Open access
Bacteriology
Opinion/Hypothesis
13 September 2023

High-throughput functional trait testing for bacterial pathogens

ABSTRACT

Functional traits are characteristics that affect the fitness and metabolic function of a microorganism. There is growing interest in using high-throughput methods to characterize bacterial pathogens based on functional virulence traits. Traditional methods that phenotype a single organism for a single virulence trait can be time consuming and labor intensive. Alternatively, machine learning of whole-genome sequences (WGS) has shown some success in predicting virulence. However, relying solely on WGS can miss functional traits, particularly for organisms lacking classical virulence factors. We propose that high-throughput assays for functional virulence trait identification should become a prominent method of characterizing bacterial pathogens on a population scale. This work is critical as we move from compiling lists of bacterial species associated with disease to pathogen-agnostic approaches capable of detecting novel microbes. We discuss six key areas of functional trait testing and how advancing high-throughput methods could provide a greater understanding of pathogens.

OPINION/HYPOTHESIS

Classifying bacterial organisms as pathogens, non-pathogens, and commensals often neglects the magnitude of the host response. Although it has been recognized that all microbes have some pathogenic potential (1), we generally refer to bacterial pathogens as organisms that cause disease in a host. To cause disease, bacterial pathogens use a variety of factors to colonize, evade, and overcome the host response. Classically, Koch’s postulates and molecular Koch’s postulates have been used to understand the pathogenesis and elucidate which factors pathogens use to cause disease (2).
Microbial functional traits are the measurable characteristics that affect organism fitness, performance, and behavior under certain conditions (3 5). Functional traits may be produced by the bacterium itself (e.g., toxin production), by the interaction of the bacterium with the host (e.g., cell death), or by the environment (e.g., competition with other microbes). Traits of individual bacteria can be difficult to identify in complex systems and can be transferred between bacteria by horizontal gene transfer, mainly when encoded by plasmids (6). For species within communities, functional traits contain information on environmental constraints, phylogenetic signals, and physiological function (4, 7, 8).
Traditionally, microbiologists analyzed functional traits based on phenotypic tests; however, high-throughput sequencing has enabled extensive efforts to infer functional traits from genomes. Because phenotypic trait testing is laborious and some microorganisms are challenging to grow in culture, data about microbial phenotypic traits are more scarcer than those about genotypic traits. Therefore, to thoroughly characterize bacterial pathogens, we must better understand how genotype translates to a functional trait and consider enzymes and morpho-physio-phenological traits as functional traits (9).
Over the past three decades, our ability to sequence DNA and RNA has increased exponentially (10). However, our ability to understand what the sequences infer is limited due to incomplete and constrained genome annotation. Furthermore, the physical, chemical, and biological dynamics that occur when a pathogenic bacterium, a conducive environment, and a susceptible host (e.g., plant, animal, and human) interact has not kept pace. Genes from newly sequenced genomes are typically annotated for function based on sequence similarity to characterized proteins, but the sheer number of possible proteins limits the accuracy of prediction. Databases now contain many proteins classified as hypothetical with unknown functions (11). A recent report from the U.S. Department of Energy’s Office of Biological and Environmental Research identified a need to “interrogate and characterize microbes and microbial communities at a scale and pace that matches genome sequence production” to enable predictive understanding of the behavior of newly discovered and emerging microbes (12). It would be a formidable challenge to characterize all functional traits of a pathogen. Therefore, in addition to genome annotation, validation of critical functional traits by high-throughput screens would improve our ability to link genomic information to pathogenic traits. Here, we describe some non-exhaustive examples of functional testing technologies that are providing insights into bacterial pathogens. Thus, this opinion article focuses on understanding recent advances in characterizing functional traits of pathogens and critical gaps in six key areas that are hallmarks of the infection process, including (i) competition, (ii) antibiotic resistance, (iii) adhesion and invasion, (iv) toxin production, (v) evasion of the immune system, and (vi) induction of cell death (Fig. 1).
Fig 1
Fig 1 Six key functional traits of bacterial pathogens that are used during infection. Clinical, environmental, and food are common sample types for pathogen testing. From these sample types, pathogens can be isolated in pure culture on agar plates or by using devices such as cell sorters and microfluidic platforms. Methods for high-throughput trait testing utilize 96-well plates, liquid handlers, rapid detection systems (e.g., optical imaging and electrochemical), reporter cell lines, microfluidics, and miniature tissues. Pathogens use some or all traits to cause disease including, competition, antibiotic resistance, adhesion and invasion, toxin production, evasion of the immune system, and induction of cell death.

