Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jan 17.
Published in final edited form as: Nat Chem Biol. 2018 Mar 19;14(4):331–341. doi: 10.1038/s41589-018-0018-3

Whole-organism phenotypic screening for anti-infectives promoting host health

Anne E Clatworthy 1,2,3, Keith P Romano 2,4, Deborah T Hung 1,2,3,*
PMCID: PMC9843822  NIHMSID: NIHMS1859060  PMID: 29556098

Abstract

To date, antibiotics have been identified on the basis of their ability to kill bacteria or inhibit their growth rather than directly for their capacity to improve clinical outcomes of infected patients. Although historically successful, this approach has led to the development of an antibiotic armamentarium that suffers from a number of shortcomings, including the inevitable emergence of resistance and, in certain infections, suboptimal efficacy leading to long treatment durations, infection recurrence, or high mortality and morbidity rates despite apparent bacterial sterilization. Conventional antibiotics fail to address the complexities of in vivo bacterial physiology and virulence, as well as the role of the host underlying the complex, dynamic interactions that cause disease. New interventions are needed, aimed at host outcome rather than microbiological cure. Here we review the role of screening models for cellular and whole-organism infection, including worms, flies, zebrafish, and mice, to identify novel therapeutic strategies and discuss their future implications.


Since the discovery of Penicillin G by Alexander Fleming in 1928, the widespread use of antibiotics in clinical practice has arguably improved human health more than any other intervention in the history of medicine. The advent of antibiotics revolutionized modern medicine, rendering infections that were formerly life threatening as trivial and dramatically decreasing morbidity and mortality from post-operative and post-partum infections. Most conventional ‘Fleming-style’ antibiotics were identified by their ability to kill or inhibit bacteria growing in rich media in vitro. Despite their early success, a number of limitations have hindered their efficacy: (1) antibiotic resistance is rising rapidly, and efforts to identify novel classes of antibiotics have been largely unsuccessful; (2) treatment durations for some infections are long and are plagued by relapse after cessation of therapy; and (3) morbidity and mortality remain high in certain infections despite therapies that achieve bacterial sterilization. There is little doubt that alternative approaches for treating bacterial infections must be explored if we are to overcome these challenges. While many reviews discuss the challenges of conventional antibiotic discovery in the face of rising resistance1,2, this Perspective will focus on the later challenges of suboptimal efficacies and persisting morbidity and mortality in some infections. Rather than selecting for therapeutic agents that simply kill bacteria, we propose a shift in strategy to discover candidate compounds that attain the desired phenotype, namely improving host health. We explore the strengths and weaknesses of host model systems amenable to chemical screening for small molecules that improve host health, including cells in culture, worms, flies, zebrafish, and mice. Finally, we discuss the feasibility of nonconventional, host-targeted therapies for bacterial infections.

Though antibiotics are currently very effective for a wide range of bacterial infections, there remain a large number of infections in which antibiotic therapy is suboptimal. For example, antibiotics have limited utility in necrotizing fasciitis, a rapidly progressive soft-tissue infection caused by monomicrobial or polymicrobial infection within fascial tissue planes3. Indeed, without early recognition and aggressive surgical management, mortality rates from necrotizing fasciitis can exceed 30%4. Similarly, antibiotics alone typically fail to clear infection of implanted hardware or devices without removal of the foreign body because of failure of conventional antibiotics to effectively address bacteria in biofilms5,6. Furthermore, in other bacterial infections such as endocarditis and tuberculosis7,8, prolonged durations of treatment with cocktails of antibiotics are required to achieve a durable cure. Finally, there is still a high degree of mortality and morbidity from common bacterial infections, such as pneumonia, despite appropriate antimicrobial treatment, resulting in sterilization9,10. The need to consider the complex dynamic between host and pathogen is perhaps best illustrated in patients with sepsis, a life-threatening infection that is characterized by a dysregulated host response and organ dysfunction9. Severe forms of sepsis account for the most deaths in hospitalized patients9, with an estimated incidence of over one million cases per year in the United States alone with mortality rates of 15–20%11, rates that are not tied to infections being untreatable with resistant organisms. Thus, it is clear that alternative interventions are needed that take into account the complex cellular biology of bacteria in vivo, host response, and dynamic host–pathogen interactions, in addition to agents that only achieve microbiological cure. Though efforts have been made to find such adjunctive therapies for sepsis, all have failed to clinically improve host outcome12,13.

Nonconventional approaches

Though by far the most pervasive approach, ‘Fleming-style’ antibiotics, or small molecules that kill or inhibit the growth of bacteria, constitute only a subset of therapies that could, in principle, be deployed to treat bacterial infections (Fig. 1). Given the current crisis of antibiotic resistance and the pressing need to develop new therapeutic agents, recent attention has turned to nonconventional approaches14. These nonconventional approaches encompass strategies that are nonetheless ‘Fleming-like’, including phage lysins, antimicrobial peptides, antibodies, and wild-type or engineered bacteriophages that ultimately kill or neutralize bacteria in vitro or in vivo14 (Fig. 1). Other nonconventional options that target the bacterium but do not have classical in vitro activity include targeting of bacterial virulence mechanisms, biofilms, and/or bacterial proteins essential for survival only during infection (in vivo essential genes; Fig. 1), which has been reviewed elsewhere1416. Examples of these nonconventional approaches include the use of small molecules or antibodies that bind and neutralize bacterial toxins or other proteins14, a strategy that is currently being employed in the treatment of Clostridium difficile with the recent US Food and Drug Administration approval of bezlotoxumab, which binds to toxin B17.

Fig. 1 |. Conventional and nonconventional approaches to treating bacterial infection.

Fig. 1 |

Conventional, ‘Fleming-like’ approaches that also kill bacteria in vitro include (1) antibiotics, (2) phage lysins, (3) bacteriophages, and (4) antimicrobial peptides. Nonconventional approaches that could improve host health but do not kill or inhibit bacterial growth in vitro include (5) neutralizing antibodies, (6) inhibition of bacterial in vivo essential genes, (7) inhibition of virulence, (8) stimulating host immunity (for example, through immune response pathways like TLR signaling (I), autophagy (II), or induction of immune response genes (III)), and (9) dampening host immunity. Investigating host targets that improve host outcome during bacterial infection could occur at the cellular, organoid, or whole-organism level.

Alternatively, one could target the host itself using small molecules or biologics to either stimulate the immune response when it is inadequate or dampen it when it is excessive (Fig. 1). In fact, this latter approach is already adjunctive care for a number of bacterial infections because of its demonstrated clinical benefit. For example, the administration of corticosteroids as adjunctive therapy has been shown to have benefits for treating meningitis and Pneumocystis carinii pneumonia in HIV patients18,19; more recent data suggests that this treatment may more generally benefit other pneumonias as well20. In contrast, its value in septic shock has been considerably more controversial21, pointing to the challenge of finding such interventions and the likelihood that they may be beneficial in only certain subsets of patients and syndromes. In this Perspective, we focus on discussing host models that are amenable to high-throughput chemical screening for small molecules that improve host health as one approach to finding such interventions. While such screens have the potential to identify molecules that have ‘Fleming-like’ activity or inhibit bacterial virulence or bacterial in vivo essential genes, we will restrict our discussion to how these models might be used to discover chemical probes that can further our understanding of the host response to infection and how one might better intervene therapeutically by targeting the host.

