison MB, et al. The versatility and adaptation of bacteria from the genus Stenotrophomonas. Nat Rev Microbiol. 2009;7:514–25.CAS 
    PubMed 

    Google Scholar 
    86.Kolton M, Erlacher A, Berg G, Cytryn E. The Flavobacterium genus in the plant holobiont: ecological, physiological, and applicative insights. In: Castro-Sowinski S, editor. Microbial models: from environmental to industrial sustainability. Singapore: Springer; 2016. p. 189–207.87.Haas D, Défago G. Biological control of soil-borne pathogens by fluorescent pseudomonads. Nat Rev Microbiol. 2005;3:307–19.CAS 
    PubMed 

    Google Scholar 
    88.Fira D, Dimkić I, Berić T, Lozo J, Stanković S. Biological control of plant pathogens by Bacillus species. J Biotechnol. 2018;285:44–55.CAS 
    PubMed 

    Google Scholar  More

  • in

    Livestock grazing impact differently on the functional diversity of dung beetles depending on the regional context in subtropical forests

    1.Herrero, M. et al. Livestock and the environment: What have we learned in the past decade?. Annu. Rev. Environ. Resour. 40, 177–202 (2015).
    Google Scholar 
    2.Robinson, T. P. et al. Mapping the global distribution of livestock. PLoS ONE 9, e96084 (2014).ADS 
    PubMed 
    PubMed Central 

    Google Scholar 
    3.Firbank, L. G., Petit, S., Smart, S., Blain, A. & Fuller, R. J. Assessing the impacts of agricultural intensification on biodiversity: A British perspective. Philos. Trans. R. Soc. B: Biol. Sci. 363, 777–787 (2007).
    Google Scholar 
    4.Laurance, W. F., Sayer, J. & Cassman, K. G. Agricultural expansion and its impacts on tropical nature. Trends Ecol. Evol. 29, 107–116 (2014).PubMed 

    Google Scholar 
    5.Steinfeld, H., de Haan, C. & Blackburn, H. Livestock—Environment Interactions 88 (WRENmedia, 1997).
    Google Scholar 
    6.Eldridge, D. J., Poore, A. G. B., Ruiz-Colmenero, M., Letnic, M. & Soliveres, S. Ecosystem structure, function, and composition in rangelands are negatively affected by livestock grazing. Ecol. Appl. 26, 1273–1283 (2016).PubMed 

    Google Scholar 
    7.Schieltz, J. M. & Rubenstein, D. I. Evidence based review: Positive versus negative effects of livestock grazing on wildlife. What do we really know?. Environ. Res. Lett. 11, 113003 (2016).ADS 

    Google Scholar 
    8.Cornwell, W. K. & Ackerly, D. D. Community assembly and shifts in plant trait distributions across an environmental gradient in coastal California. Ecol. Monogr. 79, 109–126 (2009).
    Google Scholar 
    9.Kraft, N. J. B. et al. Community assembly, coexistence and the environmental filtering metaphor. Funct. Ecol. 29, 592–599 (2015).
    Google Scholar 
    10.Keddy, P. A. Assembly and response rules: Two goals for predictive community ecology. J. Veg. Sci. 3, 157–164 (1992).
    Google Scholar 
    11.Pärtel, M., Zobel, M., Zobel, K., van der Maarel, E. & Partel, M. The species pool and its relation to species richness: Evidence from Estonian plant communities. Oikos 75, 111–117 (1996).
    Google Scholar 
    12.Temperton, V., Hobbs, R. J., Nuttle, T. & Halle, S. Assembly Rules and Restoration Ecology. Bridging the Gap Between Theory and Practice (Island Press, 2004).
    Google Scholar 
    13.Leibold, M. A. Similarity and local co-existence of species in regional biotas. Evol. Ecol. 12, 95–110 (1998).
    Google Scholar 
    14.Hortal, J. et al. Ice age climate, evolutionary constraints and diversity patterns of European dung beetles: Ice age determines European scarab diversity. Ecol. Lett. 14, 741–748 (2011).PubMed 

    Google Scholar 
    15.de Bello, F., Lepš, J. & Sebastià, M.-T. Variations in species and functional plant diversity along climatic and grazing gradients. Ecography 29, 801–810 (2006).
    Google Scholar 
    16.Reymond, A., Purcell, J., Cherix, D., Guisan, A. & Pellissier, L. Functional diversity decreases with temperature in high elevation ant fauna: Functional diversity in high elevation ant. Ecol. Entomol. 38, 364–373 (2013).
    Google Scholar 
    17.Safi, K. et al. Understanding global patterns of mammalian functional and phylogenetic diversity. Philos. Trans. R. Soc. B 366, 2536–2544 (2011).
    Google Scholar 
    18.Mason-Romo, E. D., Farías, A. A. & Ceballos, G. Two decades of climate driving the dynamics of functional and taxonomic diversity of a tropical small mammal community in western Mexico. PLoS ONE 12, e0189104 (2017).PubMed 
    PubMed Central 

    Google Scholar 
    19.Wen, Z. et al. Functional diversity overrides community-weighted mean traits in linking land-use intensity to hydrological ecosystem services. Sci. Total Environ. 682, 583–590 (2019).ADS 
    CAS 
    PubMed 

    Google Scholar 
    20Corbelli, J. M. et al. Integrating taxonomic, functional and phylogenetic beta diversities: Interactive effects with the biome and land use across taxa. PLoS ONE 10, e0126854 (2015).PubMed 
    PubMed Central 

    Google Scholar 
    21.Flynn, D. F. B. et al. Loss of functional diversity under land use intensification across multiple taxa. Ecol. Lett. 12, 22–33 (2009).PubMed 

    Google Scholar 
    22.Spector, S. Scarabaeine dung beetles (Coleoptera: Scarabaeidae: Scarabaeinae): An invertebrate focal taxon for biodiversity research and conservation. Coleopt. Bull. 60, 71–83 (2006).
    Google Scholar 
    23.Gardner, T. A. et al. The cost-effectiveness of biodiversity surveys in tropical forests: Cost-effectiveness of biodiversity surveys. Ecol. Lett. 11, 139–150 (2008).PubMed 

    Google Scholar 
    24.Mason, N. W. H., Mouillot, D., Lee, W. G. & Wilson, J. B. Functional richness, functional evenness and functional divergence: The primary components of functional diversity. Oikos 111, 112–118 (2005).
    Google Scholar 
    25.Villéger, S., Mason, N. W. H. & Mouillot, D. New multidimensional functional diversity indices for a multifaceted framework in functional ecology. Ecology 89, 2290–2301 (2008).PubMed 

    Google Scholar 
    26.Laliberté, E. & Legendre, P. A distance-based framework for measuring functional diversity from multiple traits. Ecology 91, 299–305 (2010).PubMed 

    Google Scholar 
    27.Audino, L. D., Louzada, J. & Comita, L. Dung beetles as indicators of tropical forest restoration success: Is it possible to recover species and functional diversity?. Biol. Cons. 169, 248–257 (2014).
    Google Scholar 
    28.Barragán, F., Moreno, C. E., Escobar, F., Halffter, G. & Navarrete, D. Negative impacts of human land use on dung beetle functional diversity. PLoS ONE 6, e17976 (2011).ADS 
    PubMed 
    PubMed Central 