COMPETITION

During the introduction to a host, pathogens must use functional traits to outcompete existing microbes for nutrients and niches to establish infection and colonize host tissue. Competitive exclusion is a dominant principle by which existing microbes inhibit pathogen colonization (13). Depending on the ecosystem, several factors, such as the fitness of the current members, the niche, and arrival time, can influence competition between microbes (14). We compared the advantages and limitations of conventional laboratory, genetic, and high-throughput approaches for assessing competition and other functional traits as described in Table 1. Genetic methods such as genome-scale models enable the prediction of bacterial fitness and were used to analyze the phenotypic potential of Escherichia coli genotypes (15). Traditionally in the laboratory, competition has been measured using a few strains, often a single wild-type versus a single mutant strain, and reported as the competitive index (16). The traditional competitive index is a low-throughput assay calculated as the change in the ratio of the strains after mixture and growth together (17). Previous studies have used an in vitro approach to assess the fitness of E. coli strains over a few generations and up to 60,000 generations (18, 19). To increase the throughput, mutagenesis methods using transposon libraries with sequencing have been applied to multiple strains in competitive index assays and identified several genes required for virulence, functional redundancy, and functional independency of virulence factors (20). More recently, transposon-insertion sequencing has moved from a simple growth-based selection assay to an assessment of functional traits important for outcompeting other microbes on a scale of ~107 mutant strains (21 23).
TABLE 1
TABLE 1 Comparison of technologies for testing functional traits in bacterial pathogens
TraitAssay typeRepresentative assayTime to result (h)ThroughputResolves novel microbe or traitAdvantagesLimitationsRef.
CompetitionConventionalIn vitro fitness screen2412 TestsYesDefinitively identifies novel microbial traitsDoes not identify the microbe; requires labor-intensive genetic modifications(18)
GeneticGenome-scale models168One testYesDefinitively identifies novel microbial genetic traitsDoes not predict microbial phenotype(15)
High throughputInterbacterial competition screen2496 TestsYesDefinitively identifies novel microbial traitsAssay requires robotics which are expensive(24)
Antibiotic resistanceConventionalDisc diffusion2412 TestsNoRapid sample to answerDoes not identify the microbe(25)
GeneticPCR Xpert (Cepheid)696 TestsNoRapid sample to answerTest panels do not identify engineered or emerging microbes(26)
High throughputDirect-on-target microdroplet MALDI-TOF MS≤696 TestsNoRapid sample to answerIdentification of the microbe is possible, but the microbial phenotype is not predicted(27)
Adhesion and invasionConventionalIn vitro adhesion and invasion assay7212 TestsYesDefinitively identifies novel microbial traitsDoes not identify the microbe(28)
GeneticMultiplex PCR for adhesins and invasins696 TestsNoIdentifies phenotype of interestDoes not identify the microbe(29)
High throughputVirtual colony counting infection assay4896 TestsYesDefinitively identifies novel microbial traitsDoes not identify the microbe(30)
Toxin productionConventionalToxin activity on cell monolayers48Six testsYesDefinitively identifies novel microbial traitsDoes not identify the microbe(31)
GeneticPCR of known toxin genes696 TestsNoIdentifies phenotype of interestDoes not identify the microbe(32)
High throughputAutomated patch clamp platform48384 TestsNoIdentifies phenotype of interestDoes not identify the microbe(33)
Immune system evasionConventionalAnimal model16824 TestsNoIdentifies phenotype of interestLabor and cost intensive; ethical concerns(34)
GeneticGenome search for effectors tool24One testNoIdentifies phenotype of interestRequires bioinformatics knowledge base; optimized for fungi; pipeline would need to be optimized for bacteria(35)
High throughputOrgan-on-a-chip16812 TestsNoIdentifies phenotype of interestLabor intensive(36)
Cell deathConventionalDye exclusion test2496 TestsNoRapid sample to answerDoes not identify the microbe(37)
GeneticqPCR for apoptotic nucleic acids696 TestsNoRapid sample to answerDoes not identify the microbe(38)
High throughputReal-time fluorogenic DNA dyes696 TestsNoRapid sample to answerDoes not identify the microbe(39)
Competition and bacterial growth are often viewed to have a significant relationship. Atolia et al. found that noise minimization is critical for assessing growth by a high-throughput screen and that consistent growth when inoculating many cultures from bacterial colonies grown on agar plates is challenging (40). Automated robotic colony-picking systems may reduce the challenge of inoculating several cultures starting from bacterial colonies (41). To assess competition between microbes in a functional test, a high-throughput interbacterial competition assay enabling testing of 96 competition assays simultaneously was developed for Agrobacterium tumefaciens resulting in the observation that A. tumefaciens could kill other bacteria (24). This high-throughput interbacterial competition assay requires common laboratory materials such as 96-well plates but also uses an automated pipetting system (24) and could be expanded to other microbes. Other phenotypic high-throughput screening technologies, such as the Omnilog, have made it possible to investigate nearly 2,000 phenotypes related to nutrient competition (42). A limitation of these systems is that they measure the growth of the heterogeneous bacterial population. To overcome the issue of analyzing populations, novel methods based on microfluidic platforms have now made it possible to independently evaluate and track single cells on a scale of more than 105 parallel cell lineages (43).
An additional consideration for high-throughput assessment is that the relationship between pathogenicity and growth may not be easily predicted. For example, when evaluating 61 human bacterial pathogens, the growth rate was negatively related to virulence (44). The growth rate may be considered a limitation for competition assays. Some bacterial pathogens are more amenable to high-throughput characterization because of growth characteristics, containment procedures, biosafety considerations, and ease of equipment sterilization between tests. To advance high-throughput competition assays, standardized systems should be developed that allow rapid assessment of multiple strains, standardized consortia of microorganisms relevant for different environments or host sites, and defined metrics for competition.