Improving host health through pathogen and host parameters

The relationship between the pathogen and the host can be defined by a number of parameters including pathogen burden, bacterial pathogenicity, host immune response, host immune-cell function, host genotype, and host susceptibility to death, to name a few (Box 1). Interventions to favor host survival could potentially affect one or a few of these parameters. For example, interventions that augment the host’s ability to restrict bacterial expansion and control bacterial burden, such as conventional antibiotics or molecules that stimulate host immunity to clear bacteria (for example, P4 peptide14), can result in increased host survival or improved host health. Alternatively, interventions that increase the host’s ability to ‘tolerate’ the pathogen without changing the (quantitative) pathogen burden and instead modify the (qualitative) pathogenicity or toxicity of the pathogen, such as antivirulence approaches or host-targeting molecules that blunt the effects of toxins, or those that modulate responses downstream of host immunity, such as host metabolism, can also improve host health. One approach to analyzing the impact of these parameters on host health has been recently reported through disease-tolerance curves22, which describe the relationship between host health and pathogen load within a population (Box 1; Fig. 2a). By using Drosophila melanogaster as a model host for Listeria monocytogenes infection, the relationship between host health and bacterial load was determined to be sigmoidal and described by the parameters of vigor, slope, EC50, and severity of disease22 (Box 1; Fig. 2a). In this model, interventions that decrease bacterial burden should move an individual leftward along the curve (Fig. 2b). Alternatively, mutations that decrease bacterial pathogenicity shift the entire curve to the right, thereby increasing the EC50 such that for a certain pathogen burden22 there is improved host health (Fig. 2c). This work suggests that interventions such as antivirulence approaches or host-targeting molecules that blunt the effects of toxins should shift disease-tolerance curves to the right, increasing the ability of an individual host to ‘tolerate’ the pathogen. Thus, by applying disease-tolerance-curve analysis to novel interventions such as a new compound, one could potentially gain insight into the mechanisms by which the intervention provides benefit to the host.

Box 1 |. Parameters that affect host health.

A number of variables may affect host health over the course of an infection, including the initial inoculum size of the infecting organism, the burden of the pathogen in the host over time, the pathogenicity (i.e., the ability to cause host damage) of the infecting organism, the immune response of the host to infection—which may be determined by host genotype—and susceptibility of the host to death. Ecological immunologists have defined parameters that affect host health following infection in terms of resistance, vigor, resilience, and tolerance99.

Resistance is defined as an organism’s ability to control bacterial burden22,99. Thus, immune mechanisms that restrict bacterial burden can be considered to affect host resistance.

Vigor is defined as the health of an organism before infection22. One might imagine that elderly patient populations or patients undergoing chemotherapy for cancer may have different levels of vigor compared to young, healthy adult populations.

Resilience is defined as a host’s ability to return to its original state of health before infection22. It is understood to refer to an individual host organism, unlike disease-tolerance curves, which reflect disease tolerance within a population22.

A disease-tolerance curve is the dose–response curve of host health as a function of pathogen burden measured within a population22,99.

In the context of D. melanogaster infection with L. monocytogenes, disease-tolerance curves have been determined to be sigmoidal in shape and governed by the parameters of vigor, slope, EC50, and severity of disease22 (Fig. 2a). However, these curves may not always be sigmoidal in shape; a disease-tolerance curve to cancer modeled in D. melanogaster has been determined to be linear100.

Fig. 2 |. Disease-tolerance curve analysis of parameters that affect host health.

Fig. 2 |

a, Representation of a disease-tolerance curve of D. melanogaster infected with L. monocytogenes that has been determined to be sigmoidal and is defined by the parameters of vigor, slope, EC50, and severity of disease22. b, Interventions that decrease bacterial burden (for example, ‘Fleming-like’ antibiotics) increase host resistance and should move an individual leftward along the curve. c, Interventions that decrease bacterial pathogenicity should shift disease-tolerance curves to the right (red curve), increasing the EC50 and the ability of an individual to ‘tolerate’ the pathogen. d, Disease-tolerance curves can shift in more complicated ways if the intervention affects more than one parameter (for example, EC50, slope, and the severity of disease). Images in a and c are adapted from ref.22 under a Creative Commons CC BY 4.0 license.

In reality, a given intervention may affect multiple parameters and thus have the potential to shift the pathogen–host relationship, and therefore disease-tolerance curves, in more complex ways22. For example, one could imagine that inhibition of virulence could lead to both decreased host damage and increased pathogen clearance, thus affecting host resistance, the EC50 of the curve, and potentially the severity of disease itself (Fig. 2d). Understanding how small molecules that result in improved host health impact the various factors involved in determining the outcome of infection and the ensuing disease-tolerance curves will be important for building a more informed, systematic approach to alternative therapeutic discovery. Given our current limited understanding of all these factors, how they interact, and where one could intervene to favor host health, perhaps it is not surprising that rational, target-based approaches have fallen short, necessitating consideration of other approaches such as phenotypic screening.

Phenotypic versus target-based screening

Although target-based chemical screening has dominated drug-discovery efforts, it is increasingly recognized that many ‘first-in-class’ drug-discovery success stories have been disproportionately identified in phenotypic screens23,24. Furthermore, a target-based approach is predicated on the basis of the reasonable knowledge of what is a good target, an assumption that grossly overestimates our understanding of disease processes, as illustrated by the high failure rate of candidates in clinical trials, including candidates targeting sepsis. Thus, host targets and small molecules that improve host health might be more effectively identified in phenotypic rather than target-based screening approaches. As there has been more success in identifying new antibiotics in phenotypic screens for bacterial cell killing or growth inhibition than in target-based, biochemical screens, we suggest that phenotypic screening, with a pivot toward selecting for improved host health rather than for a microbiological cure, may afford promising chemical probes to elucidate the biology of the host–pathogen balance and small-molecule leads with the potential for therapeutic benefit.

Cellular host models

On the path to performing phenotypic screens for host health, in vitro host models begin to integrate the role of the host, albeit limited to the cellular level. To date, however, such screens carried out in cellular host models have typically selected for inhibition of bacterial growth in host cells or inhibition of host cell death. Recently, the advent of new technologies such as high-content screening, including imaging-based analysis, has allowed more complex descriptions of a compound’s activity, even from the primary screen, that can provide clues to mechanisms of action25. Indeed, such models are revealing examples of small molecules or peptides that can impact infection including the following: toll-like receptor (TLR) agonists2628, small molecule inhibitors of the protein kinases Akt and Abl that have also been shown to restrict proliferation of Salmonella typhimurium and Mycobacterium tuberculosis in cultured cells2931, or small molecules like rapamycin, carbamazepine, valproic acid, EGFR inhibitors, and fluoxetine (SSRI) that reduce M. tuberculosis bacterial burden in cultured cells, in part, by inducing autophagy29,32. A number of these studies have shown that results from cellular host-based assays do extend to whole-organism infection, as Abl and EGFR inhibitors prevent M. tuberculosis replication in mice29.

The disadvantage of cellular host models as surrogates for survival of the entire organism is that many viable targets and small molecules that can improve host outcome are likely to be missed in single host cell models. First, this reductionist model lacks the rich complexity of the immune response as a whole, which is a highly orchestrated response involving different immune cell types and various organ systems, as well as the complex proximal spatial organization of of pathogens into discrete lesions surrounded by a range of host cells that include both immune and supporting cell types, and more long-range effects whereby soluble signals such as cytokines can recruit additional host cells and activate more systemic responses, including interactions with other organ systems. In addition, the phenotypic assay used for high-throughput screening of cellular models often focuses simply on the ability to inhibit bacterial expansion, an oversimplified phenotype that does not necessarily map to improved host health (for example, the host has poor resilience and cannot readily return to its state of health before infection; Box 1), highlighting the disconnect between microbiological (sterilization) and patient cures. Finally, most cellular host models suffer from the limitations inherent to all two-dimensional cell-culture-based models in that immortalized host cell lines are often characterized by mutations and chromosomal abnormalities that can ultimately affect their function, and primary cells may be difficult to obtain (if human tissue is specifically needed), may survive poorly ex vivo following harvest for the period of time needed to adequately model infection, or may themselves rapidly loose functions specific to their cell type (for example, primary human hepatocytes); this is changing, however, with the advent of more sophisticated three-dimensional (3D) culture. Though the development of new in vitro models such as organoid 3D models33,34 and models derived from human pluripotent stem cells35,36 are beginning to make exciting strides to mitigate some of these limitations, with continued progress anticipated as the ability to incorporate numerous different cell types simultaneously into these systems arises, they will not completely recapitulate the whole organism.