    Google Scholar 
    29.Correa, C. M. A., Braga, R. F., Puker, A. & Korasaki, V. Patterns of taxonomic and functional diversity of dung beetles in a human-modified variegated landscape in Brazilian Cerrado. J. Insect Conserv. 23, 89–99 (2019).
    Google Scholar 
    30.Gómez-Cifuentes, A., Munevar, A., Gimenez, V. C., Gatti, M. G. & Zurita, G. A. Influence of land use on the taxonomic and functional diversity of dung beetles (Coleoptera: Scarabaeinae) in the southern Atlantic forest of Argentina. J. Insect Conserv. 21, 147–156 (2017).
    Google Scholar 
    31.Guerra Alonso, C. B., Zurita, G. A. & Bellocq, M. I. Dung beetles response to livestock management in three different regional contexts. Sci. Rep. 10, 3702 (2020).ADS 
    CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    32de Siqueira Neves, F. et al. Successional and seasonal changes in a community of dung beetles (Coleoptera: Scarabaeinae) in a Brazilian tropical dry forest. Nat. Conserv. 08, 160–164 (2010).
    Google Scholar 
    33Kottek, M., Grieser, J., Beck, C., Rudolf, B. & Rubel, F. World Map of the Köppen-Geiger climate classification updated. Meteorol. Z. 15, 259–263 (2006).
    Google Scholar 
    34.Brown, A. La situación ambiental Argentina 2005 (Fundación Vida Silvestre Argentina, 2006).
    Google Scholar 
    35.Larsen, T. H., Lopera, A. & Forsyth, A. Extreme trophic and habitat specialization by Peruvian dung beetles (Coleoptera: Scarabaeidae: Scarabaeinae). Coleopt. Bull. 60, 315–324 (2006).
    Google Scholar 
    36Vaz-de-Mello, F. Z. A Multilingual Key to the Genera and Subgenera of the Subfamily Scarabaeinae of the New World (Coleoptera: Scarabaeidae) (Magnolia Press, 2011).
    Google Scholar 
    37.Braun-Blanquet, J. Fitosociología [Phytosociology]. Bases para el estudio de las comunidades vegetales [Basis for the study of plant communities] 820 (Editorial H. Blume, 1979).
    Google Scholar 
    38.Fick, S. E. & Hijmans, R. J. WorldClim 2: New 1-km spatial resolution climate surfaces for global land areas. Int. J. Climatol. 37, 4302–4315 (2017).
    Google Scholar 
    39Scholtz, C. H., Davis, A. L. V. & Kryger, U. Evolutionary Biology and Conservation of Dung Beetles (Pensoft, 2009).
    Google Scholar 
    40.Simmons, L. W. & Ridsdill-Smith, J. Reproductive competition and its impact on the evolution and ecology of dung beetles. In Ecology and Evolution of Dung Beetles (eds Simmons, L. W. & Ridsdill-Smith, T. J.) 1–20 (Wiley, 2011). https://doi.org/10.1002/9781444342000.ch1.Chapter 

    Google Scholar 
    41.Vaz-de-Mello, F. Scarabaeidae in Catálogo Taxonômico da Fauna do Brasil. Catálogo Taxonômico da Fauna do Brasil. http://fauna.jbrj.gov.br/fauna/faunadobrasil/128171 (2018).42.Zunino, M. Food relocation behaviour: A multivalent strategy of Coleoptera. In Advances in Coleopterology (eds Zunino, M. et al.) 297–314 (AEC, 1991).
    Google Scholar 
    43.LaBarbera, M. Analyzing body size as a factor in ecology and evolution. Ann. Rev. Ecol. Syst. 20, 97–117 (1989).
    Google Scholar 
    44Soto, C. S., Giombini, M. I., Giménez Gómez, V. C. & Zurita, G. A. Phenotypic differentiation in a resilient dung beetle species induced by forest conversion into cattle pastures. Evol. Ecol. 33, 385–402 (2019).
    Google Scholar 
    45.Laliberté, E., Legendre, P. & Shipley, B. Package ‘FD’. Measuring Functional Diversity (FD) from Multiple Traits, and Other Tools for Functional Ecology (2014).46.Gower, J. C. A general coefficient of similarity and some of its properties. Biometrics 27, 857 (1971).
    Google Scholar 
    47.Pavoine, S., Vallet, J., Dufour, A.-B., Gachet, S. & Daniel, H. On the challenge of treating various types of variables: Application for improving the measurement of functional diversity. Oikos 118, 391–402 (2009).
    Google Scholar 
    48.Moran, P. A. P. Notes on continuous stochastic phenomena. Biometrika 37, 17–23 (1950).MathSciNet 
    CAS 
    PubMed 
    MATH 

    Google Scholar 
    49.Zuur, A. F., Ieno, E. N. & Elphick, C. S. A protocol for data exploration to avoid common statistical problems: Data exploration. Methods Ecol. Evol. 1, 3–14 (2010).
    Google Scholar 
    50.Lavorel, S. et al. Assessing functional diversity in the field—Methodology matters!. Funct. Ecol. 22, 134–147 (2008).
    Google Scholar 
    51.Oksanen, J. et al. vegan: Community Ecology Package (2017).52.Clarke, K. R. & Green, R. H. Statistical design and analysis for a ‘biological effects’ study. Mar. Ecol. Prog. Ser. 46, 213–226 (1988).ADS 

    Google Scholar 
    53.da Silva, P. G. & Cassenote, S. Environmental drivers of species composition and functional diversity of dung beetles along the Atlantic Forest-Pampa transition zone. Austral. Ecol. 44, 786–799 (2019).
    Google Scholar 
    54.Giraldo, C., Escobar, F., Chará, J. D. & Calle, Z. The adoption of silvopastoral systems promotes the recovery of ecological processes regulated by dung beetles in the Colombian Andes: Ecological processes regulated by dung beetles. Insect Conserv. Divers. 4, 115–122 (2011).
    Google Scholar 
    55.Nichols, E. et al. Trait-dependent response of dung beetle populations to tropical forest conversion at local and regional scales. Ecology 94, 180–189 (2013).PubMed 

    Google Scholar 
    56Gómez-Cifuentes, A., Giménez Gómez, V. C., Moreno, C. E. & Zurita, G. A. Tree retention in cattle ranching systems partially preserves dung beetle diversity and functional groups in the semideciduous Atlantic forest: The role of microclimate and soil conditions. Basic Appl. Ecol. 34, 64–74 (2019).
    Google Scholar 
    57.Cerullo, G. R., Edwards, F. A., Mills, S. C. & Edwards, D. P. Tropical forest subjected to intensive post-logging silviculture maintains functionally diverse dung beetle communities. For. Ecol. Manage. 444, 318–326 (2019).
    Google Scholar 
    58.Filloy, J., Zurita, G. A., Corbelli, J. M. & Bellocq, M. I. On the similarity among bird communities: Testing the influence of distance and land use. Acta Oecol. 36, 333–338 (2010).ADS 

    Google Scholar 
    59.Chown, S. L., Sørensen, J. G. & Terblanche, J. S. Water loss in insects: An environmental change perspective. J. Insect Physiol. 57, 1070–1084 (2011).CAS 
    PubMed 

    Google Scholar 
    60.Duncan, F. D. & Byrne, M. J. Discontinuous gas exchange in dung beetles: Patterns and ecological implications. Oecologia 122, 452–458 (2000).ADS 
    CAS 
    PubMed 

    Google Scholar 
    61.Lobo, J. M., Lumaret, J.-P. & Jay-Robert, P. Sampling dung beetles in the French Mediterranean area: Effects of abiotic factors and farm practices. Pedobiología 42(3), 252–266 (1998).
    Google Scholar 
    62.Navarrete, D. & Halffter, G. Dung beetle (Coleoptera: Scarabaeidae: Scarabaeinae) diversity in continuous forest, forest fragments and cattle pastures in a landscape of Chiapas, Mexico: The effects of anthropogenic changes. Biodivers. Conserv. 17, 2869–2898 (2008).
    Google Scholar 
    63.Verdú, J. R., Arellano, L. & Numa, C. Thermoregulation in endothermic dung beetles (Coleoptera: Scarabaeidae): Effect of body size and ecophysiological constraints in flight. J. Insect Physiol. 52, 854–860 (2006).PubMed 

    Google Scholar 
    64.Davis, A. J., Huijbregts, H. & Krikken, J. The role of local and regional processes in shaping dung beetle communities in tropical forest plantations in Borneo. Glob. Ecol. 9, 281–292 (2000).
    Google Scholar 
    65.Tuff, K. T., Tuff, T. & Davies, K. F. A framework for integrating thermal biology into fragmentation research. Ecol. Lett. 19, 361–374 (2016).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    66.Davis, A. L. V. Habitat fragmentation in southern Africa and distributional response patterns in five specialist or generalist dung beetle families (Coleoptera). Afr. J. Ecol. 32, 192–207 (1994).
    Google Scholar 
    67.Halffter, G. & Arellano, L. Response of dung beetle diversity to human-induced changes in a tropical landscape. Biotropica 34, 144–154 (2002).
    Google Scholar 
    68.Hill, C. Habitat specificity and food preferences of an assemblage of tropical Australian dung beetles. J. Trop. Ecol. 12, 449–460 (1996).
    Google Scholar 
    69.Supp, S. R. & Ernest, S. K. M. Species-level and community-level responses to disturbance: A cross-community analysis. Ecology 95, 1717–1723 (2014).PubMed 