ANTIBIOTIC RESISTANCE

Antibiotic resistance allows microbes to colonize environments with antibiotic stressors (45). In recent years, there has been an increase in infections caused by antibiotic-resistant bacteria (46). The interplay of bacterial virulence and antibiotic resistance is complex and depends on factors associated with the microbe and the environment (47). Traditionally, antibiotic susceptibility testing is performed using several cultivation rounds of a single isolate, and accepted breakpoint values are evaluated to determine whether the microorganism is susceptible or resistant (48). Recently, Yang et al. (49) developed a phenotype-based threat assessment pipeline characterizing bacterial pathogens for adherence, toxicity, immune activation, and antibiotic resistance. This previous study developed capabilities using machine learning with phenotypic data to assess pathogenic potential, and bacterial antibiotic resistance was assessed using traditional disk diffusion assays. Traditional methods such as disk diffusion assays can only test a few antibiotics (e.g., eight antibiotics) per agar plate and rely on isolating bacterial organisms in pure culture, which poses a bottleneck to antibiotic susceptibility testing (25). Thus, nucleic acid amplification and whole-genome sequencing (WGS) technologies are used in combination with functional testing. Commercial nucleic acid amplification kits exist for testing a wide range of antibiotics (26). However, caution should be used when interpreting genetic information as some studies have shown overall low sensitivity but high specificity for detecting antimicrobial resistance by nucleic acid amplification methods (50). Additionally, virulence plasmids containing antibiotic-resistant genes are being more widely reported, which can impact treatment and determine whether specific plasmids should be monitored to limit their spread (51, 52). Functional testing for antibiotic resistance can help link the antibiotic-resistant genes found on virulence plasmids.
Functional testing is critical to determine how bacterial organisms respond to new compounds, and which existing compounds are effective against novel strains. No single antimicrobial susceptibility testing technology is broadly accepted and globally accessible for rapid, high-throughput testing (53). In conjunction, there has been a lack of newly developed antibiotics despite the rise of robust screening methods for new drugs and drug combinations (54, 55). Although the use of artificial intelligence-driven discovery has drastically decreased the number of compounds needed to test (56), it remains challenging to test multiple compounds on several microorganisms simultaneously. Identification of effective compounds is critical because approximately 50% of antibiotic treatments begin with the wrong antibiotic without diagnosing the pathogen (48).
The traditional cultivation approach for testing the function of antibiotic resistance is time consuming and labor intensive. Thus, high-throughput methods are being developed to accelerate time-to-result and increase the diversity of cultivated bacterial organisms able to be tested. Recent advances in technology have relied on building devices using microfluidics and lab-on-a-disc systems with capabilities to test the growth of up to 100 bacterial strains within droplets (57 59). Another approach uses advances in matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) to rapidly screen functional sets of antibiotic resistance by direct-on-target microdroplet growth assays at a scale of 96 samples per assay (27, 60). The current challenge these assays face is increasing throughput and accessibility for routine testing. In addition, integrating different data types, such as MALDI-TOF MS and WGS, can be challenging. For integrating various data types, existing databases such as the Antimicrobial Testing Leadership and Surveillance, which provides open access to minimal inhibitory concentration, should be leveraged and integrated with genomic information (61).

ADHESION AND INVASION

Adhesion is an important feature of bacterial pathogens that allows microbes to colonize hosts, induce cellular responses, and manipulate host signaling. To adhere to cells, bacteria use a range of factors, from pili, fimbria, flagella, and other adhesins (62 64). Conventional adherence assays are laborious and based on counting bacteria on agar plates or from stained cells (28, 65). Genetic methods, such as multi-plex PCR, test for the presence of multiple adhesin genes in a single reaction but do not provide evidence of functionality (29). Some high-throughput assays have been developed to quantify the extent of bacterial adhesion on host cells. For example, flow cytometry-based adherence assays that quantify interactions between bacteria and eukaryotic cells have been designed for fast and reproducible measurements (49, 66, 67). More recently, fluorescently tagged strains, bacteria carrying fluorescence proteins on plasmids, and live fluorescent stains have been used to quantify the level of adherence with a throughput of 96 samples per assay (49, 66, 67). Another method uses virtual colony counts, an absorbance-based measurement showing a good correlation with traditional plate-based colony counts for Salmonella with multiple eukaryotic cell lines (30). In addition, colony counting by high-throughput screens has enabled the determination of viable bacteria cell numbers in 96-well plates (68). Extensive washing to remove unbound bacteria and fixative treatment remains issues for high-throughput and traditional assays (66). To resolve finer-scale binding, host ligands can also be screened in high-throughput assays (69).
Certain pathogens also invade host cells and have an intracellular lifestyle. Along with conventional adherence assays, the laborious nature of invasion assays can be prohibitive to test multiple conditions and time points. Classically, the bacterial invasion has been quantified by a gentamicin protection assay in which internalized bacteria avoid being killed by gentamicin and can be enumerated (70). For rapid assessment, a screening system for invasion with a fluorescently tagged Salmonella strain was assessed using HeLa cells, enabling 24 samples to be tested per assay with applications for chemical libraries and potential drug testing (71). Similarly to adherence assays, the virtual colony counting high-throughput method has been applied to study Salmonella invasion, which can test 96 samples per assay (30). Tools that can characterize multiple functional traits, such as the virtual colony counting method, have greater utility.

TOXIN PRODUCTION

Several bacterial pathogens use protein toxins to disrupt signaling, degrade biochemicals, and damage host tissue to establish infection (72). A plethora of toxins have been described in a variety of bacterial species. Toxins are not simply destructive tools but may contribute to survival and escape from environmental unicellular eukaryotes (73). Several methods exist for the specific detection of clinically important toxins, such as enzyme-linked immunoassay (ELISA), lateral flow tests, western blots, cell culture, and mass spectrometry (74). However, methods to simultaneously characterize known toxins, novel toxins, toxin potency, and integrate data from other functional traits and genomic information remain underexplored.
Traditionally, bacterial toxins were discovered and functionally characterized based on observing bacterial culture filtrates causing eukaryotic cell disruption or death (31). Although bacterial toxins are critical virulence factors, new toxins are continually being discovered. For example, the pore-forming toxin exolysin (ExlA) was initially described in 2014 from a virulent strain of Pseudomonas aeruginosa causing hemorrhagic pneumonia (75). For known toxins, selective agars containing chromogenic substrates that are cleaved by toxins have been developed to differentiate strains that produce active toxins from non-producing strains (32). The throughput of using chromogenic media is often limited to one strain per agar plate and can be time consuming based on the incubation time of the bacterial strains. In conjunction, genetic methods such as PCR are often used for follow-up testing to characterize and subtype toxins which can increase the time-to-result. Also, PCR often does not provide an independent result for function but rather only tests for the presence of the gene (32). To our knowledge, high-throughput screens have primarily been developed for chemical toxins and not as robustly for bacterial toxins (76, 77). However, automated patch clamp platforms exist that measure ionic current and the state of voltage-gated ion channels (e.g., open or closed) that bacterial toxins often act upon and can be used for functional measurements. Automated patch clamp platforms such as the SyncroPatch is a high-throughput platform that can assess 384 and 768 samples and was applied to detecting tetrodotoxin produced by pathogenic bacteria such as Pseudomonas and Vibrio species (33). Although the automated patch clamp throughput is considerably higher than selective media, the construction and stable expression of channels in reporter cell lines that the bacterial toxins act on are limiting factors.