Whole-organism screening

In an ideal world, one would screen for small molecules in exactly the biological setting in which one desires a therapeutic effect, namely in infected humans. However, appropriate ethical standards force us to consider alternatives that may optimize the successful translation from model to human disease. One obvious and attractive approach is screening in animal models that serve as a surrogate for human infection. Indeed, this approach has historically been effective, with perhaps the most successful whole-organism screens for anti-infectives resulting in the discoveries of the anti-syphilitic drug salvarsan37 (Fig. 3a), the sulfa drug prontosil38 (Fig. 3b), and the anti-parasitic drug avermectin39,40. Salvarsan was discovered by Paul Ehrlich through screening in mice and rabbits37. Described as the world’s first blockbuster drug, salvarsan was the most prescribed drug for syphilis from 1910 until the introduction of penicillin in 194241. Similarly, prontosil was discovered by Gerhard Domagk in 1932 in a screen for industrial dyes that rescued mice from streptococcal sepsis38. Strikingly, prontosil had no in vitro activity against streptococcal species because it requires metabolism in vivo to the active sulfanilamide. Domagk won the Nobel Prize for this work in 193938. Finally, avermectin, which was subsequently optimized to ivermectin (Fig. 3c), was discovered in mice by William Campbell at Merck by screening bacterial fermentation extracts for antiparasitic activity and is currently used in both human and veterinary medicine to treat parasitic worm infections including river blindness, strongyloidiasis, and lymphatic filariasis39,40. William Campbell shared the 2015 Nobel Prize for Physiology and Medicine for the discovery of avermectin along with Satoshi Ōmura39,40.

Fig. 3 |. Small molecules with host-targeting activity and/or identified through whole-organism screening.

Fig. 3 |

a, Salvarsan, reported to be a mixture of cyclic As–As bonded species98, identified by screening mice and rabbits infected with Treponema pallidum37. b, Prontosil, the first sulfa drug, identified in a screen of industrial dyes that rescued mice from streptococcal sepsis38. c, Ivermectin, derivatized from avermectin, which was identified by screening bacterial fermentation extracts in mice for antiparasitic properties39,40 and targets invertebrate glutamate-gated chloride channels. d, RPW-24, identified in a screen for small molecules that rescue C. elegans from E. fecalis infection48; thought to stimulate C. elegans host response49. e, 2-Amino acetophenone, enhances the survival of D. melanogaster and M. musculus infected with P. aeruginosa62,63. f, 16,16-dimethyl-prostaglandin E2 (a stabilized derivative of prostaglandin E2 currently being evaluated as a therapeutic for graft-versus-host disease), identified in a D. rerio screen for compounds that perturbed hematopoietic stem cell numbers23,76. g, PROTO-1, protects zebrafish hair cells from aminoglycoside-induced death23,77. h, Dorsomorphin78, inhibits bone morphogenetic protein (BMP) to treat fibrodysplasia ossificans progressiva (FOP) and anemia of inflammation23. i, anthracyclines (for example, epirubicin, shown), chemotherapeutic agents found to have a profound protective effect in a murine polymicrobial sepsis model86.

Although the examples of salvarsan, metabolized protonsil, and ivermectin still work in the ‘Fleming style’ of targeting pathogen viability, they nevertheless demonstrate the potential of whole-organism screening to find effective therapeutics. In fact, whole-organism screening may be needed in cases such as salvarsan, in which adequate in vitro culture systems do not exist for some pathogens, and protonsil, wherein activation of a prodrug by the host is required. Admittedly, in this era, their choice of animal models is generally considered unfeasible, which has necessitated the consideration of other host–pathogen models that are more amenable to screening for anti-infective candidates, including model organisms like the nematode worm Caenorhabditis elegans, the fruit fly D. melanogaster and the zebrafish Danio rerio. Powerful genetic tools and the ability to carry out chemical screens in these organisms are paving the way toward a more detailed mechanistic understanding of how different cell types and organ systems work together to impact host health. Notably, studies in these organisms do often translate to mice, which are the current gold standard for modeling mammalian infection. It is through these host models that we are beginning to ask the question, how do we improve host health and not just attain microbiological cure?

C. elegans

For over 15 years, the nematode worm C. elegans has been used to model the pathogenesis of a number of clinically relevant human pathogens, including Pseudomonas aeruginosa, S. typhimurium, Staphylococcus aureus, and Enterococcus fecalis42. In many cases, the bacterial virulence factors required for infection in mammals and worms are the same, thus validating the use of worms as a model host43. Importantly, studies have also revealed conservation of innate immune pathways between worms and mammals44 (Table 1). For example, defense against bacterial infection in worms relies on activation of the PMK-1 p38 mitogen-activated protein kinase (MAPK) pathway, which is related to the p38 MAPK cascade that is activated downstream of TLR signaling in insects and mammals44. The activation of the PMK-1 p38 MAPK cassette during infection plays a role in inducing both antimicrobial peptide synthesis and the unfolded protein response (UPR)45.

Table 1 |.

Comparison of model hosts amenable to chemical screening for anti-infectives

Organism C. elegans D. melanogaster D. rerio M. musculus
Size 1 mm 2 mm 0.9-4.5 mma 10 cm
Chemical screening throughput
(molecules screened/studyb)
High (37,20048) Medium (2,000–6,1005559, c) Medium (≤1,000d) Low 54/ avermectin
606/salvarsane
Mode of inoculation Feeding Needle pricking
Injector pumping
Feeding
Automated
(or manual)
microinjection
Immersion
Orogastric, injection, inhalation

Comparison of host immunity

Toll/TLR + + + +

MyD88 + + +

p38 MAPK pathway + + + +

NF-κB + + +

Immune cells + + +

Cytokines + + +

Immune organs + +
a

Zebrafish embryo and larvae lengths range from 0.9 mm 16 h post fertilization to 4.5 mm by 7 d post fertilization, which is the relative span during which many zebrafish infection models are conducted.

b

Throughput values represent the number of small molecules screened in a given study.

c

The published number of compounds screened in flies for other phenotypes with the caveat that this this level of throughput can be impacted by the mode of inoculation.

d

Throughput for D. rerio is based on the authors’ personal experience infecting zebrafish embryos manually by microinjection, which may be increased if the inoculum is given through automated microinjection81 or immersion70.

e

Screening was conducted in both mice and rabbits in order to identify salvarsan (compound 606)37.

From a technical standpoint, the C. elegans infection model is extremely attractive for screening for chemical modulators of infection. Large numbers (~300) of genetically identical progeny can be produced from a single hermaphroditic parent worm43. Worms are transparent, allowing infection with fluorescently tagged bacteria to be visualized in real time. Their size (~1 mm in length) permits arraying in a 96- or 384-well format for chemical screening46. For these assays, worms are infected by feeding on a lawn of the pathogen of interest and then arrayed into screening plates with a large particle sorter47,48. Worm viability following infection is then quantified with automated microscopy and image analysis47,48. Automation has made this the highest throughput whole organism amenable to chemical screening, with a throughput on the order of 10,000s48.

A number of small molecules have been identified that cure the worms of infection but do not themselves have bactericidal or static activity against the infecting bacterium in vitro46,48. One of these small molecules, RPW-24 (Fig. 3d), was identified in a screen for compounds that prolonged the survival of worms infected with the Gram-positive organism E. fecalis48 but also extended the survival of worms infected with the Gram-negative bacterium P. aeruginosa49. Because the virulence factors of these two pathogens are quite disparate, the authors of the study speculated that this small molecule might target the host. Using a combination of whole-organism expression profiling, classical epistasis analysis, and RNA interference (RNAi) in C. elegans, they demonstrated that RPW-24 stimulates the induction of host immune-response genes and promotes nematode survival through the p38 MAPK cascade and the transcription factor ATF-749. Though the precise target of RPW-24 is not known, the enormous genetic tractability of the organism and readily available loss-of-function mutants were used to narrow down possible relevant pathways and suggest candidate targets49. This study demonstrates the feasibility of identifying host-targeted small molecules that improve host outcome through whole-organism screening.