    Google Scholar 
    70.Davis, A. L. V., Scholtz, C. H. & Deschodt, C. Multi-scale determinants of dung beetle assemblage structure across abiotic gradients of the Kalahari-Nama Karoo ecotone, South Africa. J. Biogeogr. 35, 1465–1480 (2008).
    Google Scholar 
    71.Nervo, B., Tocco, C., Caprio, E., Palestrini, C. & Rolando, A. The effects of body mass on dung removal efficiency in dung beetles. PLoS ONE 9, e107699 (2014).ADS 
    PubMed 
    PubMed Central 

    Google Scholar 
    72.Bui, V. B., Ziegler, T. & Bonkowski, M. Morphological traits reflect dung beetle response to land use changes in tropical karst ecosystems of Vietnam. Ecol. Ind. 108, 105697 (2020).
    Google Scholar 
    73.Giménez Gómez, V. C., Verdú, J. R. & Zurita, G. A. Thermal niche helps to explain the ability of dung beetles to exploit disturbed habitats. Sci. Rep. 10, 13364 (2020).ADS 
    PubMed 
    PubMed Central 

    Google Scholar 
    74.Verdú, J. R., Alba-Tercedor, J. & Jiménez-Manrique, M. Evidence of different thermoregulatory mechanisms between two sympatric Scarabaeus species using infrared thermography and micro-computer tomography. PLoS ONE 7, e33914 (2012).ADS 
    PubMed 
    PubMed Central 

    Google Scholar 
    75.Gómez-Cifuentes, A., Vespa, N., Semmartín, M. & Zurita, G. Canopy cover is a key factor to preserve the ecological functions of dung beetles in the southern Atlantic Forest. Appl. Soil. Ecol. 154, 103652 (2020).
    Google Scholar 
    76.Fernández, P. D. et al. Understanding the distribution of cattle production systems in the South American Chaco. J. Land Use Sci. 15, 52–68 (2020).
    Google Scholar 
    77Grau, H. R. & Aide, M. Globalization and land-use transitions in Latin America. Ecol. Soc. 13, 16 (2008).
    Google Scholar 
    78.Mastrangelo, M. E. & Gavin, M. C. Trade-offs between cattle production and bird conservation in an agricultural frontier of the Gran Chaco of Argentina. Conserv. Biol. 26, 1040–1051 (2012).PubMed 

    Google Scholar 
    79.Macchi, L. et al. Thresholds in forest bird communities along woody vegetation gradients in the South American Dry Chaco. J. Appl. Ecol. 56, 629–639 (2019).
    Google Scholar 
    80.Díaz, S. & Cabido, M. Vive la différence: Plant functional diversity matters to ecosystem processes. Trends Ecol. Evol. 16, 646–655 (2001).
    Google Scholar 
    81.Slade, E. M., Mann, D. J., Villanueva, J. F. & Lewis, O. T. Experimental evidence for the effects of dung beetle functional group richness and composition on ecosystem function in a tropical forest. J. Anim. Ecol. 76, 1094–1104 (2007).PubMed 

    Google Scholar 
    82.Ortega-Martínez, I. J., Moreno, C. E. & Escobar, F. A dirty job: manure removal by dung beetles in both a cattle ranch and laboratory setting. Entomol. Exp. Appl. 161, 70–78 (2016).
    Google Scholar  More

  • in

    Multiple roles of bamboo as a regulator of cyanobacterial bloom in aquatic systems

    1.Merel, S. et al. State of knowledge and concerns on cyanobacterial blooms and cyanotoxins. Environ. Int. 59, 303–327 (2013).CAS 
    PubMed 

    Google Scholar 
    2.Huisman, J. et al. Cyanobacterial blooms. Nat. Rev. Microbiol. 16, 471–483 (2018).CAS 
    PubMed 

    Google Scholar 
    3.Paerl, H. W. & Otten, T. G. Harmful cyanobacterial blooms: Causes, consequences, and controls. Microb. Ecol. 65, 995–1010 (2013).CAS 
    PubMed 

    Google Scholar 
    4.Ibelings, B. W. & Chorus, I. Accumulation of cyanobacterial toxins in freshwater “seafood” and its consequences for public health: A review. Environ. Pollut. 150, 177–192 (2007).CAS 
    PubMed 

    Google Scholar 
    5.Cheung, M. Y., Liang, S. & Lee, J. Toxin-producing cyanobacteria in freshwater: A review of the problems, impact on drinking water safety, and efforts for protecting public health. J. Microbiol. 51, 1–10 (2013).CAS 
    PubMed 

    Google Scholar 
    6.Rousso, B. Z., Bertone, E., Stewart, R. & Hamilton, D. P. A systematic literature review of forecasting and predictive models for cyanobacteria blooms in freshwater lakes. Water Res. 182, 115959 (2020).7.Vadeboncoeur, Y. et al. From Greenland to green lakes: Cultural eutrophication and the loss of benthic pathways in lakes. Limnol. Oceanogr. 48, 1408–1418 (2003).ADS 

    Google Scholar 
    8.Han, Z. & Cui, B. Performance of macrophyte indicators to eutrophication pressure in ponds. Ecol. Eng. 96, 8–19 (2016).
    Google Scholar 
    9.Dorgham, M. Effects of Eutrophication. In Eutrophication: Causes, Consequences and Control (eds. Ansari, A. & Gill, S.). vol. 2, 29–44. (Springer, 2014).10.Glibert, P. M. Eutrophication, harmful algae and biodiversity—Challenging paradigms in a world of complex nutrient changes. Mar. Pollut. Bull. 124, 591–606 (2017).CAS 
    PubMed 

    Google Scholar 
    11.Lürling, M. & Mucci, M. Mitigating eutrophication nuisance: In-lake measures are becoming inevitable in eutrophic waters in the Netherlands. Hydrobiologia 847, 4447–4467 (2020).
    Google Scholar 
    12.Hall, R. O., Likens, G. E. & Malcom, H. M. Trophic basis of invertebrate production in 2 streams at the Hubbard Brook Experimental Forest. J. N. Am. Benthol. Soc. 20, 432–447 (2001).
    Google Scholar 
    13.Tanentzap, A. J. et al. Forests fuel fish growth in freshwater deltas. Nat. Commun. 5, 4077 (2014).ADS 
    CAS 
    PubMed 

    Google Scholar 
    14.Fey, S. B., Mertens, A. N. & Cottingham, K. L. Autumn leaf subsidies influence spring dynamics of freshwater plankton communities. Oecologia 178, 875–885 (2015).ADS 
    PubMed 

    Google Scholar 
    15.Wondzell, S. M. & Bisson, P. A. Influence of wood on aquatic biodiversity. Am. Fish. Soc. Symp. 37, 249–263 (2003).
    Google Scholar 
    16.Czarnecka, M. Coarse woody debris in temperate littoral zones: Implications for biodiversity, food webs and lake management. Hydrobiologia 767, 13–25 (2016).
    Google Scholar 
    17.Graham, M. D. & Vinebrooke, R. D. Coupling of boreal forests and lakes: Effects of conifer pollen on littoral communities. Limnol. Oceanogr. 51, 1524–1529 (2006).ADS 

    Google Scholar 
    18.Kelly, P. T. et al. Experimental whole-lake increase of dissolved organic carbon concentration produces unexpected increase in crustacean zooplankton density. Glob. Change Biol. 22, 2766–2775 (2016).ADS 

    Google Scholar 
    19.Shao, J., Li, R., Lepo, J. E. & Gu, J. D. Potential for control of harmful cyanobacterial blooms using biologically derived substances: Problems and prospects. J. Environ. Manag. 125, 149–155 (2013).
    Google Scholar 
    20.Tan, K. et al. A review of allelopathy on microalgae. Microbiology 165, 587–592 (2019).CAS 
    PubMed 

    Google Scholar 
    21.Tsuchiya, R., Kihei, M., Sakagami, Y. & Araki, T. Assessment of inhibition effect on growth of Microcystis aeruginosa by autoclaved water extracts from leaves of 104 woody plant species. J. Jpn. Limnol. 79, 41–48 (2018) (in Japanese with English abstract).22.Neilen, A. D., Hawker, D. W., O’Brien, K. R. & Burford, M. A. Phytotoxic effects of terrestrial dissolved organic matter on a freshwater cyanobacteria and green algae species is affected by plant source and DOM chemical composition. Chemosphere 184, 969–980 (2017).ADS 
    CAS 
    PubMed 