EVASION OF THE IMMUNE SYSTEM

Many pathogens have developed defenses to evade the host innate immune system. Some pathogenic bacteria directly inject proteins termed effectors into target host cells via specialized secretion systems across the bacterial and host membranes to manipulate host cell functions. One of the best characterized secretion systems is the type III secretion system (T3SS) (78). T3SSs inject effector proteins that are diverse and specific to pathogens to induce pathogenic mechanisms, specifically immune system subversion. Pathogens use T3SSs effectors to evade host immunity in several ways, such as activating host-signaling cascades and pattern-recognition receptors, and suppressing evasion of innate and adaptive defenses (79).
There is a need for efficient methods to identify effectors in pathogenic bacteria. The Genome Search for Effectors Tool (GenSET) can predict effector sequences in bacteria (35). This tool can provide information for researchers to conduct downstream wet bench experimental validation; however, GenSET has low prediction rates. This could be caused by the various families of T3SSs found in different species, as heterologous effectors may yield other attributes when applied to specific microbes. Low prediction rates can provide inaccurate results, hence, the need for downstream validation (35). Improved computational approaches and machine-learning algorithms are needed to identify novel effectors from unannotated nucleotide and protein sequences accurately.
In the laboratory, animal models are often used to test the ability of bacterial factors to evade the innate immune system (34). In contrast, in vitro platforms are more cost-effective and rapid. There are several reporter cell lines for detecting immune system activation such as the HEK-Blue TLR2, TLR3, TLR4, TLR5, TLR7, TLR8, and TLR9 cells (InvivoGen) or the nuclear factor κB reporter line used previously (49). For the detection of immune system evasion, it is vital to characterize bacterial pathogens in relevant in vitro systems such as co-culture of epithelial and immune cells. Recent studies investigated “gut-on-chips” to simulate structure and function as an attempt to replicate in vivo microenvironments (80). These tissue chips are a promising approach for studying microbe-host interactions in a more high-throughput screen than animal testing. They can incorporate mononuclear phagocytes to respond to commensal or pathogenic bacteria in a 3D configuration. A challenge with co-culturing cell types with different tissues is that the optimum conditions for each cell type may differ, leading to inaccurate real-time interactions. Other challenges include immune cell adherence, material compatibility, selection of extracellular matrix (ECM), and immune cell migration through ECM (81). However, organ-on-chip devices provide a promising avenue of research for understanding epithelial and immune cell responses to potential pathogens.

INDUCTION OF CELL DEATH

During infection, host cells will respond to a pathogen in various ways, including cell death, to remove the infected cell from the host. Cell death has regulated pathways to initiate death and various morphological and molecular changes in the cell characterize each pathway. Some common mechanisms for cell death include apoptosis, necrosis, and pyroptosis, with the most characterized being apoptosis (82). Apoptosis is a non-inflammatory programmed cell death type characterized by changes in cell morphology, such as membrane blebbing, cell shrinkage, and DNA fragmentation. Banfalvi reviewed several assays that detect apoptosis related to structural and functional changes (83). In addition, non-programmed cell death can occur from infection (84).
Cell death assays have been developed for high-throughput screens. Cummings et al. reviewed these methodologies in detail (85). However, most assays have been designed from a drug discovery perspective rather than to assess cell death from pathogens. Drug discovery assays can determine cell death and what pathways are activated (85), but they should be more widely considered for characterizing bacterial pathogens. In addition to antibiotic resistance, immune activation, and adherence, the platform developed by Yang et al. (49) tested bacterial-induced cell death by using cell staining and flow cytometry.
Pathogenic bacteria can activate several cellular pathways. We previously engineered a fluorescent reporter lung cell line that signals when the protein kinase ERK (extracellular signal-regulated kinase) and transcription factor Fra1 (FOS-related antigen 1) pathway are activated or inhibited (86). The change in fluorescent signal occurs before the stress response/cytopathic effects of the host cell. This high-throughput image-based assay allows for functional screening of cell health with the incubation of a live bacterial strain. A limitation of this assay is that not all pathogenic bacteria can inhibit the ERK-Fra1 signaling pathway. There is an opportunity to increase the number of fluorescent reporter pathways for cell death and cell types to include a broader range of pathogens using this approach (86).

LIMITATIONS AND CONCLUDING REMARKS

Linking and integration of genotype to phenotype will enhance pathogen characterization. The amount of genetic sequence data so far surpasses functional trait data for bacterial pathogens. Thus, high-throughput functional assays are needed to keep pace with genetic sequence data. Recently, PathEngine used an integrated strategy of a phenotype-based pipeline to assess pathogenic potential, which included adherence, toxicity, antibiotic resistance, and innate immune activation (49). A significant hurdle with harnessing functional assay testing data is integrating information generated from these assays with existing information such as sequence, transcriptomic, proteomic, and metabolomic data. To harness this data, robust and agile databases are needed. As proposed previously, multiomics integration to identify pathogen-agnostic signatures of disease could detect potential pathogens without prior knowledge of the microbe (87).
Another limitation is that the integration of pathogen functional data is challenging because it can be unclear how individual proteins work as part of a global infection model. Therefore, integrating multiple bacterial functional traits to understand virulence has not been robustly developed. Developing strategies to use multiple pieces of information, such as WGS, proteomics, and the functional tests discussed in this work, will give rise to a more detailed understanding of classifying known and novel organisms for their pathogenic potential.

ACKNOWLEDGMENTS

The research described in this paper was conducted at Pacific Northwest National Laboratory, a multiprogram national laboratory operated by Battelle Memorial Institute for the U.S. Department of Energy. Battelle Memorial Institute operates Pacific Northwest National Laboratory for the U.S. DOE under Contract DE-AC05-76RL01830.
The figure was created with BioRender.com.