Although worms are clearly a powerful host model for studying bacterial infections, some aspects of host immunity are not conserved with vertebrates (Table 1). For example, worms do not have dedicated immune cells and therefore do not have adaptive immunity or express cytokines, similarly to mammals. Moreover, mammalian genes that are important for PAMP recognition and innate immune signaling such as MyD88 and NFκB do not have obvious orthologs in worms44. Finally, the single TLR homolog identified in worms does not play a central role in innate immunity to a number of pathogens, though it does appear to be important for worm avoidance of Serratia marcescens and defense against infection with Salmonella enterica44.

D. melanogaster

Flies (D. melanogaster) have long been used as a model host for bacterial pathogenesis studies due to their genetic tractability and the greater similarity of host immunity to mammals compared to worms, including the presence of phagocytic cells and more similar innate immune signaling cascades50 (Table 1). For example, the first Toll receptor was originally identified in D. melanogaster and was found to sense many pathogens, including bacteria and fungi50. Needless to say, since the original discovery of the first TLR, the voluminous body of literature in the past few decades illuminating the key role of TLRs in sensing pathogens to activate the innate immune response, including the identification of downstream signaling pathways and pathogen ligands that activate these pathways, attests to the power of flies as a host infection model and the way that understanding mechanisms of innate immune defense in flies translates to human infection.

Flies have been used to model infection with a wide array of Gram-negative, Gram-positive, and fungal pathogens, including P. aeruginosa, S. typhimurium, Mycobacterium marinum, S. aureus, Streptococcus sp., Candida albicans, and L. monocytogenes51. Like worms, large numbers of organisms can be evaluated, making flies a relatively high-throughput model compared to vertebrate infection models. Infection can be elicited through needle pricking or by more high-throughput methods of inoculation such as feeding or injector pumping, which enables more precise control of the inoculum51. Flies can be treated with small molecules through feeding or direct injection52, and assessment of bacterial burden is relatively facile in this host through homogenization and plating for colony forming units51.

Several studies in flies have played important roles in highlighting the potential disconnect between pathogen burden and host health, laying the foundation for the concept of disease tolerance22. A forward genetic screen for D. melanogaster mutants with altered sensitivity to infection from L. monocytogenes demonstrated that a number of mutant flies had a decreased mean time to death following infection but no difference in bacterial burden over time compared to wild-type flies53. Strikingly, there was no overlap between D. melanogaster genes that impacted time to death identified from a whole-organism screen and host genes identified as being important in Drosophila–L. monocytogenes cell culture infection models53, underscoring important differences between studying infection in a dish as opposed to a whole organism. Similarly, a study found that it is possible to have little correlation between bacterial burden and host survival in flies across 11 different genotypes infected with P. aeruginosa; flies can have low bacterial burden and poor survival or high bacterial burden and still remain relatively healthy54. These studies suggest that there are host factors that determine the relative health of the organism irrespective of bacterial burden, and thus interventions that focus solely on control or elimination of pathogen burden, characteristic of conventional antibiotics aiming for microbiological cure, may not be optimal for organism health.

Though chemical screening for anti-infective molecules has, to date, not been performed in flies, chemical screens have been performed for a variety of other phenotypes, including phenotypes associated with fragile X syndrome55,56, enhancers of radiation treatment for cancer57, and inhibitors of tumor cell growth58,59. Although it is not clear the degree to which compounds identified as having activity in flies will translate to mammals, the inverse often appears to be true: compounds with known mechanisms of action in mammals, like inhibition of microtubule and actin polymerization or inhibition of kinases or ion channels, do translate to flies60. Encouragingly, known antibiotics do reduce bacterial load and enhance survival in flies following infection22, suggesting that this model has potential. For example, recent work has described the ability of 2-amino acetophenone (2-AA; Fig. 3e), produced by P. aeruginosa, to enhance the survival of infected flies by inhibiting its acute virulence61; this survival benefit also translates to infected mice61. Interestingly, 2-AA appears to target the host by epigenetic reprogramming of the pro-inflammatory cytokine loci through HDAC1, ultimately resulting in downregulation of pro-inflammatory cytokine expression both in vitro and in vivo62,63. Strikingly, mice treated with 2-AA 4 days before P. aeruginosa infection had significantly improved survival compared to untreated controls, which could be reversed with an HDAC inhibitor, yet had a higher bacterial burden compared to untreated controls62,63.

Like worms, flies also have drawbacks as a host model for human infection. Although they have phagocytic cells, they lack the multilineage immune-cell complexity that is characteristic of vertebrates50. At the anatomical level, they lack vasculature and much of the organ system characteristic of mammals50, including the lymph organs that are clearly important features of mammalian immunity. Thus, despite the power of these invertebrate models, investigators have searched for additional host model organisms that better mirror mammalian—specifically human–—immunity while preserving many of the advantages of these invertebrate systems.

D. rerio

Zebrafish (D. rerio) embryos have served as a model host for a variety of pathogens including M. marinum64, S. aureus65, S. typhimurium66, and P. aeruginosa67,68, as they combine a number of advantages of both worms and flies with vertebrate immunity. Similar to worms, the dynamics of infection can be visualized microscopically in real time in living zebrafish embryos because of the availability of genetically engineered or chemically induced transparent zebrafish. Importantly, zebrafish are jawed vertebrates and have an immune system that is very similar to that of mammals (Table 1), including multilineage immune cell complexity (macrophages, neutrophils, B and T cells) and expression of pro-inflammatory cytokines and complement proteins69. Consistent and tunable inoculation of embryos and larvae is accomplished through microinjection, but is a lower throughput method than the feeding used to initiate C. elegans and some D. melanogaster infections. However, infection with M. marinum, which is a natural fish pathogen, can also be accomplished by embryo immersion in high concentrations of bacteria64,70, resulting in colonization of the developing gills and transient colonization of the gut70. Finally, bacterial expansion over time in zebrafish embryos is easily monitored either by plating for bacteria directly from homogenized fish or by using microscopy or luminometry to quantitate fluorescently or luminescently labeled bacteria, respectively, from infected embryos, which facilitates a more high-throughput way of determining bacterial expansion70,71.

In conjunction with zebrafish physiology being highly related to that of higher order vertebrates such as mammals, the technical ease of working with them makes zebrafish embryos well suited for chemical screening, as evidenced by the numerous screens in fish that have been performed for a variety of phenotypes, including but not limited to cardiotoxicity, haematopoiesis, leukocyte migration, embryogenesis, and suppressors of leukemia23. Compound administration is technically easy, as the fish are typically immersed in compound for the length of the assay, but most often requires absorption through the skin for efficacy23, which can provide some indication of tissue penetration. Though confirming the target of any compound found to have activity in a whole-organism screen is challenging, the genetic tools necessary to approach this problem are growing in fish, including CRISPR–Cas9, TALENs and morpholinos23,7275. Some hits from these screens have translated to mice and are moving forward toward human studies including, for example, 16,16-dimethyl prostaglandin E276 (16,16-dimethyl PGE2; Fig. 3f), a stabilized derivative of prostaglandin E2 used to prevent graft-versus-host disease (GvHD); PROTO-1 (Fig. 3g), which protects zebrafish hair cells from aminoglycoside-induced death23,77; and dorsomorphin78 (Fig. 3h), which inhibits bone morphogenetic protein (BMP) to treat fibrodysplasia ossificans progressiva (FOP) and anemia of inflammation23.

The feasibility of chemically screening zebrafish embryos for anti-infectives has also been demonstrated through studies showing the efficacy of conventional antibiotics. After manual microinjection of bacteria, exposure to known antibiotics in the surrounding water results in lower bacterial loads and increased survival of infected fish in multiple infection models68,79. Manual microinjection is highly laborious, which limits throughput; however, recent efforts to develop an automated microinjection pipeline to infect, array, and image infected embryos8082 has been limited to infection of pathogens into the yolk of embryos at the early developmental stage (16–128 cells)81. Although this is advantageous because of the increased throughput (~2,500 embryos per hour)8183, it remains to be seen whether infection of the yolk in early developmental stage embryos accurately replicates features of infection in mice or humans, because the bacteria are directly introduced into an extremely rich growth media (the yolk) at a developmental stage without immune cells. Some pathogens such as M. marinum and Staphylococcus epidermidis injected into the yolk in this fashion have been shown to replicate slowly and disseminate as the embryo develops8082.