    Google Scholar 
    23.Chen, J., Zhang, H., Han, Z., Ye, J. & Liu, Z. The influence of aquatic macrophytes on Microcystis aeruginosa growth. Ecol. Eng. 42, 130–133 (2012).
    Google Scholar 
    24.Zhou, B., Fu, M., Xie, J., Yang, X. & Li, Z. Ecological functions of bamboo forest: Research and application. J. For. Res. 16, 143–147 (2005).
    Google Scholar 
    25.Xu, Q. F. et al. Rapid bamboo invasion (expansion) and its effects on biodiversity and soil processes +. Glob. Change Biol. 21, e00787 (2020).26.Shinohara, Y., Misumi, Y., Kubota, T. & Nanko, K. Characteristics of soil erosion in a moso-bamboo forest of western Japan: Comparison with a broadleaved forest and a coniferous forest. CATENA 172, 451–460 (2019).
    Google Scholar 
    27.Suzuki, S. & Nakagoshi, N. Expansion of bamboo forests caused by reduced bamboo-shoot harvest under different natural and artificial conditions. Ecol. Res. 23, 641–647 (2008).
    Google Scholar 
    28.Buziquia, S. T., Lopes, P. V. F., Almeida, A. K. & de Almeida, I. K. Impacts of bamboo spreading: A review. Biodivers. Conserv. 28, 3695–3711 (2019).
    Google Scholar 
    29.Kudo, G., Amagai, Y., Hoshino, B. & Kaneko, M. Invasion of dwarf bamboo into alpine snow-meadows in Northern Japan: Pattern of expansion and impact on species diversity. Ecol. Evol. 1, 85–96 (2011).PubMed 
    PubMed Central 

    Google Scholar 
    30.Wei, Q. et al. The diversity of soil mesofauna decline after bamboo invasion in subtropical China. Sci. Total Environ. 789, 147982 (2021).31.Fujii, Y. & Kobayashi, Y. Allelopathic activities of leaf leachates of Bamboo and Sasa; sandwich method of 80 species. Weed Biol. Manag. 39, 94–95 (1994).
    Google Scholar 
    32.Ogita, S. & Sasamoto, H. In vitro bioassay of allelopathy in four bamboo species; Bambusa multiplex, Phyllostachys bambusoides, P. nigra, Sasa kurilensis, using sandwich method and protoplast co-culture method with digital image analysis. Am. J. Plant Sci. 8, 1699 (2017).33.Chuyen, N. V., Kurata, T., Kato, H. & Fujimaki, M. Antimicrobial activity of Kumazasa (Sasa albo-marginata). Agr. Biol. Chem. 46, 971–978 (1982).
    Google Scholar 
    34.Chongtham, N., Bisht, M. S. & Haorongbam, S. Nutritional properties of bamboo shoots: potential and prospects for utilization as a health food. Compr. Rev. Food Sci. Food Saf. 10, 153–168 (2011).CAS 

    Google Scholar 
    35.Singhal, P., Satya, S. & Sudhakar, P. Antioxidant and pharmaceutical potential of bamboo leaves. Bamboo Sci. Cult. 24, 19–28 (2011).
    Google Scholar 
    36.Jin, L. et al. Bamboo nutrients and microbiome affect gut microbiome of giant panda. Symbiosis 80, 293–304 (2020).CAS 

    Google Scholar 
    37.Lin, Y. T. et al. Changes in the soil bacterial communities in a cedar plantation invaded by moso bamboo. Microb. Ecol. 67, 421–429 (2014).PubMed 

    Google Scholar 
    38.Li, Y. et al. Bamboo invasion of broadleaf forests altered soil fungal community closely linked to changes in soil organic C chemical composition and mineral N production. Plant Soil 418, 507–521 (2017).CAS 

    Google Scholar 
    39.Liu, X. et al. Moso bamboo (Phyllostachys edulis) invasion effects on litter, soil and microbial PLFA characteristics depend on sites and invaded forests. Plant Soil 438, 85–99 (2019).CAS 

    Google Scholar 
    40.O’connor, P. J., Covich, A. P., Scatena, F. N. & Loope, L. L. Non-indigenous bamboo along headwater streams of the Luquillo Mountains, Puerto Rico: Leaf fall, aquatic leaf decay and patterns of invasion. J. Trop. Ecol. 16, 499–516 (2000).
    Google Scholar 
    41.Cai, L., Zhang, K., McKenzie, E. H. & Hyde, K. D. Freshwater fungi from bamboo and wood submerged in the Liput River in the Philippines. Fungal Divers. 13, 1–12 (2003).
    Google Scholar 
    42.Suto, S. Mariculture of seaweeds and its problems in Japan. NOAA Tech. Rep. NMFS Circ 388, 7–16 (1974).
    Google Scholar 
    43.Milstein, A., Azim, M. E., Wahab, M. A. & Verdegem, M. C. J. The effects of periphyton, fish and fertilizer dose on biological processes affecting water quality in earthen fish ponds. Environ. Biol. Fishes 68, 247–260 (2003).
    Google Scholar 
    44.Azim, M. E. et al. The effect of periphyton substrate density on production in freshwater polyculture ponds. Aquaculture 232, 441–453 (2004).
    Google Scholar 
    45.Khatoon, H., Yusoff, F., Banerjee, S., Shariff, M. & Bujang, J. S. Formation of periphyton biofilm and subsequent biofouling on different substrates in nutrient enriched brackishwater shrimp ponds. Aquaculture 273, 470–477 (2007).
    Google Scholar 
    46.Ma, J. F. & Takahashi, E. Soil, Fertilizer, and Plant Silicon Research in Japan. (Elsevier Science, 2002).47.Akagi, T. et al. Dissolved ion analyses of stream water from bamboo forests: Implication for enhancement of chemical weathering by bamboo. Geochem. J. 46, 505–515 (2012).ADS 
    CAS 

    Google Scholar 
    48.Umemura, M. & Takenaka, C. Biological cycle of silicon in moso bamboo (Phyllostachys pubescens) forests in central Japan. Ecol. Res. 29, 501–510 (2014).CAS 

    Google Scholar 
    49.Lürling, M. & Roessink, I. On the way to cyanobacterial blooms: impact of the herbicide metribuzin on the competition between a green alga (Scenedesmus) and a cyanobacterium (Microcystis). Chemosphere 65, 618–626 (2006).ADS 
    PubMed 

    Google Scholar 
    50.Ji, X., Verspagen, J. M., Stomp, M. & Huisman, J. Competition between cyanobacteria and green algae at low versus elevated CO2: Who will win, and why?. J. Exp. Bot. 68, 3815–3828 (2017).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    51.Kang, C. et al. Effects of macrophyte Vallisneria asiatica biomasses on the algae community. Int. J. Environ. Eng. 7, 1161–1166 (2013).
    Google Scholar 
    52.Hao, A., Haraguchi, T., Kuba, T., Kai, H., Lin, Y. & Iseri, Y. Effect of the microorganism-adherent carrier for Nitzschia palea to control the cyanobacterial blooms. Ecol. Eng. 159, 106127 (2021).53.Wang, Z., Li, G., Li, G. & Li, D. The decline process and major pathways of Microcystis bloom in Taihu Lake, China. Chin. J. Oceanol. Limnol. 30, 37–46 (2012).ADS 
    CAS 

    Google Scholar 
    54.Xiao, M., Li, M. & Reynolds, C. S. Colony formation in the cyanobacterium. Microcystis Biol. Rev. 93, 1399–1420 (2018).PubMed 

    Google Scholar 
    55.Wu, Y. et al. Allelopathic control of cyanobacterial blooms by periphyton biofilms. Environ. Microb. 13, 604–615 (2011).CAS 

    Google Scholar 
    56.Ko, S. R. et al. Bioremediation of eutrophic water and control of cyanobacterial bloom by attached periphyton. Int. J. Environ. Sci. Technol. 16, 4173–4180 (2019).CAS 

    Google Scholar 
    57.Mühlbauer, L. K., Schulze, M., Harpole, W. S. & Clark, A. T. gauseR: Simple methods for fitting Lotka-Volterra models describing Gause’s “Struggle for Existence”. Ecol. Evol. 10, 13275–13283 (2020).PubMed 
    PubMed Central 