REFERENCES

1.
Casadevall A, D’Orazio SEF. 2022. Expanding the pathogenic potential concept to incorporate fulminancy, time, and virulence factors. mSphere 7:e0102121.
2.
Falkow S. 2004. Molecular Koch’s postulates applied to bacterial pathogenicity—a personal recollection 15 years later. Nat Rev Microbiol 2:67–72.
3.
Yang Y. 2021. Emerging patterns of microbial functional traits. Trends Microbiol 29:874–882.
4.
Violle C, Navas ML, Vile D, Kazakou E, Fortunel C, Hummel I, Garnier E. 2007. Let the concept of trait be functional! Oikos 116:882–892.
5.
Krause S, Le Roux X, Niklaus PA, Van Bodegom PM, Lennon JT, Bertilsson S, Grossart H-P, Philippot L, Bodelier PLE. 2014. Trait-based approaches for understanding microbial biodiversity and ecosystem functioning. Front Microbiol 5:251.
6.
Schjørring S, Struve C, Krogfelt KA. 2008. Transfer of antimicrobial resistance plasmids from Klebsiella pneumoniae to Escherichia coli in the mouse intestine. J Antimicrob Chemother 62:1086–1093.
7.
He N, Liu C, Piao S, Sack L, Xu L, Luo Y, He J, Han X, Zhou G, Zhou X, Lin Y, Yu Q, Liu S, Sun W, Niu S, Li S, Zhang J, Yu G. 2019. Ecosystem traits linking functional traits to macroecology. Trends Ecol Evol 34:200–210.
8.
Kraft NJB, Cornwell WK, Webb CO, Ackerly DD. 2007. Trait evolution, community assembly, and the phylogenetic structure of ecological communities. Am Nat 170:271–283.
9.
Escalas A, Hale L, Voordeckers JW, Yang Y, Firestone MK, Alvarez-Cohen L, Zhou J. 2019. Microbial functional diversity: from concepts to applications. Ecol Evol 9:12000–12016.
10.
Koboldt DC, Steinberg KM, Larson DE, Wilson RK, Mardis ER. 2013. The next-generation sequencing revolution and its impact on genomics. Cell 155:27–38.
11.
Consortium TU. 2021. UniProt: the universal protein knowledgebase in 2021. Nucleic Acids Res 49:D480–D489.
12.
Buell R, Deutschbauer A, Adin D, Ronning C. 2019. Breaking the bottleneck of genomes: understanding gene function across taxa. Workshop Report United States.
13.
Lange ME, Uwiera RRE, Inglis GD. 2022. Enteric Escherichia Coli O157:H7 in cattle, and the use of mice as a model to elucidate key aspects of the host-pathogen-microbiota interaction: a review. Front Vet Sci 9:937866.
14.
Segura Munoz RR, Mantz S, Martínez I, Li F, Schmaltz RJ, Pudlo NA, Urs K, Martens EC, Walter J, Ramer-Tait AE. 2022. Experimental evaluation of ecological principles to understand and modulate the outcome of bacterial strain competition in gut microbiomes. ISME J 16:1681–1682.
15.
Chen K, Anand A, Olson C, Sandberg TE, Gao Y, Mih N, Palsson BO. 2021. Bacterial fitness landscapes stratify based on proteome allocation associated with discrete aero-types. PLoS Comput Biol 17:e1008596.
16.
Krypotou E, Scortti M, Grundström C, Oelker M, Luisi BF, Sauer-Eriksson AE, Vázquez-Boland J. 2019. Control of bacterial virulence through the peptide signature of the habitat. Cell Rep 26:1815–1827.
17.
Taylor RK, Miller VL, Furlong DB, Mekalanos JJ. 1987. Use of phoA gene fusions to identify a pilus colonization factor coordinately regulated with cholera toxin. Proc Natl Acad Sci U S A 84:2833–2837.
18.
Linkevicius M, Anderssen JM, Sandegren L, Andersson DI. 2016. Fitness of Escherichia coli mutants with reduced susceptibility to tigecycline. J Antimicrob Chemother 71:1307–1313.
19.
Lenski RE. 2017. Experimental evolution and the dynamics of adaptation and genome evolution in microbial populations. ISME J 11:2181–2194.
20.
Karlinsey JE, Stepien TA, Mayho M, Singletary LA, Bingham-Ramos LK, Brehm MA, Greiner DL, Shultz LD, Gallagher LA, Bawn M, Kingsley RA, Libby SJ, Fang FC. 2019. Genome-wide analysis of salmonella enterica serovar Typhi in humanized mice reveals key virulence features. Cell Host Microbe 26:426–434.
21.
Cain AK, Barquist L, Goodman AL, Paulsen IT, Parkhill J, van Opijnen T. 2020. A decade of advances in transposon-insertion sequencing. Nat Rev Genet 21:526–540.
22.
Nolan LM, Whitchurch CB, Barquist L, Katrib M, Boinett CJ, Mayho M, Goulding D, Charles IG, Filloux A, Parkhill J, Cain AK. 2018. A global genomic approach uncovers novel components for twitching motility-mediated biofilm expansion in Pseudomonas aeruginosa. Microb Genom 4:e000229.
23.
Kakkanat A, Phan M-D, Lo AW, Beatson SA, Schembri MA. 2017. Novel genes associated with enhanced motility of Escherichia coli ST131. PLoS One 12:e0176290.
24.
Lin H-H, Yu M, Sriramoju MK, Hsu S-T, Liu C-T, Lai E-M. 2019. A high-throughput Interbacterial competition screen identifies ClpAP in enhancing recipient susceptibility to type VI secretion system-mediated attack by Agrobacterium tumefaciens. Front Microbiol 10:3077.
25.
Gajic I, Kabic J, Kekic D, Jovicevic M, Milenkovic M, Mitic Culafic D, Trudic A, Ranin L, Opavski N. 2022. Antimicrobial susceptibility testing: a comprehensive review of currently used methods. Antibiotics (Basel) 11:427.
26.
Yee R, Fisher S, Bergman Y, Chambers KK, Tamma PD, Carroll KC, Simner PJ. 2021. Combined selective culture and molecular methods for the detection of carbapenem-resistant organisms from fecal specimens. Eur J Clin Microbiol Infect Dis 40:2315–2321.
27.
Li R, Tang H, Xu H, Ren Y, Li S, Shen J. 2021. Direct-on-target microdroplet growth assay applications for clinical antimicrobial susceptibility testing. Infect Drug Resist 14:1423–1425.
28.
Bucior I, Tran C, Engel J. 2014. Assessing Pseudomonas virulence using host cells. Methods Mol Biol 1149:741–755.
29.
Frömmel U, Lehmann W, Rödiger S, Böhm A, Nitschke J, Weinreich J, Groß J, Roggenbuck D, Zinke O, Ansorge H, Vogel S, Klemm P, Wex T, Schröder C, Wieler LH, Schierack P. 2013. Adhesion of human and animal Escherichia coli strains in association with their virulence-associated genes and phylogenetic origins. Appl Environ Microbiol 79:5814–5829.
30.