Importantly, it is clear that targeting host mechanisms in combination with antibiotics to improve the outcome of infection in a zebrafish model can indeed translate to biology relevant to human infection. In a landmark study, the lta4h gene was identified in a zebrafish forward genetic screen as a gene required for defense against M. mariunum infection84. Lta4h catalyzes production of the pro-inflammatory eicosanoid LTB484. Interestingly, two SNPs identified in the human homolog of lta4H were associated with pulmonary and meningeal tuberculosis84. Though homozygosity of either allele was associated with increased disease severity in humans, heterozygosity was shown to be protective, resulting in a less severe disease84. Studies of Lta4H in fish revealed that its deficiency results in greater production of anti-inflammatory lipoxins (LXA4) and decreased levels of the pro-inflammatory cytokine TNFα, together contributing ultimately to poor control of M. marinum replication in vivo85. Conversely, overexpression of Lta4H results in excessive eicosanoid LTB4 production and excessive TNFα expression, contributing again to poor control of M. marinum replication in vivo, now due to an excessive inflammatory response85. Importantly, bacterial load could be controlled in fish by tuning the inflammatory response with small molecules that antagonized either LTB4 or TNFα in fish with increased levels of Lta4H or in Lta4H-deficient fish to improve host survival85. These observations in fish could be extended to human patient populations. The study showed a profound effect of glucocorticoid treatment on tubercular meningitis patient survival that was dependent on a patient’s genotype at a polymorphism in the LTA4H gene85. Although dexamethasone treatment had only a very modest effect on patient survival as a whole in the patients studied, independent of host genotype, patients homozygous for the LTA4H allele predicted to have increased LTA4H expression and excessive pro-inflammatory cytokine responses showed greatly increased survival following dexamethasone treatment85. This study is particularly important, as it demonstrates how studies in fish can be extrapolated to those in humans in which adjunctive glucocorticoid treatment is the standard of care for tubercular meningitis yet clearly has differential effects on host outcome based on lta4H genotype. Furthermore, it is illustrative of the fact that interventions targeting the host may be sensitive to host genotype, necessitating the identification of patient subpopulations who will benefit most from an intervention.

Mice

The story of the discoveries of salvarsan, prontosil, and ivermectin demonstrate the power of whole-organism phenotypic screens in mice. Though use of this model for screening is less feasible in the current era, except perhaps for small chemical collections such as repurposing libraries, several recent studies demonstrate the critical role of whole-organism models in the goal of targeting host health and not simply bacterial sterilization.

To date, many pathogenesis studies define whether a given bacterial gene is important for infection or whether a given small molecule has potential therapeutic efficacy by considering their effect solely on bacterial burden, often irrespective of the outcome on host survival. Yet, recent work in mice highlights the dramatic effects that interventions can have on host survival even without a substantial impact on bacterial burden. This point was best exemplified in a study exploring the effects of the anthracycline class of chemotherapeutic agents on host survival in a murine polymicrobial sepsis model (Fig. 3i)86. The authors found that low-dose administration of anthracyclines (epirubicin, doxorubicin, and daunorubicin) provided a profound protective effect on host survival yet had little to no effect on bacterial burden in multiple organs in a cecal ligation and puncture (CLP) model86. The protective effect of anthracyclines on host survival was found to be dependent on the DNA damage response mediated through the ATM serine/threonine kinase, (as Atm−/− mice were not protected with anthracycline treatment, and the autophagy pathway; ATM is known to negatively regulate mTOR, an inhibitor of autophagy)86. Perhaps the most striking finding of the paper was that the protective effect of anthracyclines on host survival was mediated through the activity of ATM and the autophagy pathway in the lung rather than in myeloid cell populations following CLP, as tissue-specific deletion of the Atm gene and the autophagy gene Atg7 in the lung, but not in myeloid cells, abrogated the protective effect of anthracyclines during sepsis86. This study not only underscores the disconnect between bacterial burden and host health, but also emphasizes how little we understand about which host tissue types might even be optimally targeted to promote survival during infection, an aspect that cannot be discovered or explored in reductionist models.

The elegant and complex interplay between various host cell types and organ systems that support organism health during infection is only beginning to be understood and clearly differs between different pathogens. A recent study on the host requirements for Bacillus anthracis toxin–induced lethality in mice also yielded unexpected findings on the tissue requirements for host death. B. anthracis causes disease primarily through the effects of two toxins: anthrax lethal toxin (LT), a metalloproteinase that cleaves MAPK kinases and the inflammasome component NLRP187, and edema toxin87 (ET), an adenylate cyclase. While the precise mechanism of toxin entry into host cells and the subsequent molecular effects of each toxin have been extensively studied, the mechanism by which these toxins ultimately lead to host death was not known until a number of tissue-specific knockout mice of the anthrax toxin receptor, which can bind both LT and ET and is required for their transport into host cells, were evaluated for their susceptibility to LT and ET88. The authors surprisingly found that mortality is dependent on LT targeting of cardiomyocytes and vascular smooth muscle cells, whereas ET-dependent mortality is dependent on targeting hepatocytes88. The examples of anthracycline and anthrax toxin both demonstrate that it is not necessarily obvious which tissue types and which organ systems would have the greatest impact on host survival during infection and that, indeed, this may differ between different infecting bacteria. Importantly, addressing these critical questions is now possible using whole-organism models of infection and the latest genetic tools such as tissue-specific siRNA or CRISPR–Cas9, which may ultimately guide the discovery of small molecules that specifically target a tissue that will have the greatest impact on host health improvement.

Though mice have historically been and continue to be the workhorse model for studying human disease, many questions have been raised regarding whether mice are indeed a good model for human infection. While mice are often currently used as the gold standard for evaluating whether a given intervention might translate to humans, it cannot be ignored that the translation rate of host-targeted therapeutic candidates for sepsis from mice to humans is extremely poor89. For conventional antibiotics, the high correlation between success based on in vitro efficacy and pharmacodynamics models and success in clinical translation in comparison to that of small-molecule candidates targeting other disease areas is likely tied to the fact that antibiotics are targeting the bacterium rather than the human. Thus, targeting the host in infection will likely be vulnerable to the lower translation rates due to the increased complexity of human disease and physiology compared to bacteria and inherent differences between mice and humans, such as the fact that mice are relatively refractory to inflammatory stimuli, such as endotoxin, in comparison to humans where the lethal dose in humans can be at least five orders of magnitude lower than that of mice90. The recent literature examining the effect of inflammatory stimuli on gene-expression responses in whole blood cell populations isolated from human patients and mice suggests that although there are statistically significant similarities in gene-expression changes between mice and humans91,92, the overlap in similarity of genes that were either up- or downregulated between the two species ranged from only 5.9%–15% in the samples analyzed92. Certainly, this somewhat low level of concordance could potentially be attributed to inherent differences in the proportions of immune-cell populations in whole blood between mice and humans, differences in the time frame of blood harvest between species, and large genetic differences expected in samples derived from human patients compared to the smaller genetic variations in inbred mouse lines93, making it quite difficult to make an even-handed comparison between the gene-expression responses of mice and humans. This point highlights one important caveat to finding candidates based on screening in alternative host models: the efficacy of the candidate and the relevance of its underlying biology must be validated in humans rather than extrapolated from surrogate models. Thus, while mice may seemingly be a better model for human infection than other models including worms, flies, and zebrafish, all are imperfect.

Implications and future directions

The value of whole-organism models for elucidating the biology of infection is abundantly clear given the impact that that these models have had on our understanding of mammalian and even human immunity. At the same time, the limitations of such models in completely reflecting human infection biology must also be recognized, as evidenced by the limited translation of therapeutic candidates from animal models to human studies9496. In light of this, it is worth contemplating what fair expectations are for whole-organism screens in worms, flies, fish, and mice. We propose that one valuable role that these models may play is in the identification of novel chemical probes to further elucidate the biology of infection and potentially identify new targets. Of course, this value might be dependent on the ability to elucidate the mechanisms of action of novel small-molecule probes of interest. The availability of rapidly improving, comprehensive genetic, genomic, and proteomic tools among these organisms is making this challenge increasingly tractable7275. As worms, flies, and fish become progressively high-throughput, cost-effective models, they offer the tremendous benefit of increasing the feasibility to discover small-molecule probes in models that better recapitulate the complexity of the host response in the organism as a whole, wherein extremely diverse sets of cell-types organized in complex anatomical structures must communicate information regarding the health of the organism over both short and long distances.