    Google Scholar 
    58.Li, J. et al. Growth inhibition and oxidative damage of Microcystis aeruginosa induced by crude extract of Sagittaria trifolia tubers. J. Environ. Sci. 43, 40–47 (2016).
    Google Scholar 
    59.Ma, J. et al. Environmental factors controlling colony formation in blooms of the cyanobacteria Microcystis spp. in Lake Taihu, China. Harmful Algae 31, 136–142 (2014).60.Hua, Q. et al. Allelopathic effect of the rice straw aqueous extract on the growth of Microcystis aeruginosa. Ecotoxicol. Environ. Saf. 148, 953–959 (2018).CAS 

    Google Scholar 
    61.Zhao, W., Zheng, Z., Zhang, J., Roger, S. F. & Luo, X. Allelopathically inhibitory effects of eucalyptus extracts on the growth of Microcystis aeruginosa. Chemosphere 225, 424–433 (2019).ADS 
    CAS 
    PubMed 

    Google Scholar 
    62.Ball, A. S., Williams, M., Vincent, D. & Robinson, J. Algal growth control by a barley straw extract. Bioresour. Technol. 77, 177–181 (2001).CAS 
    PubMed 

    Google Scholar 
    63.Park, M. H., Kim, B. H., Chung, I. M. & Hwang, S. J. Selective bactericidal potential of rice (Oryza sativa L. var. japonica) hull extract on Microcystis strains in comparison with green algae and zooplankton. Bull. Environ. Contam. Toxicol. 83, 97–101 (2009).64.Le Rouzic, B., Thiébaut, G. & Brient, L. Selective growth inhibition of cyanobacteria species (Planktothrix agardhii) by a riparian tree leaf extract. Ecol. Eng. 97, 74–78 (2016).
    Google Scholar 
    65.Eladel, H., Battah, M., Dawa, A., Abd-Elhay, R. & Anees, D. Effect of rice straw extracts on growth of two phytoplankton isolated from a fish pond. J. Appl. Phycol. 31, 3557–3563 (2019).
    Google Scholar 
    66.Yang, J. et al. High temperature and pH favor Microcystis aeruginosa to outcompete Scenedesmus obliquus. Environ. Sci. Pollut. Res. 25, 4794–4802 (2018).CAS 

    Google Scholar 
    67.Grover, J. P. Phosphorus-dependent growth kinetics of 11 species of freshwater algae. Limnol. Oceanogr. 34, 341–348 (1989).ADS 
    CAS 

    Google Scholar 
    68.Shia, L. et al. Community structure of bacteria associated with Microcystis colonies from cyanobacterial blooms. J. Freshwat. Ecol. 25, 193–203 (2010).
    Google Scholar 
    69.Smith, D. J. et al. Individual Microcystis colonies harbour distinct bacterial communities that differ by Microcystis oligotype and with time. Environ. Microbiol. 23, 3020–3036 (2021).CAS 
    PubMed 

    Google Scholar  More

  • in

    Fecal and soil microbiota composition of gardening and non-gardening families

    1.Turnbaugh, P. J. et al. The human microbiome project. Nature 449, 804–810 (2007).ADS 
    CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    2.Sonnenburg, J. L. & Bäckhed, F. Diet-microbiota interactions as moderators of human metabolism. Nature 535, 56–64 (2016).ADS 
    CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    3.Rook, G. A. Regulation of the immune system by biodiversity from the natural environment: An ecosystem service essential to health. Proc. Natl. Acad. Sci. U.S.A. 110, 18360–18367 (2013).ADS 
    CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    4.Huang, Y. J. et al. The microbiome in allergic disease: Current understanding and future opportunities. J. Allergy Clin. Immunol. 139, 1099–1110 (2018).
    Google Scholar 
    5.Morgan, X. C. et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 13, 2 (2012).
    Google Scholar 
    6.Schnabl, B. Linking intestinal homeostasis and liver disease. Curr. Opin. Gastroenterol. 29, 264–270 (2013).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    7.Scott, F. W., Pound, L. D., Patrick, C., Eberhard, C. E. & Crookshank, J. A. Where genes meet environment—integrating the role of gut luminal contents, immunity and pancreas in type 1 diabetes. Transl. Res. 179, 183–198 (2017).CAS 
    PubMed 

    Google Scholar 
    8.Rook, G. A. W. Hygiene hypothesis and autoimmune diseases. Clin. Rev. Allergy Immunol. 42, 5–15 (2012).CAS 
    PubMed 

    Google Scholar 
    9.Deehan, E. C. & Walter, J. The fiber gap and the disappearing gut microbiome: implications for human nutrition. Trends Endocrinol. Metab. 27, 239–242 (2016).CAS 
    PubMed 

    Google Scholar 
    10.Rothschild, D. et al. Environment dominates over host genetics in shaping human gut microbiota. Nature 555, 210–215 (2018).ADS 
    CAS 
    PubMed 

    Google Scholar 
    11.Schloss, P. D., Iverson, K. D., Petrosino, J. F. & Schloss, S. J. The dynamics of a family’s gut microbiota reveal variations on a theme. Microbiome 2, 1–13 (2014).
    Google Scholar 
    12.Song, S. J. et al. Cohabiting family members share microbiota with one another and with their dogs. ELife 2, 1–22 (2013).
    Google Scholar 
    13.Seedorf, H. et al. Bacteria from diverse habitats colonize and compete in the mouse gut. Cell 159, 253–266 (2014).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    14.Zhou, D. et al. Exposure to soil, house dust and decaying plants increases gut microbial diversity and decreases serum immunoglobulin E levels in BALB/c mice. Environ. Microbiol. 18, 1326–1337 (2016).CAS 
    PubMed 

    Google Scholar 
    15.Schnorr, S. L. et al. Gut microbiome of the Hadza hunter-gatherers. Nat. Commun. 5, 2 (2014).
    Google Scholar 
    16.Tasnim, N., Abulizi, N., Pither, J., Hart, M. M. & Gibson, D. L. Linking the gut microbial ecosystem with the environment: Does gut health depend on where we live?. Front. Microbiol. 8, 1–8 (2017).
    Google Scholar 
    17.Schnorr, S. L. The soil in our microbial DNA informs about environmental interfaces across host and subsistence modalities: Soil taxa in human gut microbiome. Philos. Trans. R. Soc. B Biol. Sci. 375, 2 (2020).
    Google Scholar 
    18.Rook, G. A. W. 99th Dahlem conference on infection, inflammation and chronic inflammatory disorders: Darwinian medicine and the “hygiene” or “old friends” hypothesis. Clin. Exp. Immunol. 160, 70–79 (2010).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    19.de Filippo, C. et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc. Natl. Acad. Sci. 107, 14691–14696 (2010).ADS 
    PubMed 
    PubMed Central 

    Google Scholar 
    20.Martínez, I. et al. The gut microbiota of rural papua new guineans: Composition, diversity patterns, and ecological processes. Cell Rep. 11, 527–538 (2015).PubMed 

    Google Scholar 
    21.Senghor, B., Sokhna, C., Ruimy, R. & Lagier, J. C. Gut microbiota diversity according to dietary habits and geographical provenance. Hum. Microbiome J. 7–8, 1–9 (2018).
    Google Scholar 
    22.Holscher, H. D. Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut Microbes 8, 172–184 (2017).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    23.McDonald, D. et al. American gut: an open platform for citizen-science microbiome research. mSystems 3, 1–28 (2018).
    Google Scholar 
    24.Mills, J. G. et al. Urban habitat restoration provides a human health benefit through microbiome rewilding: The Microbiome Rewilding Hypothesis. Restor. Ecol. 25, 866–872 (2017).
    Google Scholar 
    25.Shenhav, L. et al. FEAST: Fast expectation-maximization for microbial source tracking. Nat. Methods 16, 627–632 (2019).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    26.Dhillon, J., Li, Z. & Ortiz, R. M. Almond snacking for 8 wk increases alpha-diversity of the gastrointestinal microbiome and decreases bacteroides fragilis abundance compared with an isocaloric snack in college freshmen. Curr. Dev. Nutr. 3, 1–9 (2019).CAS 

    Google Scholar 
    27.Thompson, S. V. et al. Avocado consumption alters gastrointestinal bacteria abundance and microbial metabolite concentrations among adults with overweight or obesity: A randomized controlled trial. J. Nutr. 151, 753–762 (2021).PubMed 