Hoffmann S, Walter S, Blume A-K, Fuchs S, Schmidt C, Scholz A, Gerlach RG. 2018. High-throughput quantification of bacterial-cell interactions using virtual colony counts. Front Cell Infect Microbiol 8:43.
31.
Konowalchuk J, Speirs JI, Stavric S. 1977. Vero response to a cfytotoxin of Escherichia coli. Infect Immun 18:775–779.
32.
Darkoh C, Dupont HL, Kaplan HB. 2011. Novel one-step method for detection and isolation of active-toxin-producing Clostridium difficile strains directly from stool samples. J Clin Microbiol 49:4219–4224.
33.
Li T, Lu G, Chiang EY, Chernov-Rogan T, Grogan JL, Chen J, Pignataro G. 2017. High-throughput electrophysiological assays for voltage gated ion channels using SyncroPatch 768PE. PLoS ONE 12:e0180154.
34.
Freire CA, Silva RM, Ruiz RC, Pimenta DC, Bryant JA, Henderson IR, Barbosa AS, Elias WP. 2022. Secreted autotransporter toxin (sat) mediates innate immune system evasion. Front Immunol 13:844878.
35.
Hobbs CK, Porter VL, Stow MLS, Siame BA, Tsang HH, Leung KY. 2016. Computational approach to predict species-specific type III secretion system (T3SS) effectors using single and multiple genomes. BMC Genomics 17:1048.
36.
Feaugas T, Sauvonnet N. 2021. Organ-on-chip to investigate host-pathogens interactions. Cell Microbiol 23:e13336.
37.
Strober W. 2015. Trypan blue exclusion test of cell viability. Curr Protoc Immunol 111:A3.
38.
Hooker DJ, Mobarok M, Anderson JL, Rajasuriar R, Gray LR, Ellett AM, Lewin SR, Gorry PR, Cherry CL. 2012. A new way of measuring apoptosis by absolute quantitation of inter-nucleosomally fragmented genomic DNA. Nucleic Acids Res 40:e113.
39.
Kepp O, Galluzzi L, Lipinski M, Yuan J, Kroemer G. 2011. Cell death assays for drug discovery. Nat Rev Drug Discov 10:221–237.
40.
Atolia E, Cesar S, Arjes HA, Rajendram M, Shi H, Knapp BD, Khare S, Aranda-Díaz A, Lenski RE, Huang KC. 2020. Environmental and physiological factors affecting high-throughput measurements of bacterial growth. mBio 11:e01378-20.
41.
Hughes SR, Riedmuller SB, Mertens JA, Li X-L, Bischoff KM, Qureshi N, Cotta MA, Farrelly PJ. 2006. High-throughput screening of cellulase F mutants from multiplexed plasmid sets using an automated plate assay on a functional proteomic robotic workcell. Proteome Sci 4:1–14.
42.
Acin-Albiac M, Filannino P, Gobbetti M, Di Cagno R. 2020. Microbial high throughput phenomics: the potential of an irreplaceable omics. Comput Struct Biotechnol J 18:2290–2299.
43.
Bakshi S, Leoncini E, Baker C, Cañas-Duarte SJ, Okumus B, Paulsson J. 2021. Tracking bacterial lineages in complex and dynamic environments with applications for growth control and persistence. Nat Microbiol 6:783–791.
44.
Leggett HC, Cornwallis CK, Buckling A, West SA. 2017. Growth rate, transmission mode and virulence in human pathogens. Philos Trans R Soc Lond B Biol Sci 372:20160094.
45.
Pan Y, Zeng J, Li L, Yang J, Tang Z, Xiong W, Li Y, Chen S, Zeng Z, Gilbert JA. 2020. Coexistence of antibiotic resistance genes and virulence factors deciphered by large-scale complete genome analysis. mSystems 5:e00821-19.
46.
Uddin TM, Chakraborty AJ, Khusro A, Zidan BRM, Mitra S, Emran TB, Dhama K, Ripon MKH, Gajdács M, Sahibzada MUK, Hossain MJ, Koirala N. 2021. Antibiotic resistance in microbes: history, mechanisms, therapeutic strategies and future prospects. J Infect Public Health 14:1750–1766.
47.
Beceiro A, Tomás M, Bou G. 2013. Antimicrobial resistance and virulence: a successful or deleterious association in the bacterial world? Clin Microbiol Rev 26:185–230.
48.
Vasala A, Hytönen VP, Laitinen OH. 2020. Modern tools for rapid diagnostics of antimicrobial resistance. Front Cell Infect Microbiol 10:308.
49.
Yang J, Eslami M, Chen Y-P, Das M, Zhang D, Chen S, Roberts A-J, Weston M, Volkova A, Faghihi K, Moore RK, Alaniz RC, Wattam AR, Dickerman A, Cucinell C, Kendziorski J, Coburn S, Paterson H, Obanor O, Maples J, Servetas S, Dootz J, Qin Q-M, Samuel JE, Han A, van Schaik EJ, de Figueiredo P. 2022. Phenotype-based threat assessment. Proc Natl Acad Sci U S A 119:e2112886119.
50.
Sigmund IK, Renz N, Feihl S, Morgenstern C, Cabric S, Trampuz A. 2020. Value of multiplex PCR for detection of antimicrobial resistance in samples retrieved from patients with orthopaedic infections. BMC Microbiol 20:88.
51.
Turton J, Davies F, Turton J, Perry C, Payne Z, Pike R. 2019. Hybrid resistance and virulence plasmids in “high-risk” clones of Klebsiella pneumoniae, including those carrying blaNDM-5. Microorganisms 7:326.
52.
Sunde M, Ramstad SN, Rudi K, Porcellato D, Ravi A, Ludvigsen J, das Neves CG, Tryland M, Ropstad E, Slettemeås JS, Telke AA. 2021. Plasmid-associated antimicrobial resistance and virulence genes in Escherichia coli in a high arctic reindeer subspecies. J Glob Antimicrob Resist 26:317–322.
53.
van Belkum A, Bachmann TT, Lüdke G, Lisby JG, Kahlmeter G, Mohess A, Becker K, Hays JP, Woodford N, Mitsakakis K, Moran-Gilad J, Vila J, Peter H, Rex JH, Dunne WM, JPIAMR AMR-RDT Working Group on Antimicrobial Resistance and Rapid Diagnostic Testing. 2019. Developmental roadmap for antimicrobial susceptibility testing systems. Nat Rev Microbiol 17:51–62.
54.
Murray EM, Allen CF, Handy TE, Huffine CA, Craig WR, Seaton SC, Wolfe AL. 2019. Development of a robust and quantitative high-throughput screening method for antibiotic production in bacterial libraries. ACS Omega 4:15414–15420.
55.
Sun W, Weingarten RA, Xu M, Southall N, Dai S, Shinn P, Sanderson PE, Williamson PR, Frank KM, Zheng W. 2016. Rapid antimicrobial susceptibility test for identification of new therapeutics and drug combinations against multidrug-resistant bacteria. Emerg Microbes Infect 5:e116.
56.