Optimistically, one can hope that these studies will extend beyond the discovery of chemical probes and eventually translate to therapeutic intervention of human infection. Is this a reasonable expectation? Aside from the challenge of discovering new antibiotics with new mechanisms, for conventional antibiotic candidates targeting the bacterium in which preclinical in vivo pharmacodynamics models are highly predictive of clinical efficacy97, this is a reasonable expectation. For host-targeting small-molecule candidates, the jury is still out. However, the fact that some, though not all, important immune mechanisms are evolutionarily conserved across the different host model organisms and the fact that many current drugs used in clinical medicine have been shown to also be efficacious in the different model organisms together suggest that indeed some candidates identified in these model organisms are expected to translate. Humanized versions of these model hosts might also improve translation potential. Furthermore, it is reasonable to speculate that the candidates most likely to translate to human infection may be ones that target host functions that are evolutionarily conserved across a large range of models, including worms, flies, zebrafish, and mice, as their conservation may be indicative of the importance of such functions.

Finally, the current lack of therapeutic approaches targeting the host in bacterial infection raises the more general question of the feasibility of this approach in the treatment of infection. However, current practice argues that this is indeed possible, with several examples of bacterial infections in which adjunctive therapy in combination with a conventional antibiotic results in superior survival compared to that with treatment with an antibiotic alone1820. Nevertheless, it is a valid concern that manipulating the host response to infection could be potentially detrimental, with augmentation of immunity resulting in uncontrolled inflammation and dampening of immunity causing immunocompromise and uncontrolled pathogen proliferation. Indeed, the study of Lta4H in zebrafish and its extrapolation to patients with tubercular meningitis attests to the fact that too much as well as too little inflammation both lead to a worse outcome for the host85. Thus, one of the great challenges of finding effective host-targeting therapeutics may be the need to tune host immunity rather than completely inhibiting or augmenting it. To increase the complexity of the problem, the degree of tuning required to maximize host health may vary as a function of time throughout the infection course and as a function of underlying host genotype as it relates to the magnitude of host response. The desire for candidates that are able to tune immunity in subtle ways mirrors the quest for inhibitors of kinases in cancer that modulate function to dampen excessive activity without completely eliminating the kinase activity important for homeostasis of normal cells. The ability to obtain this level of control may well determine whether a particular host-targeting therapy will be effective14,85. In this regard, the development of host-targeted therapies for infection may mirror innovative approaches now being taken in other disease areas, such as cancer, that are pushing toward personalized medicine that is tailored to the timing of intervention based on disease biomarkers and to the unique genotype of the patient.

Infection is still one of the leading causes of death worldwide. Given the growing problem of antibiotic resistance, the current poor rate of return in the development of new ‘Fleming-style’ antibiotics, and the ongoing problem that antibiotics alone are insufficient for improving host health in a variety of settings, there is a clear need to consider nonconventional approaches, such as targeting the host, to treat bacterial infections14. Multiple approaches will need to be explored to meet these challenges, and whole-organism screening is one valuable tool through which we can begin to explore how we might improve host health following infection instead of simply settling for microbiological cure.

Acknowledgements

We would like to acknowledge E. Office and S. Son for assistance with preparing the graphical abstract.

Footnotes

Competing interests

The authors declore no competing interests.