    Google Scholar 
    28.Yu, D. et al. Long-term diet quality is associated with gut microbiome diversity and composition among urban Chinese adults. Am. J. Clin. Nutr. 113, 684–694 (2021).PubMed 
    PubMed Central 

    Google Scholar 
    29.Koh, A., de Vadder, F., Kovatcheva-Datchary, P. & Bäckhed, F. From dietary fiber to host physiology: Short-chain fatty acids as key bacterial metabolites. Cell 165, 1332–1345 (2016).CAS 
    PubMed 

    Google Scholar 
    30.Chung, W. S. F. et al. Modulation of the human gut microbiota by dietary fibres occurs at the species level. BMC Biol. 14, 1–13 (2016).
    Google Scholar 
    31.Kaczmarek, J. L. et al. Broccoli consumption affects the human gastrointestinal microbiota. J. Nutr. Biochem. 63, 27–34 (2019).CAS 
    PubMed 

    Google Scholar 
    32.Rose, D. J., DeMeo, M. T. & Keshavarzian, A. Influence of dietary fiber on inflammatory bowel disease and colon cancer: Importance of fermentation pattern. Nutr. Rev. 65, 51–62 (2007).PubMed 

    Google Scholar 
    33.Keohane, D. M. et al. Microbiome and health implications for ethnic minorities after enforced lifestyle changes. Nat. Med. 26, 1089–1095 (2020).CAS 
    PubMed 

    Google Scholar 
    34.Fierer, N. Embracing the unknown: Disentangling the complexities of the soil microbiome. Nat. Rev. Microbiol. 15, 579–590 (2017).CAS 
    PubMed 

    Google Scholar 
    35.Ottman, N. et al. Soil exposure modifies the gut microbiota and supports immune tolerance in a mouse model. J. Allergy Clin. Immunol. 143, 1198-1206.e12 (2019).CAS 
    PubMed 

    Google Scholar 
    36.Turnbaugh, P. J. et al. A core gut microbiome in obese and lean twins. Nature 457, 480–484 (2009).ADS 
    CAS 

    Google Scholar 
    37.Rook, G. A. W., Lowry, C. A. & Raison, C. L. Microbial, “Old Friends”, immunoregulation and stress resilience. Evol. Med. Public Heal. 2013, 46–64 (2013).
    Google Scholar 
    38.Brame, J. E., Liddicoat, C., Abbott, C. A. & Breed, M. F. The potential of outdoor environments to supply beneficial butyrate-producing bacteria to humans. Sci. Total Environ. 777, 2 (2021).
    Google Scholar 
    39.Caporaso, J. G. et al. Ultra-high-throughput microbial community analysis on the illumina HiSeq and MiSeq platforms. ISME J. 6, 1621–1624 (2012).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    40.Venable, E. B. et al. Effects of feeding management on the equine cecal microbiota. J. Equine Vet. Sci. 49, 113–121 (2017).
    Google Scholar 
    41.Bolyen, E. et al. Reproducible, interactive, scalable, and extensible microbiome data science using QIIME2. Nat. Biotechnol. 37, 852–857 (2019).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    42.Callahan, B. J. et al. DADA2: High-resolution sample inference from Illumina amplicon data. Nat. Methods 13, 581–583 (2016).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    43.Yilmaz, P. et al. The SILVA and “all-species living tree project (LTP)” taxonomic frameworks. Nucleic Acids Res. 42, 643–648 (2014).
    Google Scholar 
    44.Subar, A. F. et al. The automated self-administered 24-hour dietary recall (ASA24): A resource for researchers, clinicians, and educators from the national cancer institute. J. Acad. Nutr. Diet 112, 1134–1137 (2012).PubMed 
    PubMed Central 

    Google Scholar 
    45.Miller, P. E. et al. Development and evaluation of a method for calculating the Healthy Eating Index-2005 using the Nutrition Data System for Research. Public Health Nutr. 14, 306–313 (2011).PubMed 

    Google Scholar 
    46.Krebs-Smith, S. M. et al. Update of the healthy eating index: HEI-2015. J. Acad. Nutr. Diet 118, 1591–1602 (2018).PubMed 
    PubMed Central 

    Google Scholar 
    47.Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: A practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B 57, 289–300 (1995).MathSciNet 
    MATH 

    Google Scholar 
    48.Segata, N. et al. Metagenomic biomarker discovery and explanation. Genome Biol 12, 2 (2011).
    Google Scholar  More

  • in

    Influence of state reopening policies in COVID-19 mortality

    1.

    2.Li, Q. et al. Early transmission dynamics in Wuhan, China, of novel coronavirus-infected pneumonia. N. Engl. J. Med. 382, 1199 (2020).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    3.Anderson, R. M., Heesterbeek, H., Klinkenberg, D. & Hollingsworth, T. D. How will country-based mitigation measures influence the course of the COVID-19 epidemic? Lancent 395, 931 (2020).CAS 

    Google Scholar 
    4.WHO. Coronavirus disease (COVID 2019) situation report-30.5.Linton, N. M. et al. Incubation period and other epidemiological characteristics of 2019 novel coronavirus infections with right truncation: A statistical analysis of publicly available case data. J. Clin. Med. 9, 538 (2020).PubMed Central 

    Google Scholar 
    6.
    https://www.washingtonpost.com/graphics/2020/national/states-reopening-coronavirus-map/
    7.Kaufman, G. B. et al. Comparing associations of state reopening strategies with COVID-19 burden. J. Gen. Intern. Med. 35, 3627 (2020).PubMed 
    PubMed Central 

    Google Scholar 
    8.Woolf, S. H. et al. Excess deaths from COVID-19 and other causes, March-July 2020. JAMA 324, 1562 (2020).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    9.Faust, J. S. et al. All-cause excess mortality and COVID-19-related mortality among US adults aged 25–44 years, March–July 2020. JAMA 325, 785 (2021).CAS 
    PubMed 

    Google Scholar 
    10.Huppert, A. & Katriel, G. Mathematical modelling and prediction in infectious disease epidemiology. Clin. Microbiol. Infect. 19, 999 (2003).
    Google Scholar 
    11.Kermack, W. O. & McKendrick, A. G. A contribution to the mathematical theory of epidemics. Proc. R. Soc. Lond. 115, 700 (1927).ADS 
    MATH 

    Google Scholar 
    12.Crokidakis, C. Modeling the early evolution of the COVID-19 in Brazil: Results from a susceptible-infectious-quarantined-recovered (SIQR) model. Int. J. Mod. Phys. C 31, 2050135 (2020).ADS 
    MathSciNet 
    CAS 

    Google Scholar 
    13.Bin, M. et al. Post-lockdown abatement of COVID-19 by fast periodic switching. PLoS Comput. Biol. 17, e1008604 (2021).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    14.Pedersen, M. G., & Meneghini, M. Quantifying undetected COVID-19 cases and effects of containment measures in Italy: Predicting phase 2 dynamics. https://doi.org/10.13140/RG.2.2.11753.85600 (2020).15.Calafiore, G. C., Novara, C., & Possieri, C. A Modified SIR Model for the COVID-19 Contagion in Italy. arXiv:2003.14391 (2020).16.Bastos, S. B., & Cajueiro, D. O. Modeling and forecasting the early evolution of the Covid-19 pandemic in Brazil. arXiv:2003.14288 (2020).17.Gaeta, G. Chaos, social distancing versus early detection and contacts tracing in epidemic management. Solitons Fractals 140, 110074 (2020).MathSciNet 

    Google Scholar 
    18.Gaeta, G. Asymptomatic infectives and R0 for COVID. arXiv:2003.14098 (2020).19.te Vrugt, M., Bickmann, J., & Wittkowski, R. Effects of social distancing and isolation on epidemic spreading: a dynamical density functional theory model. arXiv:2003.13967 (2020).20.Schulz, R. A., Coimbra-Araújo, C. H., & Costiche, S. W. S. COVID-19: A model for studying the evolution of contamination in Brazil. arXiv:2003.13932 (2020).21.Zhang, Y., Yu, X., Sun, H., Tick, Geoffrey R., Wei, W., & Jin, B. COVID-19 infection and recovery in various countries: Modeling the dynamics and evaluating the non-pharmaceutical mitigation scenarios. arXiv:2003.13901 (2020).22.Dell’Anna, L. Solvable delay model for epidemic spreading: the case of Covid-19 in Italy. Sci. Rep. 10, 15763 (2020).ADS 
    PubMed 
    PubMed Central 

    Google Scholar 
    23.Sonnino, G. & Nardone, P. Annals of clinical and medical case reports dynamics of the COVID-19 comparison between the theoretical predictions and the real data, and predictions about returning to normal life. Ann. Clin. Med. Case Rep. 4, 1 (2020).
    Google Scholar 
    24.Notari, A. Temperature dependence of COVID-19 transmission. arXiv:2003.12417 (2020).25.Amaro, J. E. The D model for deaths by COVID-19. arXiv:2003:13747 (2020).26.Simha, A., Prasad, R. V., & Narayana, S. A simple stochastic SIR model for COVID 19 infection dynamics for Karnataka: Learning from Europe. arXiv:2003.11920 (2020).27.Acioli, P. H. Diffusion as a first model of spread of viral infection. arXiv:2003.11449 (2020).28.Zullo, F. Some numerical observations about the COVID-19 epidemic in Italy. arXiv:2003.11363 (2020).29.Sameni, R. Mathematical modeling of epidemic diseases; a case study of the COVID-19 coronavirus. arXiv:2003.11371 (2020).30.Radulescu, A., & Cavanagh, K. Management strategies in a SEIR model of COVID 19 community spread. arXiv:2003.11150 (2020).31.Roques, L., Klein, E., Papaix, J. & Soubeyrand, S. Using early data to estimate the actual infection fatality ratio from COVID-19 in France. MDPI Biol. 9, 97 (2020).CAS 

    Google Scholar 
    32.Teles, P. Predicting the evolution Of SARS-Covid-2 in Portugal using an adapted SIR Model previously used in South Korea for the MERS outbreak. arXiv:2003.10047 (2020).33.Piccolomini, E. L., & Zama, F. Preliminary analysis of COVID-19 spread in Italy with an adaptive SEIRD model. arXiv:2003.09909 (2020).34.Brugnano, L., & Iavernaro, F. A multi-region variant of the SIR model and its extensions. arXiv:2003.09875 (2020).35.Giordano, G. et al. Modelling the COVID-19 epidemic and implementation of population-wide interventions in Italy. the COVID19 IRCCS San Matteo Pavia Task Force. Nat. Med. 16, 855 (2020).
    Google Scholar 
    36.Zlatić, V., Barjašć, I., Kadović, A., Štefančić, H. & Gabrielli, A. Bi-stability of SUDR+ K model of epidemics and test kits applied to COVID-19. Nonlinear Dyn. 101, 1635 (2020).
    Google Scholar 
    37.Baker, R. Reactive Social distancing in a SIR model of epidemics such as COVID-19. arXiv:2003.08285 (2020).38.Biswas, K., Khaleque, A., & Sen, P. Covid-19 spread: Reproduction of data and prediction using a SIR model on Euclidean network. arXiv:2003.07063 (2020).39.Zhang, J., Wang, L., & Wang, J. SIR model-based prediction of infected population of coronavirus in Hubei Province. arXiv:2003.06419 (2020).40.Chen, Y.-C., Lu, P.-E., Chang, C.-S., & Liu, T.-H. A Time-dependent SIR model for COVID-19 with Undetectable Infected Persons. arXiv:2003.00122 (2020).41.Lloyd, A. L. Realistic distributions of infectious periods in epidemic models: Changing patterns of persistence and dynamics. Theor. Popul. Biol. 60, 59 (2001).CAS 
    PubMed 

    Google Scholar 
    42.Fokas, A. S., Dikaios, N. & Kastis, G. A. Mathematical models and deep learning for predicting the number of individuals reported to be infected with SARS-CoV-2. J. R. Soc. Interface 17, 20200494 (2020).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    43.Vadyala, S. R., Betgeri, S. N., Sherer, E. A., & Amritphale, A. Prediction of the number of COVID-19 confirmed cases based on K-means-LSTM. arXiv:2006.14752 (2020).44.Fokas, A. S., Cuevas-Maraver, J. & Kevrekidis, P. G. A quantitative framework for exploring exit strategies from the COVID-19 lockdown. Chaos Solitons Fractals 140, 11024 (2020).MathSciNet 

    Google Scholar 
    45.Coopera, I., Mondal, A. & Antonopoulos, C. G. A SIR model assumption for the spread of COVID-19 in different communities. Chaos Solitons Fractals 139, 110057 (2020).MathSciNet 

    Google Scholar 
    46.Bertozzi, A. L., Franco, E., Mohler, G., Short, M. B. & Sledge, D. The challenges of modeling and forecasting the spread of COVID-19. PNAS 117, 16732 (2020).MathSciNet 
    CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    47.Prem, K. et al. The effect of control strategies to reduce social mixing on outcomes of the COVID-19 epidemic in Wuhan, China: A modelling study. The Lancet Public Health 5, e261 (2020).PubMed 
    PubMed Central 

    Google Scholar 
    48.He, S., Peng, Y. & Sun, K. SEIR modeling of the COVID-19 and its dynamics. Nonlinear Dyn. 101, 1667 (2020).
    Google Scholar 
    49.Mwalili, S., Kimathi, M., Ojiambo, V., Gathungu, D. & Mbogo, R. SEIR model for COVID-19 dynamics incorporating the environment and social distancing. BMC Res. Notes 13, 352 (2020).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    50.Dehning, J. et al. Inferring change points in the spread of COVID-19 reveals the effectiveness of interventions. Science 369, 160 (2020).
    Google Scholar 
    51.Atkeson, A., Kopecky, K. A., & Zha, T. A. Estimating and forecasting disease scenarios for COVID-19 with an SIR Model. NBER Working Paper w27335 (2020).52.Wang, N., Fu, Y., Zhang, H. & Shi, H. An evaluation of mathematical models for the outbreak of COVID-19. Precis. Clin. Med. 3, 85 (2020).
    Google Scholar 
    53.Chang, S. et al. Mobility network models of COVID-19 explain inequities and inform reopening. Nature 589, 82 (2021).ADS 
    CAS 
    PubMed 

    Google Scholar 
    54.Alfano, V. & Ercolano, S. The efficacy of lockdown against COVID-19: A cross-country panel analysis. Appl. Health Econ. Health Policy 18, 509 (2020).PubMed 

    Google Scholar 
    55.Arshed, N., Meo, M. S. & Farooq, F. Empirical assessment of government policies and flattening of the COVID19 curve. J. Public Aff. 20, e2333 (2000).
    Google Scholar 
    56.Auger, K. A. et al. Association between statewide school closure and COVID-19 incidence and mortality in the US. JAMA 324, 859 (2020).CAS 
    PubMed 

    Google Scholar 
    57.Banerjee, T. & Nayak, A. Coping with being cooped up: Social distancing during COVID-19 among 60+ in the United States. Revista Panamericana de Salud Pública. 44, e81 (2020).
    Google Scholar 
    58.Castex, G., Dechter, E. & Lorca, M. COVID-19: The impact of social distancing policies, cross-country analysis. Econ. Disasters Clim. Change 5, 135 (2021).
    Google Scholar 
    59.Bennett, M. All things equal? Heterogeneity in policy effectiveness against COVID-19 spread in Chile. World Dev. 137, 105208 (2021).PubMed 

    Google Scholar 
    60.Castillo, R. C., Staguhn, E. D. & Weston-Farber, E. The effect of state-level stay-at-home orders on COVID-19 infection rates. Am. J. Infect. Control 48, 958 (2020).PubMed 
    PubMed Central 

    Google Scholar 
    61.Cobb, J. S. & Seale, M. A. Examining the effect of social distancing on the compound growth rate of COVID-19 at the county level (United States) using statistical analyses and a random forest machine learning model. Public Health 185, 27 (2020).CAS 
    PubMed 

    Google Scholar 
    62.Courtemanche, C., Garuccio, J., Le, A., Pinkston, J. & Yelowitz, A. Strong social distancing measures in the United States reduced The COVID-19 growth rate. Health Aff. 39, 1237 (2020).
    Google Scholar 
    63.Dave, D., Friedson, A. I., Matsuzawa, K. & Sabia, J. J. When do shelter-in-place orders fight COVID-19 best? Policy heterogeneity across states and adoption time. Econ Inq. 59, 29 (2021).
    Google Scholar 
    64.Dave, D., Friedson, A., Matsuzawa, K., Sabia, J. J. & Safford, S. JUE insight: Were urban cowboys enough to control COVID-19? Local shelter-in-place orders and coronavirus case growth. J. Urban Econ. 2020, 103294 (2020).
    Google Scholar 
    65.Edelstein, M. et al. SARS-CoV-2 infection in London, England: Changes to community point prevalence around lockdown time, March–May 2020. J. Epidemiol. Commun. Health 75, 185 (2021).
    Google Scholar 
    66.Gallaway, M. S. et al. Trends in COVID-19 incidence after implementation of mitigation measures–Arizona, January 22–August 7, 2020. MMWR Morb. Mortal. Wkly. Rep. 69, 1460 (2020).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    67.Hsiang, S. et al. The effect of large-scale anti-contagion policies on the COVID-19 pandemic. Nature 584, 262 (2020).ADS 
    CAS 
    PubMed 

    Google Scholar 
    68.Hyafil, A. & Moriña, D. Analysis of the impact of lockdown on the reproduction number of the SARS-Cov-2 in Spain. Gac. Sanit. 35, 453 (2021).PubMed 

    Google Scholar 
    69.Islam, N., Sharp, S. J. & Chowell, G. Physical distancing interventions and incidence of coronavirus disease 2019: Natural experiment in 149 countries. BMJ 2020, m2743 (2019).
    Google Scholar 
    70.Lyu, W. & Wehby, G. L. Comparison of estimated rates of coronavirus disease 2019 (COVID-19) in border counties in Iowa without a stay-at-home order and border counties in Illinois with a stay-at-home order. JAMA Netw. Open 3, e2011102 (2020).PubMed 
    PubMed Central 

    Google Scholar 
    71.Lyu, W. & Wehby, G. L. Community use of face masks and COVID-19: Evidence from a natural experiment of state mandates in the US. Health Aff. 39, 1419 (2020).
    Google Scholar 
    72.Zhang, R., Li, Y., Zhang, A. L., Wang, Y. & Molina, M. J. Identifying airborne transmission as the dominant route for the spread of COVID-19. PNAS 117, 14857 (2020).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    73.Fokas, A. S., Athanassios, S., Jesus, C.-M. & Panayotis, G. K. A quantitative framework for exploring exit strategies from the COVID-19 lockdown. Chaos Solitons Fractals 140, 110244 (2020).MathSciNet 
    CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    74.Olumoyin, K. D., Khaliq, A. Q. M., & Furati, F. M. A quantitative framework for exploring exit strategies from the COVID-19 lockdown. arXiv:2104.02603 (2021).75.Tam, K.-M., Walker, N. & Moreno, J. Effect of mitigation measures on the spreading of COVID-19 in hard-hit states in the U.S.. PLoS ONE 15, e0240877 (2020).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    76.Tam, K.-M., Walker, N., & Moreno, J. Projected Development of COVID-19 in Louisiana. arXiv:2004.02859 (2020).77.Marchant, R., Samia, N. I., Rosen, O., Tanner, M. A., & Cripps, S. Learning as we go: An examination of the statistical accuracy of COVID19 daily death count predictions. arXiv:2004.04734 (2020).78.Wu, Z. & McGoogan, J. M. Characteristics of and important lessons from the coronavirus disease 2019 (COVID-19) outbreak in China. JAMA 323, 1239 (2020).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    79.Mizumoto, K., Kagaya, K., Zarebski, A. & Chowell, G. Estimating the asymptomatic proportion of coronavirus disease 2019 (COVID-19) cases on board the Diamond Princess cruise ship, Yokohama, Japan, 2020. Eurosurveill 25, 10 (2020).
    Google Scholar 
    80.
    https://github.com/nytimes/covid-19-data
    81.Holmdahl, I. & Buckee, C. Wrong but useful—What covid-19 epidemiologic models can and cannot tell us. N. Engl. J. Med. 383, 303 (2020).CAS 
    PubMed 

    Google Scholar 
    82.
    https://www.cdc.gov/coronavirus/2019-ncov/prevent-getting-sick/cloth-face-cover-guidance.html
    83.
    https://www.cdc.gov/mmwr/volumes/69/wr/mm695152a8.htm
    84.Abedi, V. et al. Racial, economic, and health inequality and COVID-19 infection in the United States. J. Racial Ethnic Health Disparities 8, 732 (2021).
    Google Scholar 
    85.Merow, C. & Urban, M. C. Seasonality and uncertainty in global COVID-19 growth rates. PNAS 117(44), 27456 (2020).CAS 
    PubMed 
    PubMed Central 

    Google Scholar 
    86.Carlson, C. J., Gomez, A. C. R., Bansal, S. & Ryan, S. J. Misconceptions about weather and seasonality must not misguide COVID-19 response. Nat. Comm. 11, 412 (2020).
    Google Scholar 
    87.Burra, P. et al. Temperature and latitude correlate with SARS-CoV-2 epidemiological variables but not with genomic change worldwide. Evol. Bioinform. https://doi.org/10.1177/1176934321989695 (2021).Article 

    Google Scholar  More

  • in

    Grape expectations: making Australian wine more sustainable

    Download PDF

    This photograph was taken at the Angullong estate in New South Wales, Australia, which hosts some of my field trials. The aim is to study sustainable agriculture in vineyards. You have to dodge the odd brown snake, but, as offices go, this one — among the grapevines of such a picturesque part of the world — makes my job quite a privilege.It’s a November evening, which is springtime here in the Southern Hemisphere, and this time of year is when pests such as the light brown apple moth (Epiphyas postvittana) start to emerge. That means that ecologists such as myself, as well as the commercial winemakers we collaborate with, move into data-capture mode to track the presence of the insects. These moths produce multiple generations every year, so they can be quite numerous by harvest time, and can cause real damage by getting into the grapes.We’re conducting experiments to see whether positioning various plant species between and under grapevines can help to reduce the population of pests by encouraging their predators. Parasitoid wasps, for example, target the eggs of light brown apple moths, injecting them with their own eggs. When the wasp larvae hatch, they eat the moth larvae from the inside out. Although quite gruesome, parasitoid wasps could provide an environmentally friendly way to control moth populations.In my laboratory at Charles Sturt University in Orange, we’re incubating moth eggs that we then put on special cards in the vineyard. Because parasitoids love nectar, we expect to see more attacks on the moth eggs in areas where we’ve planted flowering shrubs than in the control areas, where grass predominates. We collect the cards after about 48 hours in the field, and incubate the moth eggs to measure the level of parasitism. In the next couple of years, with more data, we hope to identify the optimum mix of plant species to manage pests without resorting to chemicals.

    Nature 602, 176 (2022)
    doi: https://doi.org/10.1038/d41586-022-00218-z

    Related Articles

    ‘For a brown invertebrate’: rescuing native UK oysters

    Breeding the sweetest biofuels in the business

    An IPCC reviewer shares his thoughts on the climate debate

    Collection: Fieldwork

    Subjects

    Careers

    Ecology

    Agriculture

    Latest on:

    Careers

    A nutritionist in Kenya shares advice for prospective students
    Career Q&A 31 JAN 22

    Why early-career researchers should step up to the peer-review plate
    Career Feature 31 JAN 22

    I’m a lip-reading scientist: here’s how I can discuss science with you
    Career Column 28 JAN 22

    Ecology

    Richard Leakey (1944–2022)
    Obituary 28 JAN 22

    Emphasizing declining populations in the Living Planet Report
    Matters Arising 26 JAN 22

    Reply to: Do not downplay biodiversity loss
    Matters Arising 26 JAN 22

    Agriculture

    A hard graft problem solved for key global food crops
    News & Views 25 JAN 22

    Countries should boycott Brazil over export-driven deforestation
    Correspondence 18 JAN 22

    From the archive
    News & Views 18 JAN 22

    Jobs

    Postdoctoral Research Assistant

    Queen Mary University of London (QMUL)
    London, United Kingdom

    Faculty Position in Environmental Sensing Technologies Joint Appointement between the Swiss Federal Laboratories for Materials Science and Technology (Empa) and the Ecole polytechnique fédérale de Lausanne (EPFL)

    Swiss Federal Institute of Technology in Lausanne (EPFL)
    Dübendorf and Lausanne, Switzerland

    Private Secretary, Chief Scientist

    Alan Turing Institute
    London, United Kingdom

    Research Fellow/Senior Research Fellow – Mathematical Modeller/Bioinformatician

    University College London (UCL)
    London, United Kingdom More