Nguyen M, Long SW, McDermott PF, Olsen RJ, Olson R, Stevens RL, Tyson GH, Zhao S, Davis JJ. 2019. Using machine learning to predict antimicrobial MICs and associated genomic features for nontyphoidal Salmonella. J Clin Microbiol 57:e01260-18.
57.
Postek W, Garstecki P. 2022. Droplet microfluidics for high-throughput analysis of antibiotic susceptibility in bacterial cells and populations. Acc Chem Res 55:605–615.
58.
Lim T, Kim E-G, Choi J, Kwon S. 2020. A high-throughput cell culture system based on capillary and centrifugal actions for rapid antimicrobial susceptibility testing. Lab Chip 20:4552–4560.
59.
Watterson WJ, Tanyeri M, Watson AR, Cham CM, Shan Y, Chang EB, Eren AM, Tay S. 2020. Droplet-based high-throughput cultivation for accurate screening of antibiotic resistant gut microbes. Elife 9:e56998.
60.
Nix ID, Idelevich EA, Storck LM, Sparbier K, Drews O, Kostrzewa M, Becker K. 2020. Detection of methicillin resistance in Staphylococcus aureus from agar cultures and directly from positive blood cultures using MALDI-TOF mass spectrometry-based direct-on-target microdroplet growth assay. Front Microbiol 11:232.
61.
Catalán P, Wood E, Blair JMA, Gudelj I, Iredell JR, Beardmore RE. 2022. Seeking patterns of antibiotic resistance in ATLAS, an open, raw MIC database with patient metadata. Nat Commun 13:2917.
62.
Pizarro-Cerdá J, Cossart P. 2006. Bacterial adhesion and entry into host cells. Cell 124:715–727.
63.
Sheng H, Lim JY, Knecht HJ, Li J, Hovde CJ. 2006. Role of Escherichia coli O157:H7 virulence factors in colonization at the bovine terminal rectal mucosa. Infect Immun 74:4685–4693.
64.
Hartmann I, Carranza P, Lehner A, Stephan R, Eberl L, Riedel K. 2010. Genes involved in Cronobacter sakazakii biofilm formation. Appl Environ Microbiol 76:2251–2261.
65.
Thakur SD, Obradovic M, Dillon J-A, Ng SH, Wilson HL. 2019. Development of flow cytometry based adherence assay for Neisseria gonorrhoeae using 5'-carboxyfluorosceinsuccidyl ester. BMC Microbiol 19:67.
66.
Zanaboni E, Arato V, Pizza M, Seubert A, Leuzzi R. 2016. A novel high-throughput assay to quantify the vaccine-induced inhibition of Bordetella pertussis adhesion to airway epithelia. BMC Microbiol 16:215.
67.
Akinsola RO, Adewoyin M, Lee C-W, Sim EU-H, Narayanan K. 2021. RFP-based method for real-time tracking of invasive bacteria in a heterogeneous population of cells. Anal Biochem 634:114432.
68.
Hazan R, Que Y-A, Maura D, Rahme LG. 2012. A method for high throughput determination of viable bacteria cell counts in 96-well plates. BMC Microbiol 12:1–7.
69.
Petrie LE, Leonard AC, Murphy J, Cox G. 2020. Development and validation of a high-throughput whole cell assay to investigate Staphylococcus aureus adhesion to host ligands. J Biol Chem 295:16700–16712.
70.
Elsinghorst EA. 1994. Measurement of invasion by gentamicin resistance, p 405–420. In Methods in Enzymology. Elsevier.
71.
Steinberg BE, Scott CC, Grinstein S. 2007. High-throughput assays of phagocytosis, phagosome maturation, and bacterial invasion. Am J Physiol Cell Physiol 292:C945–C952.
72.
Rudkin JK, McLoughlin RM, Preston A, Massey RC. 2017. Bacterial toxins: offensive, defensive, or something else altogether? PLoS Pathog 13:e1006452.
73.
Loś JM, Loś M, Węgrzyn A, Węgrzyn G. 2012. Altruism of shiga toxin-producing Escherichia coli: recent hypothesis versus experimental results. Front Cell Infect Microbiol 2:166.
74.
Duracova M, Klimentova J, Fucikova A, Dresler J. 2018. Proteomic methods of detection and quantification of protein toxins. Toxins (Basel) 10:99.
75.
Elsen S, Huber P, Bouillot S, Couté Y, Fournier P, Dubois Y, Timsit J-F, Maurin M, Attrée I. 2014. A type III secretion negative clinical strain of Pseudomonas aeruginosa employs a two-partner secreted exolysin to induce hemorrhagic pneumonia. Cell Host Microbe 15:164–176.
76.
Zhu H, Zhang J, Kim MT, Boison A, Sedykh A, Moran K. 2014. Big data in chemical toxicity research: the use of high-throughput screening assays to identify potential toxicants. Chem Res Toxicol 27:1643–1651.
77.
Renner H, Becker KJ, Kagermeier TE, Grabos M, Eliat F, Günther P, Schöler HR, Bruder JM. 2021. Cell-type-specific high throughput toxicity testing in human midbrain organoids. Front Mol Neurosci 14:715054.
78.
Galán JE. 2009. Common themes in the design and function of bacterial effectors. Cell Host Microbe 5:571–579.
79.
Coburn B, Sekirov I, Finlay BB. 2007. Type III secretion systems and disease. Clin Microbiol Rev 20:535–549.
80.
Xiang Y, Wen H, Yu Y, Li M, Fu X, Huang S. 2020. Gut-on-chip: recreating human intestine in vitro. J Tissue Eng 11:2041731420965318.
81.
Cherne MD, Sidar B, Sebrell TA, Sanchez HS, Heaton K, Kassama FJ, Roe MM, Gentry AB, Chang CB, Walk ST, Jutila M, Wilking JN, Bimczok D. 2021. A synthetic hydrogel, VitroGel ORGANOID-3, improves immune cell-epithelial interactions in a tissue chip co-culture model of human gastric organoids and dendritic cells. Front Pharmacol 12:707891.
82.
Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW. 2018. Molecular mechanisms of cell death. Cell Death Differ 25:486–541.
83.
Banfalvi G. 2017. Methods to detect apoptotic cell death. Apoptosis 22:306–323.
84.
Tait SWG, Ichim G, Green DR. 2014. Die another way–non-apoptotic mechanisms of cell death. J Cell Sci 127:2135–2144.
85.
Cummings J, Ward TH, Ranson M, Dive C. 2004. Apoptosis pathway-targeted drugs—from the bench to the clinic. Biochim Biophys Acta 1705:53–66.
86.
Phillips SMB, Bergstrom C, Walker B, Wang G, Alfaro T, Stromberg ZR, Hess BM. 2022. Engineered cell line imaging assay differentiates pathogenic from non-pathogenic bacteria. Pathogens 11:209.
87.
Leiser OP, Hobbs EC, Sims AC, Korch GW, Taylor KL. 2021. Beyond the list: bioagent-agnostic signatures could enable a more flexible and resilient biodefense posture than an approach based on priority agent lists alone. Pathogens 10:1497.

Information & Contributors

Information

Published In

cover image mSphere
mSphere
Volume 8Number 524 October 2023
eLocator: e00315-23
Editor: Alfredo G. Torres, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
PubMed: 37702517

History

Published online: 13 September 2023

Keywords

  1. bacterial pathogen
  2. high-throughput methods
  3. microbial functional traits
  4. pathogen agnostic detection
  5. pathogenesis

Contributors

Authors

Chemical and Biological Signatures Group, Pacific Northwest National Laboratory, Richland, Washington, USA
Author Contributions: Conceptualization, Data curation, Funding acquisition, Investigation, Methodology, Resources, Writing – original draft, and Writing – review and editing.
Shelby M. B. Phillips
Chemical and Biological Signatures Group, Pacific Northwest National Laboratory, Richland, Washington, USA
Author Contributions: Conceptualization, Formal analysis, Writing – original draft, and Writing – review and editing.
Kristin M. Omberg
Chemical and Biological Signatures Group, Pacific Northwest National Laboratory, Richland, Washington, USA
Author Contributions: Funding acquisition, Supervision, and Writing – review and editing.
Chemical and Biological Signatures Group, Pacific Northwest National Laboratory, Richland, Washington, USA

Editor

Alfredo G. Torres
Editor
The University of Texas Medical Branch at Galveston, Galveston, Texas, USA

Notes

The authors declare no conflict of interest.

Metrics & Citations

Metrics

Note:

  • For recently published articles, the TOTAL download count will appear as zero until a new month starts.
  • There is a 3- to 4-day delay in article usage, so article usage will not appear immediately after publication.
  • Citation counts come from the Crossref Cited by service.

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. For an editable text file, please select Medlars format which will download as a .txt file. Simply select your manager software from the list below and click Download.

View Options

Figures

Tables

Media

Share

Share

Share the article link

Share with email

Email a colleague

Share on social media

American Society for Microbiology ("ASM") is committed to maintaining your confidence and trust with respect to the information we collect from you on websites owned and operated by ASM ("ASM Web Sites") and other sources. This Privacy Policy sets forth the information we collect about you, how we use this information and the choices you have about how we use such information.
FIND OUT MORE about the privacy policy