References

  • 1.Brown ED & Wright GD Antibacterial drug discovery in the resistance era. Nature 529, 336–343 (2016). [DOI] [PubMed] [Google Scholar]
  • 2.Fernandes P The global challenge of new classes of antibacterial agents: an industry perspective. Curr. Opin. Pharmacol 24, 7–11 (2015). [DOI] [PubMed] [Google Scholar]
  • 3.Young MH, Aronoff DM & Engleberg NC Necrotizing fasciitis: pathogenesis and treatment. Expert Rev. Anti Infect. Ther 3, 279–294 (2005). [DOI] [PubMed] [Google Scholar]
  • 4.Young MH, Engleberg NC, Mulla ZD & Aronoff DM Therapies for necrotising fasciitis. Expert Opin. Biol. Ther 6, 155–165 (2006). [DOI] [PubMed] [Google Scholar]
  • 5.Vinh DC & Embil JM Device-related infections: a review. J. Long Term Eff. Med. Implants 15, 467–488 (2005). [DOI] [PubMed] [Google Scholar]
  • 6.Song T, Duperthuy M & Wai SN Sub-optimal treatment of bacterial biofilms. Antibiotics (Basel) 5, 23 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Horsburgh CR Jr., Barry CE III & Lange C Treatment of tuberculosis. N. Engl. J. Med 373, 2149–2160 (2015). [DOI] [PubMed] [Google Scholar]
  • 8.Baddour LM et al. Infective endocarditis in adults: diagnosis, antimicrobial therapy, and management of complications: a scientific statement for healthcare professionals from the american heart association. Circulation 132, 1435–1486 (2015). [DOI] [PubMed] [Google Scholar]
  • 9.Deutschman CS & Tracey KJ Sepsis: current dogma and new perspectives. Immunity 40, 463–475 (2014). [DOI] [PubMed] [Google Scholar]
  • 10.Prina E, Ranzani OT & Torres A Community-acquired pneumonia. Lancet 386, 1097–1108 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kaukonen KM, Bailey M, Suzuki S, Pilcher D & Bellomo R Mortality related to severe sepsis and septic shock among critically ill patients in Australia and New Zealand, 2000–2012. J. Am. Med. Assoc 311, 1308–1316 (2014). [DOI] [PubMed] [Google Scholar]
  • 12.Giamarellos-Bourboulis EJ The failure of biologics in sepsis: where do we stand? Int. J. Antimicrob. Agents 42, S45–S47 (2013). [DOI] [PubMed] [Google Scholar]
  • 13.Marshall JC Such stuff as dreams are made on: mediator-directed therapy in sepsis. Nat. Rev. Drug Discov 2, 391–405 (2003). [DOI] [PubMed] [Google Scholar]
  • 14.Czaplewski L et al. Alternatives to antibiotics-a pipeline portfolio review. Lancet Infect. Dis 16, 239–251 (2016). [DOI] [PubMed] [Google Scholar]
  • 15.Allen RC, Popat R, Diggle SP & Brown SP Targeting virulence: can we make evolution-proof drugs? Nat. Rev. Microbiol 12, 300–308 (2014). [DOI] [PubMed] [Google Scholar]
  • 16.Rasko DA & Sperandio V Anti-virulence strategies to combat bacteria-mediated disease. Nat. Rev. Drug Discov 9, 117–128 (2010). [DOI] [PubMed] [Google Scholar]
  • 17.Orth P et al. Mechanism of action and epitopes of Clostridium difficile toxin B-neutralizing antibody bezlotoxumab revealed by X-ray crystallography. J. Biol. Chem 289, 18008–18021 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ewald H et al. Adjunctive corticosteroids for Pneumocystis jiroveci pneumonia in patients with HIV infection. Cochrane Database Syst. Rev 2015, CD006150 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Prasad K, Singh MB & Ryan H Corticosteroids for managing tuberculous meningitis. Cochrane Database Syst. Rev 4, CD002244 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Feldman C & Anderson R Corticosteroids in the adjunctive therapy of community-acquired pneumonia: an appraisal of recent meta-analyses of clinical trials. J. Thorac. Dis 8, E162–E171 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Yadav H & Cartin-Ceba R Balance between hyperinflammation and immunosuppression in sepsis. Semin. Respir. Crit. Care Med 37, 42–50 (2016). [DOI] [PubMed] [Google Scholar]
  • 22.Louie A, Song KH, Hotson A, Thomas Tate A & Schneider DS How many parameters does it take to describe disease tolerance? PLoS Biol. 14, e1002435 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study describes disease-tolerance curves in L. monocytogenes infected D. melanogaster as sigmoidal and governed by the parameters of vigor, slope, EC50, and the severity of disease.
  • 23.MacRae CA & Peterson RT Zebrafish as tools for drug discovery. Nat. Rev. Drug Discov 14, 721–731 (2015). [DOI] [PubMed] [Google Scholar]
  • 24.Swinney DC & Anthony J How were new medicines discovered? Nat. Rev. Drug Discov 10, 507–519 (2011). [DOI] [PubMed] [Google Scholar]
  • 25.Mattiazzi Usaj M et al. High-content screening for quantitative cell biology. Trends Cell Biol. 26, 598–611 (2016). [DOI] [PubMed] [Google Scholar]
  • 26.Raby AC et al. Targeting the TLR co-receptor CD14 with TLR2-derived peptides modulates immune responses to pathogens. Sci. Transl. Med 5, 185ra64 (2013). [DOI] [PubMed] [Google Scholar]
  • 27.Hancock RE, Nijnik A & Philpott DJ Modulating immunity as a therapy for bacterial infections. Nat. Rev. Microbiol 10, 243–254 (2012). [DOI] [PubMed] [Google Scholar]
  • 28.Dowling JK & Mansell A Toll-like receptors: the swiss army knife of immunity and vaccine development. Clin. Transl. Immunology 5, e85 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Stanley SA et al. Identification of host-targeted small molecules that restrict intracellular Mycobacterium tuberculosis growth. PLoS Pathog. 10, e1003946 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Napier RJ et al. Imatinib-sensitive tyrosine kinases regulate mycobacterial pathogenesis and represent therapeutic targets against tuberculosis. Cell Host Microbe 10, 475–485 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kuijl C et al. Intracellular bacterial growth is controlled by a kinase network around PKB/AKT1. Nature 450, 725–730 (2007). [DOI] [PubMed] [Google Scholar]
  • 32.Schiebler M et al. Functional drug screening reveals anticonvulsants as enhancers of mTOR-independent autophagic killing of Mycobacterium tuberculosis through inositol depletion. EMBO Mol. Med 7, 127–139 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Fatehullah A, Tan SH & Barker N Organoids as an in vitro model of human development and disease. Nat. Cell Biol 18, 246–254 (2016). [DOI] [PubMed] [Google Scholar]
  • 34.Ranga A, Gjorevski N & Lutolf MP Drug discovery through stem cell-based organoid models. Adv. Drug Deliv. Rev 69-70, 19–28 (2014). [DOI] [PubMed] [Google Scholar]
  • 35.Bartfeld S et al. In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection. Gastroenterology 148, 126–136.e6 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ng S et al. Human iPSC-derived hepatocyte-like cells support Plasmodium liver-stage infection in vitro. Stem Cell Rep. 4, 348–359 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kaufmann SH Paul Ehrlich: founder of chemotherapy. Nat. Rev. Drug Discov 7, 373 (2008). [DOI] [PubMed] [Google Scholar]
  • 38.Raju TN The Nobel chronicles. 1939: Gerhard Domagk (1895–1964). Lancet 353, 681 (1999). [DOI] [PubMed] [Google Scholar]
  • 39.Campbell WC Ivermectin: a reflection on simplicity (Nobel Lecture). Angew. Chem. Int. Edn. Engl 55, 10184–10189 (2016). [DOI] [PubMed] [Google Scholar]
  • 40.Omura S & Crump A The life and times of ivermectin - a success story. Nat. Rev. Microbiol 2, 984–989 (2004). [DOI] [PubMed] [Google Scholar]
  • 41.Yarnell A in Chemical and Engineering News, Vol. 83 (ed. Baum RM) p. 63 (American Chemical Society, Washington DC, 2005). [Google Scholar]
  • 42.Powell JR & Ausubel FM Models of Caenorhabditis elegans infection by bacterial and fungal pathogens. Methods Mol. Biol 415, 403–427 (2008). [DOI] [PubMed] [Google Scholar]
  • 43.Ewbank JJ & Zugasti OC elegans: model host and tool for antimicrobial drug discovery. Dis. Model. Mech 4, 300–304 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Irazoqui JE, Urbach JM & Ausubel FM Evolution of host innate defence: insights from Caenorhabditis elegans and primitive invertebrates. Nat. Rev. Immunol 10, 47–58 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Richardson CE, Kooistra T & Kim DH An essential role for XBP-1 in host protection against immune activation in C. elegans. Nature 463, 1092–1095 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Moy TI et al. Identification of novel antimicrobials using a live-animal infection model. Proc. Natl Acad. Sci. USA 103, 10414–10419 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Conery AL, Larkins-Ford J, Ausubel FM & Kirienko NV High-throughput screening for novel anti-infectives using a C. elegans pathogenesis model. Curr. Protoc. Chem. Biol 6, 25–37 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Moy TI et al. High-throughput screen for novel antimicrobials using a whole animal infection model. ACS Chem. Biol 4, 527–533 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Pukkila-Worley R et al. Stimulation of host immune defenses by a small molecule protects C. elegans from bacterial infection. PLoS Genet. 8, e1002733 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study describes the use of whole-organism transcriptional profiling, classical epistasis analysis, and RNAi screens in C. elegans to define pathways perturbed by the small molecule RPW-24, identified through whole-organism screening in C. elegans.
  • 50.Buchon N, Silverman N & Cherry S Immunity in Drosophila melanogaster—from microbial recognition to whole-organism physiology. Nat. Rev. Immunol 14, 796–810 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Apidianakis Y & Rahme LG Drosophila melanogaster as a model host for studying Pseudomonas aeruginosa infection. Nat. Protoc 4, 1285–1294 (2009). [DOI] [PubMed] [Google Scholar]
  • 52.Tzelepis I, Kapsetaki SE, Panayidou S & Apidianakis Y Drosophila melanogaster: a first step and a stepping-stone to anti-infectives. Curr. Opin. Pharmacol 13, 763–768 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Ayres JS, Freitag N & Schneider DS Identification of Drosophila mutants altering defense of and endurance to Listeria monocytogenes infection. Genetics 178, 1807–1815 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study identified D. melanogaster mutants that were more susceptible to L. monocytogenes infection, though their ability to control bacterial burden was unchanged compared to wild type.
  • 54.Corby-Harris V, Habel KE, Ali FG & Promislow DE Alternative measures of response to Pseudomonas aeruginosa infection in Drosophila melanogaster. J. Evol. Biol 20, 526–533 (2007). [DOI] [PubMed] [Google Scholar]
  • 55.Chang S et al. Identification of small molecules rescuing fragile X syndrome phenotypes in Drosophila. Nat. Chem. Biol 4, 256–263 (2008). [DOI] [PubMed] [Google Scholar]
  • 56.Qurashi A et al. Chemical screen reveals small molecules suppressing fragile X premutation rCGG repeat-mediated neurodegeneration in Drosophila. Hum. Mol. Genet 21, 2068–2075 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Gladstone M et al. A translation inhibitor identified in a Drosophila screen enhances the effect of ionizing radiation and taxol in mammalian models of cancer. Dis. Model. Mech 5, 342–350 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Willoughby LF et al. An in vivo large-scale chemical screening platform using Drosophila for anti-cancer drug discovery. Dis. Model. Mech 6, 521–529 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Markstein M et al. Systematic screen of chemotherapeutics in Drosophila stem cell tumors. Proc. Natl Acad. Sci. USA 111, 4530–4535 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Fernández-Hernández I, Scheenaard E, Pollarolo G & Gonzalez C The translational relevance of Drosophila in drug discovery. EMBO Rep. 17, 471–472 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kesarwani M et al. A quorum sensing regulated small volatile molecule reduces acute virulence and promotes chronic infection phenotypes. PLoS Pathog. 7, e1002192 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bandyopadhaya A et al. The quorum sensing volatile molecule 2-amino acetophenon modulates host immune responses in a manner that promotes life with unwanted guests. PLoS Pathog. 8, e1003024 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrated that pretreatment with 2-AA prior to lethal infection with P. aeruginosa resulted in downregulation of pro-inflammatory cytokines, upregulation of anti-inflammatory cytokines, higher bacterial burden, and a significant survival benefit in treated animals, suggesting that 2-AA is an immunomodulatory molecule that ‘tolerizes’ the host to the presence of the pathogen.
  • 63.Bandyopadhaya A, Tsurumi A, Maura D, Jeffrey KL & Rahme LG A quorum-sensing signal promotes host tolerance training through HDAC1-mediated epigenetic reprogramming. Nat. Microbiol 1, 16174 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrated that the effect of 2-AA on downregulation of pro-inflammatory cytokine induction is mediated through HDAC-1, and the survival benefit provided by 2-AA in infected animals could be inhibited with HDAC inhibitors.
  • 64.Davis JM et al. Real-time visualization of mycobacterium-macrophage interactions leading to initiation of granuloma formation in zebrafish embryos. Immunity 17, 693–702 (2002). [DOI] [PubMed] [Google Scholar]
  • 65.Prajsnar TK, Cunliffe VT, Foster SJ & Renshaw SA A novel vertebrate model of Staphylococcus aureus infection reveals phagocyte-dependent resistance of zebrafish to non-host specialized pathogens. Cell. Microbiol 10, 2312–2325 (2008). [DOI] [PubMed] [Google Scholar]
  • 66.van der Sar AM et al. Zebrafish embryos as a model host for the real time analysis of Salmonella typhimurium infections. Cell. Microbiol 5, 601–611 (2003). [DOI] [PubMed] [Google Scholar]
  • 67.Brannon MK et al. Pseudomonas aeruginosa Type III secretion system interacts with phagocytes to modulate systemic infection of zebrafish embryos. Cell. Microbiol 11, 755–768 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Clatworthy AE et al. Pseudomonas aeruginosa infection of zebrafish involves both host and pathogen determinants. Infect. Immun 77, 1293–1303 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Renshaw SA & Trede NS A model 450 million years in the making: zebrafish and vertebrate immunity. Dis. Model. Mech 5, 38–47 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Dalton JP et al. Screening of anti-mycobacterial compounds in a naturally infected zebrafish larvae model. J. Antimicrob. Chemother 72, 421–427 (2017). [DOI] [PubMed] [Google Scholar]
  • 71.Takaki K, Cosma CL, Troll MA & Ramakrishnan L An in vivo platform for rapid high-throughput antitubercular drug discovery. Cell Rep. 2, 175–184 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Deans RM et al. Parallel shRNA and CRISPR-Cas9 screens enable antiviral drug target identification. Nat. Chem. Biol 12, 361–366 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Neggers JE et al. Identifying drug-target selectivity of small-molecule CRM1/XPO1 inhibitors by CRISPR/Cas9 genome editing. Chem. Biol 22, 107–116 (2015). [DOI] [PubMed] [Google Scholar]
  • 74.Nijman SM Functional genomics to uncover drug mechanism of action. Nat. Chem. Biol 11, 942–948 (2015). [DOI] [PubMed] [Google Scholar]
  • 75.Ziegler S, Pries V, Hedberg C & Waldmann H Target identification for small bioactive molecules: finding the needle in the haystack. Angew. Chem. Int. Edn. Engl 52, 2744–2792 (2013). [DOI] [PubMed] [Google Scholar]
  • 76.North TE et al. Prostaglandin E2 regulates vertebrate haematopoietic stem cell homeostasis. Nature 447, 1007–1011 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Owens KN et al. Identification of genetic and chemical modulators of zebrafish mechanosensory hair cell death. PLoS Genet. 4, e1000020 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Yu PB et al. Dorsomorphin inhibits BMP signals required for embryogenesis and iron metabolism. Nat. Chem. Biol 4, 33–41 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Adams KN et al. Drug tolerance in replicating mycobacteria mediated by a macrophage-induced efflux mechanism. Cell 145, 39–53 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Carvalho R et al. A high-throughput screen for tuberculosis progression. PLoS One 6, e16779 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Veneman WJ et al. Establishment and optimization of a high throughput setup to study Staphylococcus epidermidis and Mycobacterium marinum infection as a model for drug discovery. J. Vis. Exp 2014, e51649 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Veneman WJ et al. A zebrafish high throughput screening system used for Staphylococcus epidermidis infection marker discovery. BMC Genomics 14, 255 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Spaink HP et al. Robotic injection of zebrafish embryos for high-throughput screening in disease models. Methods 62, 246–254 (2013). [DOI] [PubMed] [Google Scholar]
  • 84.Tobin DM et al. The lta4h locus modulates susceptibility to mycobacterial infection in zebrafish and humans. Cell 140, 717–730 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study identified the requirement of the lta4h gene for defense against M. marinum infection in zebrafish and SNPs in the human homolog of lta4H that were associated with and provided heterozygous advantage for pulmonary and meningeal tuberculosis.
  • 85.Tobin DM et al. Host genotype-specific therapies can optimize the inflammatory response to mycobacterial infections. Cell 148, 434–446 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrated that host-directed therapies can be used to fine tune the inflammatory response—which may be either excessive or deficient dependening on host genotype—and improve host outcome during mycobacterial infection in both zebrafish and human tuberculous meningitis patients.
  • 86.Figueiredo N et al. Anthracyclines induce DNA damage response-mediated protection against severe sepsis. Immunity 39, 874–884 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrated that anthracyclines provide a profound protective effect on host survival yet have no effect on bacterial burden in a severe sepsis model and that the survival effect is mediated through ATM and the autophagy pathway in the lung.
  • 87.Moayeri M, Leppla SH, Vrentas C, Pomerantsev AP & Liu S Anthrax Pathogenesis. Annu. Rev. Microbiol 69, 185–208 (2015). [DOI] [PubMed] [Google Scholar]
  • 88.Liu S et al. Key tissue targets responsible for anthrax-toxin-induced lethality. Nature 501, 63–68 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrated that anthrax-toxin-induced mortality in mice is dependent on LT targeting of cardiomyocytes and vascular smooth muscle cells and ET targeting of hepatocytes.
  • 89.Fink MP Animal models of sepsis. Virulence 5, 143–153 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Munford RS Murine responses to endotoxin: another dirty little secret? J. Infect. Dis 201, 175–177 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Seok J et al. Genomic responses in mouse models poorly mimic human inflammatory diseases. Proc. Natl Acad. Sci. USA 110, 3507–3512 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Takao K & Miyakawa T Genomic responses in mouse models greatly mimic human inflammatory diseases. Proc. Natl Acad. Sci. USA 112, 1167–1172 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Shay T, Lederer JA & Benoist C Genomic responses to inflammation in mouse models mimic humans: we concur, apples to oranges comparisons won’t do. Proc. Natl Acad. Sci. USA 112, E346 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Hackam DG & Redelmeier DA Translation of research evidence from animals to humans. J. Am. Med. Assoc 296, 1731–1732 (2006). [DOI] [PubMed] [Google Scholar]
  • 95.van der Worp HB et al. Can animal models of disease reliably inform human studies? PLoS Med. 7, e1000245 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Mak IW, Evaniew N & Ghert M Lost in translation: animal models and clinical trials in cancer treatment. Am. J. Transl. Res 6, 114–118 (2014). [PMC free article] [PubMed] [Google Scholar]
  • 97.Rex JH et al. A comprehensive regulatory framework to address the unmet need for new antibacterial treatments. Lancet Infect. Dis 13, 269–275 (2013). [DOI] [PubMed] [Google Scholar]
  • 98.Lloyd NC, Morgan HW, Nicholson BK & Ronimus RS The composition of Ehrlich’s salvarsan: resolution of a century-old debate. Angew. Chem. Int. Ed. Engl 44, 941–944 (2005). [DOI] [PubMed] [Google Scholar]
  • 99.Ayres JS & Schneider DS Tolerance of infections. Annu. Rev. Immunol 30, 271–294 (2012). [DOI] [PubMed] [Google Scholar]
  • 100.Dillman AR & Schneider DS Defining resistance and tolerance to Cancer. Cell Rep. 13, 884–887 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES