Abstract
In recent years, tremendous advances have been made in our ability to characterize complex microbial communities such as the gut microbiota, and numerous surveys of the human gut microbiota have identified countless associations between different compositional attributes of the gut microbiota and adverse health conditions. However, most of these findings in humans are purely correlative and animal models are required for prospective evaluation of such changes as causative factors in disease initiation or progression. As in most fields of biomedical research, microbiota-focused studies are predominantly performed in mouse or rat models. Depending on the field of research and experimental question or objective, non-rodent models may be preferable due to better translatability or an inability to use rodents for various reasons. The following review describes the utility and limitations of several non-rodent model species for research on the microbiota and its influence on host physiology and disease. In an effort to balance the breadth of potential model species with the amount of detail provided, four model species are discussed: zebrafish, dogs, pigs, and rabbits.
Keywords: animal model, comparative medicine, microorganisms, organism models
Résumé
Ces dernières années, des progrèss énormes ont été réalisés dans notre capacité à caractériser les communautés microbiennes complexes telles que le microbiote intestinal (MI), et de nombreuses enquêtes menées sur le MI ont identifié un nombre incalculable d’associations entre différents attributs de composition du MI et les effets indésirables sur la santé. Cependant, la majorité de ces résultats chez les humains sont purement corrélatifs et les modèles animaux sont nécessaires à l’évaluation prospective de ces changements en tant que facteurs étiologiques de l’initiation ou de la progression de la maladie. Comme dans la plupart des domaines de la recherche biomédicale, les études axées sur le microbiote sont principalement effectuées sur des modèles de souris ou de rat. Selon le domaine de la recherche et la question ou l’objectif de l’expérience, des modèles autres que des rongeurs peuvent être préférables en raison de meilleures possibilités de translation ou du fait de l’impossibilité d’utiliser des rongeurs pour diverses raisons. L’étude ci-après décrit l’utilité et les limites de plusieurs modèles issus d’espèces autres que les rongeurs pour la recherche sur la santé humaine, et leur influence sur la physiologie de l’hôte et la maladie. Afin d’équilibrer l’étendue des espèces de modèles potentiels avec la quantité de détails fournie, quatre espèces de modèles sont discutées : le poisson-zèbre, le chien, le porc et le lapin.
Abstract
In den letzten Jahren haben sich unsere Fähigkeiten zur Charakterisierung komplexer mikrobieller Gemeinschaften wie der Darmflora (intestinale Mikrobiota, GM) wesentlich verbessert, und bei zahlreichen Untersuchungen der menschlichen GM wurden unzählige Zusammenhänge zwischen verschiedenen Merkmalen der Zusammensetzung der GM und ungünstigen gesundheitlichen Umständen erkannt. Die meisten dieser Ergebnisse beim Menschen sind jedoch rein korrelativ, und Tiermodelle sind für die prospektive Bewertung von solchen Veränderungen als ursächliche Faktoren bei der Entstehung oder dem Fortschreiten von Krankheiten erforderlich. Wie in den meisten Bereichen der biomedizinischen Forschung werden Mikrobiota-Studien überwiegend mit Maus- oder Rattenmodellen durchgeführt. Je nach Forschungsgebiet und experimenteller Fragestellung oder Zielsetzung sind Nicht-Nagetier-Modelle aufgrund besserer Übertragbarkeit oder eines aus unterschiedlichen Gründen gespeisten Unvermögens der Nutzung von Nagetieren möglicherweise vorzuziehen. Der folgende Bericht beschreibt den Nutzen und die Grenzen mehrerer Nicht-Nagetier-Modellarten für die Mikrobiota-Forschung und ihren Einfluss auf Physiologie und Krankheit des Wirts. Um die Breite der potenziellen Modellarten ausgewogen mit der gelieferten Detailmenge in Einklang zu bringen, werden vier Modellarten diskutiert: Zebrafische, Hunde, Schweine und Kaninchen.
Resumen
En los últimos años, se han logrado grandes avances en nuestra capacidad para caracterizar comunidades microbiales complejas como la microbiota intestinal (GM), y numerosas encuestas sobre la GM humana han identificado muchas asociaciones entre distintos atributos composicionales de la GM y unas condiciones de salud adversas. Sin embargo, la mayoría de estas indagaciones en los humanos es puramente correlativa y se requieren modelos animales para una evaluación prospectiva de dichos cambios como factores causales en el inicio o avance de la enfermedad. Como ocurre en muchos campos de la investigación biomédica, los estudios centrados en la microbiota se realizan predominantemente con ratones o ratas. Dependiendo del campo de investigación y la cuestión experimental u objetivo, los modelos de no roedores pueden ser preferibles debido a una mejor traducibilidad o una incapacidad de usar roedores por distintos motivos. La siguiente reseña describe la utilidad y las limitaciones de distintas especies de modelos de no roedores para investigar la microbiota y su influencia en la enfermedad y fisiología del huésped. En un esfuerzo por equilibrar la gama de posibles especies modelo con la cantidad de datos suministrados, se ponen sobre el tapete cuatro especies: pez cebra, perros, cerdos y conejos.
Introduction
The development and increasing availability of methods to characterize complex microbial communities has led to a staggering number of associations between characteristics of the human gut microbiota (GM) and health or disease. However, the bulk of those associations are purely correlative and causative relationships in humans are difficult to identify due the ethical implications of prospective experiments in human subjects. Moreover, the genetic and environmental heterogeneity of human populations makes it incredibly difficult to conclusively associate characteristics of the GM and disease phenotypes. Animal models can often be used to circumvent those limitations and allow for prospective experimentation in controlled conditions. Additionally, the disease or condition replicated by many animal models often occurs in a compressed timespan relative to the human condition being modeled. As in most fields of biomedical research, investigations related to the GM and host interactions are dominated by rodent models due to their high fecundity, convenience, availability, tractable genetics, and other reasons. That said, there are limitations to rodent models and certain procedures simply preclude their use. The current review will focus on GM-centric research performed in certain non-rodent model species, and specific benefits or attributes of each meriting consideration in experimental design. Specifically, we address invertebrate and zebrafish models as cost-effective precursors, alternatives, or adjuncts to rodent models, which should be considered in the context of the three Rs (reduction, refinement, and replacement of animal models). The discussion then switches its focus to three higher vertebrate models (rabbits, dogs, and pigs), which may be more appropriate model species for certain lines of investigation and, in the case of dogs and pigs, may serve as more relevant models for humans due to their shared environmental exposure. With that in mind, readers are also directed to more focused reviews on the aforementioned model species1–4 as well as other species not discussed here, such as non-human primates5,6 and other non-murine rodent species.7
Invertebrates
Invertebrate hosts such as worms and insects represent multicellular in vivo systems with variably compartmentalized digestive tracts and homologs of many of the differentiated cell types found in vertebrate hosts. There are several invertebrate model species that are frequently used to study certain interactions between the host and its microbiota. Frequently, invertebrate species are used in studies focused on symbioses between the host and microbiota and the mechanisms by which those interdependent relationships are established and maintained. Two characteristics of invertebrates making them particularly amenable to such research are their reliance on the innate immune system8 and a highly restricted (i.e., low α-diversity) gut microbiota.9 Much work has been done in this regard using entomopathogenic nematode hosts such as Heterorhabditis bacteriophora and Steinernema carpocapsae and their respective symbionts. Photorhabdus luminescens and Xenorhabdus nematophila,10–13 as well as marine organisms such as the freshwater leech (Hirudo verbana)14,15 and bobtail squid (Euprymna scolopes) and their respective symbionts Aeromonas veronii and Vibrio fischeri.16,17
For studies necessitating genetically tractable model organisms, Drosophila melanogaster and Caenorhabditis elegans are the two most commonly used invertebrate models. Although these host species can also be used to study the nature of microbial symbioses, their relatively short lifespan and the aforementioned ability to manipulate their well-characterized genomes18–23 make them attractive models to study the influence of the microbiota on host aging, including methods involving caloric restriction.24–28 Additionally, it is possible to render both Drosophila sp. and C. elegans axenic or germfree (GF) for use in gnotobiotic experiments.29,30 Moreover, their small size allows for well-powered studies and high-throughput whole animal testing including in vivo fluorescent imaging in the case of C. elegans. As one final advantage, the use of invertebrate models abrogates much of the regulatory concerns associated with vertebrate animal research. The primary limitations associated with invertebrate models include the stark differences in gastrointestinal anatomy relative to mammalian hosts, differences in microbial community structure at the most basic taxonomic levels, and an inability to model many of the immune-mediated and neoplastic conditions occurring in humans such as those reliant on the adaptive immune system. Table 1 summarizes these benefits and limitations, alongside those of the host species discussed below.
Table 1.
Model organism(s) | Advantages/uses | Limitations |
---|---|---|
Invertebrates |
|
|
Zebrafish |
|
|
Dogs |
|
|
Pigs |
|
|
Rabbits |
|
|
IBD: inflammatory bowel disease; GI: gastrointestinal; GIT: GI tract; GM: gut microbiota.
Zebrafish (Danio rerio)
The use of zebrafish in biomedical research in general has been increasing steadily over the last couple of decades. As in mice, the limited requirements related to housing space and cost and high fecundity allow studies with larger sample sizes. Physiologically, zebrafish also possess several basic similarities to mammalian hosts including a well-differentiated adaptive immune system,31 a stress response axis typified by the same neuronal transmitters,32 corticosteroid mediators,33 and responses to pharmaceutical interventions in accordance with the responses observed in humans.34
Zebrafish first gained popularity in the field of developmental biology and teratology due to their transparent body wall during embryonic and larval stages, allowing direct visualization of events during organogenesis. That same trait, along with their ex utero development, can be exploited to directly study early events during colonization by the gastrointestinal tract (GIT). Specifically, surface sterilization of embryos with various antibiotic cocktails results in GF zebrafish larvae,35,36 and whole mount larvae can be labeled with in situ hybridization or other assays to localize gene expression or specific cell types in GF and colonized fish.37 Alternatively, fluorescently labeled bacteria can be visualized directly through the transparent body wall.38,39
Zebrafish are also increasingly being used to determine the role of the microbiota in disease models,40 including chemically induced models of inflammatory bowel disease (IBD).41,42 In these and other studies, their aquatic environment allows test compounds and antibiotics to be delivered directly into the tank water.43,44 Notably, zebrafish have gained considerable attention over the last decade for their utility in determining the influence of probiotic bacteria, such as Lactobacillus spp., on stress- and anxiety-related behaviour,45 appetite and feeding behaviour,46 metabolism, reproduction,47,48 immunity and pathogen resistance,49,50 and candidate microbial taxa with possible effects on these parameters can also be delivered via the tank water.36,45,51 Allowing for investigations of the effect of host gene expression on the GM, genetic manipulation can be accomplished in zebrafish using any of the recently developed platforms employed in rodents.52–54
Limitations of the zebrafish model in microbiota-related research include differences in environmental conditions and exposures when compared to humans and other model organisms. Perhaps not surprisingly, the intestinal microbiota of zebrafish is thus quite different from that of mammalian hosts and, unlike most other host species, differs dramatically between institutions (and depending on diet) at the phylum level.36,55–58 Although the fecal microbiota of humans and rodents may vary significantly at the genus level depending on host genetics, geography, age, and environmental factors, it is consistently dominated by the phyla Firmicutes and Bacteroidetes. Depending on the source, the GM of zebrafish is dominated by Proteobacteria and Fusobacteria, with variable presence of Firmicutes, Cyanobacteria, Actinobacteria, and other phyla;55,57 Bacteroidetes are usually, but not always,36,58 negligible. Although they are beyond the scope of the current review, there are also considerable differences between zebrafish and mammalian hosts in gut anatomy that may be of relevance to particular studies such as a lack of Paneth cells and organized lymphoid structures.59 Specifically, the intestinal microbiota stimulates Myd88-mediated signaling in Paneth cells to induce the production of antimicrobial peptides (e.g. defensins),60 which, in turn, modulate microbial ecology within the gut.61,62 Additionally, the GIT of zebrafish lacks the clearly differentiated segments (e.g. stomach, small and large intestines) found in higher vertebrates. In higher vertebrates, there is a distinct division between the small and large intestines with regard to bacterial composition63,64 and the lack of a clear anatomic division between these regions in zebrafish should be considered a limitation with regard to translational research.
Dogs (Canis familiaris)
Companion animals such as dogs possess attributes that make them an appropriate model species for certain microbiota-focused studies. First, the canine GIT is more similar in size and structure to that of humans than the rodent (or zebrafish) GIT. Specifically, although mice and rats have large ceca that serve as sites of hindgut fermentation, dogs are functionally monogastric, like humans. Dogs do possess a well-developed cecal structure when compared to the human cecum (or appendix), although it is relatively small compared to rodent ceca and the bacterial populations present in this region are of unknown relevance to host health. Of importance for translational research, privately owned dogs are often exposed to the same environmental influences as humans and, in some cases, may share compositional similarities to humans in the same household,65,66 begging the question of whether there is direct exchange between humans and their pets or that both host species are exposed to the same factors. Considering the functional capacity of the canine microbiota, metagenomic analyses based on the collective genomic content of the microbiota (rather than marker genes such as 16S rRNA) suggest that diet-induced differences in the composition of the microbiota do not necessitate changes in function, and that canine, human, and mouse fecal microbiota demonstrate a high degree of metabolic and phylogenetic similarity.67 The functional or metabolic capacity of microbial communities can be interrogated using whole metagenome sequencing or metatranscriptomic approaches to reveal microbial genetic content and expression respectively,68 and metabolomic69 or metaproteomic70 approaches to quantify specific classes of molecules in the gut. It is important to remember that these and other functional outputs are essential adjuncts to composition-based studies.
For these and other reasons, microbiota-related research using dogs as the model species is growing and some of the most salient research focuses on gingival and periodontal disease, dental implants, and inflammatory bowel disease. Links between the oral/gingival microbiota and periodontal/gingival disease are intuitive and dogs have proven to be apt model species for decades.71–73 Like humans (and unlike rodents), dogs are subject to diet-induced periodontal disease74,75 and host genetics also influence susceptibility. Although the cultivable oral microbiota of dogs differs depending on whether saliva, gingival sulci, or plaque proper is analysed,76–78 dominant genera include Streptococcus, Staphylococcus, Pseudomonas, Actinomyces, Pasteurella, Neisseria, and Porphyromonas, genera repeatedly identified in the human oral microbiota.79,80 A recent culture-independent analysis of composite oral samples (combined gingival, dental, buccal, and lingual samples) collected from six privately owned dogs revealed Porphyromonas spp. as the dominant colonizer of the canine oral cavity, along with high relative abundance of Fusobacterium and Capnocytophaga spp., among others.81 The apparent discrepancies between these two studies could be explained by differences in sampling techniques, and the presence or absence of microbiological culture prior to sequencing. Of particular interest are the changes associated with canine periodontal disease as similar changes may occur in humans.82–85 For example, despite differences in the composition of the oral microbiota between healthy dogs and humans at the species level, the shift from predominantly Gram-positive aerobic and facultative anaerobic membership to greater numbers of Gram-negative anaerobic species during the development of periodontal disease in both host species suggests common ecological progression and mechanisms.83
Dogs (and cats) also develop chronic inflammatory conditions very similar to human inflammatory bowel disease. Considering the common environmental exposures with humans, there is interest in whether common mechanisms are involved.86 Fusobacteria, Firmicutes, Bacteroidetes, and Proteobacteria constitute the dominant phyla in the healthy canine fecal microbiota87,88 and associations between certain compositional changes and incidence of idiopathic canine IBD have been identified in multiple studies.89,90 Notably, these studies support the notion that chronic inflammatory conditions may be the result of dysbiosis in genetically susceptible individuals, resulting from myriad pressures, and a decrease in the beneficial functions of the GM such as production of short-chain fatty acids.90,91 As in the aforementioned studies on oral microbiota, the fact that the relevant bacterial communities differ at a finer taxonomic resolution between dogs and humans may be irrelevant if common pathways are involved in disease susceptibility. Thus, efforts to enhance the beneficial functions of the GM and prevent or ameliorate inflammation via oral probiotics using canine models is also a growing area of research.
Lastly, dogs may also represent an ideal model species for investigations of the microbiota present in other internal organ systems such as the respiratory tract. Although the lungs and lower airways were historically regarded as sterile environments based on negative culture results, molecular approaches have revealed rich, low-biomass bacterial communities in the healthy lungs of humans,92 cats,93 dogs,94 sheep,95 and mice.96,97 This newly appreciated factor potentially affecting human development, physiology, and disease susceptibility might be most appropriately studied in a canine model based on the comparable size and environmental exposures relative to humans. Moreover, the four dominant genera found in the lungs of healthy humans (i.e. Pseudomonas, Streptococcus, Prevotella, and Fusobacterium)98 were all detected at appreciable relative abundance in canine lung samples,94 suggesting specific attributes of those taxa render them capable of colonization in the lower airways of both species.
There are several limitations to the use of dogs as research models. Clearly, dogs are quite expensive to purchase and house relative to rodents, and studies using purpose-bred research dogs often suffer from poor statistical power owing to this burden. Additionally, there is increased scrutiny of research performed in companion animals from an animal welfare standpoint, and less acceptance by the general public. Lastly, although the use of defined dog breeds can provide some degree of genetic homogeneity, they are nonetheless outbred, and genetic manipulation of dogs is not a readily available practice.
Pigs (Sus scrofa domesticus)
Domestic and miniature pigs have become standard model species in several areas of translational research including xenotransplantation,99 cardiovascular physiology,100,101 and more recently, gastrointestinal physiology102 and immuno-ontogeny.103 The appeal of pigs as research models stems from their comparable size, physiology, and developmental trajectories relative to humans, as well as the ability to manipulate their genome.104,105 As omnivores with a similar GIT structure to humans, the well-characterized fecal microbiota of juvenile and adult domestic pigs106 and other select strains used in research107–109 also possesses compositional similarities to that of humans. Notably, many of these strains are used to study diet-induced obesity in genetically susceptible individuals and the same differences (i.e. an increase in the ratio of Firmicutes to Bacteroidetes) observed between lean and obese humans110 are mirrored in these pig models during the development of obesity.111 Moreover, when comparing the gut microbiota of humans and a wide range of farm animals including horse, cow, goat, sheep, rabbit, and pig via real-time polymerase chain reaction (PCR), probes targeting the prominent butyrate-producing C. leptum (i.e. Clostridial cluster IV or family Ruminococcaceae) and C. coccoides (i.e. Clostridial cluster XIVa or family Lachnospiraceae) groups, as well as the Bacteroides/Prevotella group, were able to discriminate between humans and all farm animals except for pigs and rabbits.112 Thus, with regard to several metabolically active and physiologically relevant clades of gut bacteria, pigs appear to harbor the most compositionally similar fecal microbiota relative to humans.
Aside from rodents, pigs appear to be the only other host species that has been stably colonized with human GM,113 and, unlike so-called “humanized” mice,114 cesarean-delivered pigs (i.e. not colonized with bacteria at parturition) colonized with human GM at birth develop relatively normal gastrointestinal morphology with no overt deficiencies in immune system development.115 In fact, pigs colonized with human GM at birth develop the same or greater numbers of IgA- and IgG-producing cells, CD4+ T helper cells, and MHC class II antigen-presenting cells in the small and large intestines, when compared to control pigs colonized experimentally with porcine GM in a similar fashion.116 Based on these findings, other groups have now begun using human GM-colonized pigs to evaluate the influence of prebiotics and probiotics on community structure and pathogen resistance.117–119 A more comprehensive review of the use of pigs colonized with human GM is provided by Wang and Donovan.113 Similarly, piglets delivered via cesarean section can be monocolonized120,121 or colonized with a highly restricted defined microbiota similar to Altered Schaedler Flora used in mice122,123 to tightly control GM composition.
Given the financial implications of pigs as livestock, the vast majority of GM-focused research in pigs has been done from a production perspective, rather than as biomedical or microbial ecology research. Numerous studies have reported the effects of resistant starch,124 high- and low-fat diets,125 antibiotics126,127 prebiotics,128 probiotics,129,130 and myriad other compounds131 on the GM of pigs. That said, many of these studies may have translational merit owing to the similar functions of the GM, regardless of the host (e.g. butyrate production, stimulation of the immune system, and colonization resistance). A thorough review of pigs as research models of dietary interventions in humans provides a more exhaustive discussion of their beneficial attributes and limitations.132
The major limitations associated with pigs in studies of the microbiota are related to their size and expense to house and feed. These factors, in turn, make large sample sizes difficult to achieve due to the necessary physical space and budget. Moreover, although pigs can be genetically manipulated, it is much more difficult to generate knockout and transgenic pigs than rodents.
Rabbits (Oryctolagus cuniculus)
In general, rabbits are used sparingly and for select purposes as animal models, and the same is true of research targeting the GM. As herbivorous hindgut fermenters, rabbits possess a large and metabolically active cecum, the contents of which have been characterized using culture-independent methods at different stages of life.133–135 Within days of birth, rabbit kits actively ingest fecal material from the doe and thus inoculate their own GIT. Failure to do so results in increased post-weaning mortality and a decreased rate of GM maturation.136 Interestingly, GF rabbits fail to practice coprophagy.137
Beginning in the mid-1960s, there was considerable interest in GF rabbits as axenic models complementary to their rodent predecessors, particularly in the areas of nutrition and digestion.137–139 However, the same factors that drive the use of mice and rats as experimental models in general (i.e. low costs and high fecundity) likely explain the relative dearth of research being performed with GF rabbits; it is simply more cost effective to work with GF mice or rats than it is to work with GF rabbits. That said, there are still investigators taking advantage of the precocious nature of rabbits and hand-rearing cesarean-born rabbits as GF models.140,141
Prior to the development of GF rabbits (or mice),142 rabbits had gained favor as model organisms in investigations of cultivable infectious agents. Specifically, in 1953 De and Chatterje pioneered an acute rabbit model to study GI pathogens wherein a portion of the small intestine was ligated intra-operatively, injected with a pure culture of the candidate bacteria, and placed back in the abdomen with the ligations intact.143 At 24 hours post-surgery, rabbits were euthanized and the ligated section was removed and evaluated to assess in vivo effects of the bacteria in question. Since that initial study on the effects of Vibrio cholerae, numerous pathogenic agents have been studied using similar techniques.144–148 As this approach completely stops intestinal transit of ingesta and the associated microbiota, rabbits must be sacrificed shortly after the procedure. Recently, however, a related approach has been developed that allows for longer studies and can be used to investigate complex microbial communities. Specifically, within 24 hours of birth, the appendix is flushed with a broad-spectrum antibiotic cocktail and ligated at the cecal-appendix junction to prevent influx and colonization of commensal bacteria. At 4 weeks post-ligation, the sterile lumen of the appendix can then be inoculated with cecal contents or pure isolates to assess influence on the immune system and inflammatory responses.140,149–151 Although undeniably artificial, this model is somewhat unique in that it renders only a portion of the gut a sterile environment and allows otherwise normal gut development and physiology.
There are, of course, limitations of rabbits as model species in microbiota-focused research including their cost relative to rodents. As with dogs and pigs, this often precludes large sample sizes. Also, as in swine, although the technology to create knockout and transgenic rabbits does exist, these are not commonly performed procedures and existing mutant rabbit models are few.
Conclusion
In translational medicine, model species are selected based on some similarity to humans with regard to anatomy, molecular basis, pathogenesis, or response to treatment. In studies of host-associated microbiota and interactions between host and microbes, the investigator must now consider other factors. For example, traditional rodent models may be less preferable than dogs when studying the effects of environmental factors on the GM of an outbred population. Similarly, larger omnivorous animals such as pigs may be preferable to rodents for nutrition research or certain procedures. The question of which host species harbors a GM most similar to humans may be, to some degree, irrelevant. Although it is difficult to argue against the possible merits of working with host species in which the GM resembles humans, it is also well established that each host has co-evolved with its cognate GM and xenotransplantation of the GM usually fails to recapitulate the same effects as the “correct” GM on development of the immune system and other physiological parameters.114 Moreover, the concept of a “core microbiota” of any host species, humans included, has largely been supplanted by the concept of a core microbiome. In other words, although the GM composition may vary widely between and within a host species, the functions provided by those microbes are highly conserved. As such, readers are encouraged to fully consider the experimental question being asked during study design and consider whether alternative model species might better serve their goals.
Funding
The author(s) received no financial support for the research, authorship, and/or publication of this article.
Footnotes
Declaration of Conflicting Interests
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
References
- 1.Douglas AE. The Drosophila model for microbiome research. Lab Anim (NY) 2018; 47: 157–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Gerbaba TK, Green-Harrison L and Buret AG. Modeling host-microbiome interactions in Caenorhabditis elegans. J Nematol 2017; 49: 348–356. [PMC free article] [PubMed] [Google Scholar]
- 3.Zhang F, Berg M, Dierking K, et al. Caenorhabditis elegans as a model for microbiome research. Frontiers Microbiol 2017; 8: 485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Chandler JA, Lang JM, Bhatnagar S, et al. Bacterial communities of diverse Drosophila species: Ecological context of a host-microbe model system. PLoS Genet 2011; 7: e1002272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Amato KR. Incorporating the gut microbiota into models of human and non-human primate ecology and evolution. Am J Phys Anthropol 2016; 159: S196–215. [DOI] [PubMed] [Google Scholar]
- 6.Hajishengallis G. The inflammophilic character of the periodontitis-associated microbiota. Mol Oral Microbiol 2014; 29: 248–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Martinez I, Wallace G, Zhang C, et al. Diet-induced metabolic improvements in a hamster model of hypercholesterolemia are strongly linked to alterations of the gut microbiota. Appl Environ Microbiol 2009; 75: 4175–4184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Chu H and Mazmanian SK. Innate immune recognition of the microbiota promotes host-microbial symbiosis. Nat Immunol 2013; 14: 668–675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chaston J and Goodrich-Blair H. Common trends in mutualism revealed by model associations between invertebrates and bacteria. FEMS Microbiol Reviews 2010; 34: 41–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Clarke DJ. Photorhabdus: A model for the analysis of pathogenicity and mutualism. Cellular Microbiol 2008; 10: 2159–2167. [DOI] [PubMed] [Google Scholar]
- 11.Wollenberg AC, Jagdish T, Slough G, et al. Death becomes them: Bacterial community dynamics and stil-bene antibiotic production in cadavers of Galleria mello-nella killed by Heterorhabditis and Photorhabdus spp. Appl Environ Microbiol 2016; 82: 5824–5837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Singh S, Orr D, Divinagracia E, et al. Role of secondary metabolites in establishment of the mutualistic partnership between Xenorhabdus nematophila and the entomopathogenic nematode Steinernema carpocapsae. Appl Environ Microbiol 2015; 81: 754–764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Sicard M, Ferdy JB, Pages S, et al. When mutualists are pathogens: An experimental study of the symbioses between Steinernema (entomopathogenic nematodes) and Xenorhabdus (bacteria). J Evol Biol 2004; 17: 985–993. [DOI] [PubMed] [Google Scholar]
- 14.Rio RV, Maltz M, McCormick B, et al. Symbiont succession during embryonic development of the European medicinal leech, Hirudo verbana. Appl Environ Microbiol 2009; 75: 6890–6895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Graf J, Kikuchi Y and Rio RV. Leeches and their microbiota: Naturally simple symbiosis models. Trends Microbiol 2006; 14: 365–371. [DOI] [PubMed] [Google Scholar]
- 16.McFall-Ngai M. Divining the essence of symbiosis: Insights from the squid-vibrio model. PLoS Biol 2014; 12: e1001783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Schleicher TR and Nyholm SV. Characterizing the host and symbiont proteomes in the association between the Bobtail squid, Euprymna scolopes, and the bacterium, Vibrio fischeri. PloS one 2011; 6: e25649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Beumer KJ and Carroll D. Targeted genome engineering techniques in Drosophila. Methods 2014; 68: 29–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Carroll D. Genome editing by targeted chromosomal mutagenesis. Methods Molecular Biol 2015; 1239: 1–13. [DOI] [PubMed] [Google Scholar]
- 20.Marygold SJ, Crosby MA, Goodman JL, et al. Using FlyBase, a database of Drosophila genes and genomes. Meth Molecular Biol 2016; 1478: 1–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Fire A, Xu S, Montgomery MK, et al. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998; 391: 806–811. [DOI] [PubMed] [Google Scholar]
- 22.Stein L, Sternberg P, Durbin R, et al. WormBase: Network access to the genome and biology of Caenorhabditis elegans. Nucleic Acids Res 2001; 29: 82–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Howe KL, Bolt BJ, Cain S, et al. WormBase 2016: Expanding to enable helminth genomic research. Nucleic Acids Res 2016; 44: D774–780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Cabreiro F, Au C, Leung KY, et al. Metformin retards aging in C. elegans by altering microbial folate and methionine metabolism. Cell 2013; 153: 228–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Greer EL and Brunet A. Different dietary restriction regimens extend lifespan by both independent and overlapping genetic pathways in C. elegans. Aging Cell 2009; 8: 113–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kaeberlein TL, Smith ED, Tsuchiya M, et al. Lifespan extension in Caenorhabditis elegans by complete removal of food. Aging Cell 2006; 5: 487–494. [DOI] [PubMed] [Google Scholar]
- 27.Erkosar B and Leulier F. Transient adult microbiota, gut homeostasis and longevity: Novel insights from the Drosophila model. FEBS Lett 2014; 588: 4250–4257. [DOI] [PubMed] [Google Scholar]
- 28.Clark RI, Salazar A, Yamada R, et al. Distinct shifts in microbiota composition during Drosophila aging impair intestinal function and drive mortality. Cell Rep 2015; 12: 1656–1667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kietz C, Pollari V and Meinander A. Generating germfree Drosophila to study gut-microbe interactions: Protocol to rear Drosophila under axenic conditions. Curr Protoc Toxicol 2018; e52. [DOI] [PubMed] [Google Scholar]
- 30.Berg M, Stenuit B, Ho J, et al. Assembly of the Caenorhabditis elegans gut microbiota from diverse soil microbial environments. ISME J 2016; 10: 1998–2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Trede NS, Langenau DM, Traver D, et al. The use of zebrafish to understand immunity. Immunity 2004; 20: 367–379. [DOI] [PubMed] [Google Scholar]
- 32.Norton WH, Folchert A and Bally-Cuif L. Comparative analysis of serotonin receptor (HTR1A/HTR1B families) and transporter (slc6a4a/b) gene expression in the zebrafish brain. J Comp Neurol 2008; 511: 521–542. [DOI] [PubMed] [Google Scholar]
- 33.Alsop D and Vijayan MM. Molecular programming of the corticosteroid stress axis during zebrafish development. Comp Biochem Physiol A Mol Integr Physiol 2009; 153: 49–54. [DOI] [PubMed] [Google Scholar]
- 34.Wong RY, Oxendine SE and Godwin J. Behavioral and neurogenomic transcriptome changes in wild-derived zebrafish with fluoxetine treatment. BMC Genomics 2013; 14: 348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bates JM, Mittge E, Kuhlman J, et al. Distinct signals from the microbiota promote different aspects of zebrafish gut differentiation. Dev Biol 2006; 297: 374–386. [DOI] [PubMed] [Google Scholar]
- 36.Davis DJ, Bryda EC, Gillespie CH, et al. Microbial modulation of behavior and stress responses in zebrafish larvae. Behavioural Brain Res 2016; 311: 219–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bates JM, Akerlund J, Mittge E, et al. Intestinal alkaline phosphatase detoxifies lipopolysaccharide and prevents inflammation in zebrafish in response to the gut microbiota. Cell Host Microbe 2007; 2: 371–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Russo P, Iturria I, Mohedano ML, et al. Zebrafish gut colonization by mCherry-labelled lactic acid bacteria. Applied Microbiol Biotechnol 2015; 99: 3479–3490. [DOI] [PubMed] [Google Scholar]
- 39.Singer JT, Phennicie RT, Sullivan MJ, et al. Broad-host-range plasmids for red fluorescent protein labeling of gram-negative bacteria for use in the zebrafish model system. Appl Environ Microbiol 2010; 76: 3467–3474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Yang Y, Tomkovich S and Jobin C. Could a swimming creature inform us on intestinal diseases? Lessons from zebrafish. Inflamm Bowel Dis 2014; 20: 956–966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Geiger BM, Gras-Miralles B, Ziogas DC, et al. Intestinal upregulation of melanin-concentrating hormone in TNBS-induced enterocolitis in adult zebrafish. PloS one 2013; 8: e83194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Brugman S, Liu KY, Lindenbergh-Kortleve D, et al. Oxazolone-induced enterocolitis in zebrafish depends on the composition of the intestinal microbiota. Gastroenterol 2009; 137: 1757–1767 e1751. [DOI] [PubMed] [Google Scholar]
- 43.Gaulke CA, Barton CL, Proffitt S, et al. Triclosan exposure is associated with rapid restructuring of the microbiome in adult zebrafish. PloS one 2016; 11: e0154632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Oehlers SH, Flores MV, Okuda KS, et al. A chemical enterocolitis model in zebrafish larvae that is dependent on microbiota and responsive to pharmacological agents. Dev Dyn 2011; 240: 288–298. [DOI] [PubMed] [Google Scholar]
- 45.Davis DJ, Doerr HM, Grzelak AK, et al. Lactobacillus plantarum attenuates anxiety-related behavior and protects against stress-induced dysbiosis in adult zebrafish. Scientific Reports 2016; 6: 33726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Falcinelli S, Rodiles A, Unniappan S, et al. Probiotic treatment reduces appetite and glucose level in the zebrafish model. Scientific Reports 2016; 6: 18061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Qin C, Xu L, Yang Y, et al. Comparison of fecundity and offspring immunity in zebrafish fed Lactobacillus rhamnosus CICC 6141 and Lactobacillus casei BL23. Reproduction 2014; 147: 53–64. [DOI] [PubMed] [Google Scholar]
- 48.Giorgini E, Conti C, Ferraris P, et al. Effects of Lactobacillus rhamnosus on zebrafish oocyte maturation: An FTIR imaging and biochemical analysis. Anal Bioanal Chem 2010; 398: 3063–3072. [DOI] [PubMed] [Google Scholar]
- 49.Wang Y, Ren Z, Fu L, et al. Two highly adhesive lactic acid bacteria strains are protective in zebrafish infected with Aeromonas hydrophila by evocation of gut mucosal immunity. J Applied Microbiol 2016; 120: 441–451. [DOI] [PubMed] [Google Scholar]
- 50.Qin C, Zhang Z, Wang Y, et al. EPSP of L. casei BL23 Protected against the infection caused by Aeromonas veronii via enhancement of immune response in zebrafish. Frontiers Microbiol 2017; 8: 2406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Falcinelli S, Picchietti S, Rodiles A, et al. Lactobacillus rhamnosus lowers zebrafish lipid content by changing gut microbiota and host transcription of genes involved in lipid metabolism. Scientific Reports 2015; 5: 9336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Meng X, Noyes MB, Zhu LJ, et al. Targeted gene inactivation in zebrafish using engineered zinc-finger nucleases. Nat Biotechnol 2008; 26: 695–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Hwang WY, Fu Y, Reyon D, et al. Efficient genome editing in zebrafish using a CRISPR-Cas system. Nat Biotechnol 2013; 31: 227–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Sander JD, Cade L, Khayter C, et al. Targeted gene disruption in somatic zebrafish cells using engineered TALENs. Nat Biotechnol 2011; 29: 697–698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Roeselers G, Mittge EK, Stephens WZ, et al. Evidence for a core gut microbiota in the zebrafish. ISME J 2011; 5: 1595–1608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Hart ML, Meyer A, Johnson PJ, et al. Comparative evaluation of DNA extraction methods from feces of multiple host species for downstream next-generation sequencing. PloS one 2015; 10: e0143334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Rawls JF, Mahowald MA, Ley RE, et al. Reciprocal gut microbiota transplants from zebrafish and mice to germfree recipients reveal host habitat selection. Cell 2006; 127: 423–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Semova I, Carten JD, Stombaugh J, et al. Microbiota regulate intestinal absorption and metabolism of fatty acids in the zebrafish. Cell Host Microbe 2012; 12: 277–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Brugman S. The zebrafish as a model to study intestinal inflammation. Dev Comp Immunol 2016; 64: 82–92. [DOI] [PubMed] [Google Scholar]
- 60.Vaishnava S, Behrendt CL, Ismail AS, et al. Paneth cells directly sense gut commensals and maintain homeostasis at the intestinal host-microbial interface. Proc Natl Acad Sci USA 2008; 105: 20858–20863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Vaishnava S, Yamamoto M, Severson KM, et al. The antibacterial lectin RegIIIgamma promotes the spatial segregation of microbiota and host in the intestine. Science 2011; 334: 255–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Salzman NH, Hung K, Haribhai D, et al. Enteric defensins are essential regulators of intestinal microbial ecology. Nat Immunol 2010; 11: 76–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Ericsson AC, Johnson PJ, Lopes MA, et al. A microbiological map of the healthy equine gastrointestinal tract. PloS one 2016; 11: e0166523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ericsson AC, Gagliardi J, Bouhan D, et al. The influence of caging, bedding, and diet on the composition of the microbiota in different regions of the mouse gut. Scientific Reports 2018; 8: 4065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Song SJ, Lauber C, Costello EK, et al. Cohabiting family members share microbiota with one another and with their dogs. eLife 2013; 2: e00458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Misic AM, Davis MF, Tyldsley AS, et al. The shared microbiota of humans and companion animals as evaluated from Staphylococcus carriage sites. Microbiome 2015; 3: 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Swanson KS, Dowd SE, Suchodolski JS, et al. Phylogenetic and gene-centric metagenomics of the canine intestinal microbiome reveals similarities with humans and mice. ISME J 2011; 5: 639649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Aguiar-Pulido V, Huang W, Suarez-Ulloa V, et al. Metagenomics, metatranscriptomics, and metabolomics approaches for microbiome analysis. Evol Bioinform Online 2016; 12: 5–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Vernocchi P, Del Chierico F and Putignani L. Gut microbiota profiling: Metabolomics-based approach to unravel compounds affecting human health. Frontiers Microbiol 2016; 7: 1144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Petriz BA and Franco OL. Metaproteomics as a complementary approach to gut microbiota in health and disease. Front Chem 2017; 5: 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Lindhe J, Hamp S and Loe H. Experimental periodontitis in the beagle dog. J Periodontal Res 1973; 8: 1–10. [DOI] [PubMed] [Google Scholar]
- 72.Charalampakis G, Abrahamsson I, Carcuac O, et al. Microbiota in experimental periodontitis and periimplantitis in dogs. Clin Oral Implants Res 2014; 25: 1094–1098. [DOI] [PubMed] [Google Scholar]
- 73.Berglundh T, Zitzmann NU and Donati M. Are periimplantitis lesions different from periodontitis lesions? J Clin Periodontol 2011; 38(Suppl 11): 188–202. [DOI] [PubMed] [Google Scholar]
- 74.Gorrel C. Periodontal disease and diet in domestic pets. J Nutrition 1998; 128: 2712S–2714S. [DOI] [PubMed] [Google Scholar]
- 75.Harvey CE. Periodontal disease in dogs. Etiopathogenesis, prevalence, and significance. Vet Clin North Am Small Anim Pract 1998; 28: 1111–1128. (vi). [DOI] [PubMed] [Google Scholar]
- 76.Elliott DR, Wilson M, Buckley CM, et al. Cultivable oral microbiota of domestic dogs. J Clinical Microbiol 2005; 43: 5470–5476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Saphir DA and Carter GR. Gingival flora of the dog with special reference to bacteria associated with bites. J Clinical Microbiology 1976; 3: 344–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wunder JA, Briner WW and Calkins GP. Identification of the cultivable bacteria in dental plaque from the beagle dog. J Dental Res 1976; 55: 1097–1102. [DOI] [PubMed] [Google Scholar]
- 79.Dewhirst FE, Chen T, Izard J, et al. The human oral microbiome. J Bacteriol 2010; 192: 5002–5017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Sakamoto M, Umeda M and Benno Y. Molecular analysis of human oral microbiota. J Periodontal Res 2005; 40: 277–285. [DOI] [PubMed] [Google Scholar]
- 81.Sturgeon A, Stull JW, Costa MC, et al. Metagenomic analysis of the canine oral cavity as revealed by high-throughput pyrosequencing of the 16S rRNA gene. Vet Microbiol 2013; 162: 891–898. [DOI] [PubMed] [Google Scholar]
- 82.Senhorinho GN, Nakano V, Liu C, et al. Occurrence and antimicrobial susceptibility of Porphyromonas spp. and Fusobacterium spp. in dogs with and without periodontitis. Anaerobe 2012; 18: 381–385. [DOI] [PubMed] [Google Scholar]
- 83.Wallis C, Marshall M, Colyer A, et al. A longitudinal assessment of changes in bacterial community composition associated with the development of periodontal disease in dogs. Vet Microbiol 2015; 181: 271–282. [DOI] [PubMed] [Google Scholar]
- 84.Riggio MP, Lennon A, Taylor DJ, et al. Molecular identification of bacteria associated with canine periodontal disease. Vet Microbiol 2011; 150: 394–400. [DOI] [PubMed] [Google Scholar]
- 85.Davis IJ, Wallis C, Deusch O, et al. A cross-sectional survey of bacterial species in plaque from client owned dogs with healthy gingiva, gingivitis or mild periodontitis. PloS one 2013; 8: e83158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Cerquetella M, Spaterna A, Laus F, et al. Inflammatory bowel disease in the dog: Differences and similarities with humans. World J Gastroenterol 2010; 16: 1050–1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Hand D, Wallis C, Colyer A, et al. Pyrosequencing the canine faecal microbiota: Breadth and depth of biodiversity. PloS one 2013; 8: e53115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Suchodolski JS, Camacho J and Steiner JM. Analysis of bacterial diversity in the canine duodenum, jejunum, ileum, and colon by comparative 16S rRNA gene analysis. FEMS Microbiol Ecol 2008; 66: 567–578. [DOI] [PubMed] [Google Scholar]
- 89.Suchodolski JS, Xenoulis PG, Paddock CG, et al. Molecular analysis of the bacterial microbiota in duodenal biopsies from dogs with idiopathic inflammatory bowel disease. Vet Microbiol 2010; 142: 394–400. [DOI] [PubMed] [Google Scholar]
- 90.Vazquez-Baeza Y, Hyde ER, Suchodolski JS, et al. Dog and human inflammatory bowel disease rely on overlapping yet distinct dysbiosis networks. Nat Microbiol 2016; 1: 16177. [DOI] [PubMed] [Google Scholar]
- 91.Suchodolski JS. Diagnosis and interpretation of intestinal dysbiosis in dogs and cats. Veterinary J 2016; 215: 30–37. [DOI] [PubMed] [Google Scholar]
- 92.Dickson RP, Erb-Downward JR, Freeman CM, et al. Bacterial topography of the healthy human lower respiratory tract. mBio 2017; 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Vientos-Plotts AI, Ericsson AC, Rindt H, et al. Dynamic changes of the respiratory microbiota and its relationship to fecal and blood microbiota in healthy young cats. PloS one 2017; 12: e0173818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Ericsson AC, Personett AR, Grobman ME, et al. Composition and predicted metabolic capacity of upper and lower airway microbiota of healthy dogs in relation to the fecal microbiota. PloS one 2016; 11: e0154646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Glendinning L, Wright S, Pollock J, et al. Variability of the sheep lung microbiota. Appl Environ Microbiol 2016; 82: 3225–3238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Poroyko V, Meng F, Meliton A, et al. Alterations of lung microbiota in a mouse model of LPS-induced lung injury. Am J Physiol Lung Cell Mol Physiol 2015; 309: L76–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Gollwitzer ES, Saglani S, Trompette A, et al. Lung microbiota promotes tolerance to allergens in neonates via PD-L1. Nat Med 2014; 20: 642–647. [DOI] [PubMed] [Google Scholar]
- 98.Erb-Downward JR, Thompson DL, Han MK, et al. Analysis of the lung microbiome in the “healthy” smoker and in COPD. PloS one 2011; 6: e16384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Vodicka P, Smetana K Jr., Dvorankova B, et al. The miniature pig as an animal model in biomedical research. Annals New York Acad Sci 2005; 1049: 161–171. [DOI] [PubMed] [Google Scholar]
- 100.Hughes HC. Swine in cardiovascular research. Lab Anim Sci 1986; 36: 348–350. [PubMed] [Google Scholar]
- 101.Gallo M, Poser H, Bottio T, et al. The Vietnamese pig as a translational animal model to evaluate tissue engineered heart valves: Promising early experience. Int J Artif Organs 2017; 40: 142–149. [DOI] [PubMed] [Google Scholar]
- 102.Roura E, Koopmans SJ, Lalles JP, et al. Critical review evaluating the pig as a model for human nutritional physiology. Nutrition Res Rev 2016; 29: 60–90. [DOI] [PubMed] [Google Scholar]
- 103.Sinkora M and Butler JE. Progress in the use of swine in developmental immunology of B and T lymphocytes. Dev Comp Immunol 2016; 58: 1–17. [DOI] [PubMed] [Google Scholar]
- 104.Perleberg C, Kind A and Schnieke A. Genetically engineered pigs as models for human disease. Disease Mod Mechan 2018; 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Ryu J, Prather RS and Lee K. Use of gene-editing technology to introduce targeted modifications in pigs. J Anim Sci Biotechnol 2018; 9: 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Zhao W, Wang Y, Liu S, et al. The dynamic distribution of porcine microbiota across different ages and gastrointestinal tract segments. PloS one 2015; 10: e0117441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Panasevich MR, Wankhade UD, Chintapalli SV, et al. Cecal versus fecal microbiota in Ossabaw swine and implications for obesity. Physiological Genomics 2018; 50: 355–368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Pedersen R, Ingerslev HC, Sturek M, et al. Characterisation of gut microbiota in Ossabaw and Gottingen minipigs as models of obesity and metabolic syndrome. PloS one 2013; 8: e56612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Ji Y, Guo Q, Yin Y, et al. Dietary proline supplementation alters colonic luminal microbiota and bacterial metabolite composition between days 45 and 70 of pregnancy in Huanjiang mini-pigs. J Anim Sci Biotechnol 2018; 9: 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Turnbaugh PJ, Ley RE, Mahowald MA, et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006; 444: 1027–1031. [DOI] [PubMed] [Google Scholar]
- 111.Pedersen R, Andersen AD, Molbak L, et al. Changes in the gut microbiota of cloned and non-cloned control pigs during development of obesity: Gut microbiota during development of obesity in cloned pigs. BMC Microbiol 2013; 13: 30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Furet JP, Firmesse O, Gourmelon M, et al. Comparative assessment of human and farm animal faecal microbiota using real-time quantitative PCR. FEMS Microbio Ecol 2009; 68: 351–362. [DOI] [PubMed] [Google Scholar]
- 113.Wang M and Donovan SM. Human microbiota-associated swine: Current progress and future opportunities. ILAR J / National Research Council, Institute Lab Animal Res 2015; 56: 63–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Chung H, Pamp SJ, Hill JA, et al. Gut immune maturation depends on colonization with a host-specific microbiota. Cell 2012; 149: 1578–1593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Pang X, Hua X, Yang Q, et al. Inter-species transplantation of gut microbiota from human to pigs. ISME J 2007; 1: 156–162. [DOI] [PubMed] [Google Scholar]
- 116.Che C, Pang X, Hua X, et al. Effects of human fecal flora on intestinal morphology and mucosal immunity in human flora-associated piglet. Scand J Immunol 2009; 69: 223–233. [DOI] [PubMed] [Google Scholar]
- 117.Shen J, Zhang B, Wei H, et al. Assessment of the modulating effects of fructo-oligosaccharides on fecal microbiota using human flora-associated piglets. Archives Microbiol 2010; 192: 959–968. [DOI] [PubMed] [Google Scholar]
- 118.Zhang H, Wang H, Shepherd M, et al. Probiotics and virulent human rotavirus modulate the transplanted human gut microbiota in gnotobiotic pigs. Gut Pathog 2014; 6: 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Wen K, Tin C, Wang H, et al. Probiotic Lactobacillus rhamnosus GG enhanced Th1 cellular immunity but did not affect antibody responses in a human gut microbiota transplanted neonatal gnotobiotic pig model. PloS one 2014; 9: e94504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Wochtl B, Gunzer F, Gerner W, et al. Comparison of clinical and immunological findings in gnotobiotic piglets infected with Escherichia coli O104:H4 outbreak strain and EHEC O157:H7. Gut Pathog 2017; 9: 30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Willing BP and Van Kessel AG. Intestinal microbiota differentially affect brush border enzyme activity and gene expression in the neonatal gnotobiotic pig. J Anim Physiol Anim Nutr (Berl) 2009; 93: 586–595. [DOI] [PubMed] [Google Scholar]
- 122.Laycock G, Sait L, Inman C, et al. A defined intestinal colonization microbiota for gnotobiotic pigs. Vet Immunol Immunopathol 2012; 149: 216–224. [DOI] [PubMed] [Google Scholar]
- 123.Huang HC, Vlasova AN, Kumar A, et al. Effect of antibiotic, probiotic, and human rotavirus infection on colonisation dynamics of defined commensal microbiota in a gnotobiotic pig model. Benef Microbes 2018; 9: 71–86. [DOI] [PubMed] [Google Scholar]
- 124.Haenen D, Zhang J, Souza da Silva C, et al. A diet high in resistant starch modulates microbiota composition, SCFA concentrations, and gene expression in pig intestine. J Nutrition 2013; 143: 274–283. [DOI] [PubMed] [Google Scholar]
- 125.Heinritz SN, Weiss E, Eklund M, et al. Intestinal microbiota and microbial metabolites are changed in a pig model fed a high-fat/low-fiber or a low-fat/high-fiber diet. PloS one 2016; 11: e0154329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Looft T, Johnson TA, Allen HK, et al. In-feed antibiotic effects on the swine intestinal microbiome. Proc Natl Acad Sci USA 2012; 109: 1691–1696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Allen HK, Looft T, Bayles DO, et al. Antibiotics in feed induce prophages in swine fecal microbiomes. mBio 2011; 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Berding K, Wang M, Monaco MH, et al. Prebiotics and bioactive milk fractions affect gut development, microbiota, and neurotransmitter expression in piglets. J Pediatr Gastroenterol Nutr 2016; 63: 688–697. [DOI] [PubMed] [Google Scholar]
- 129.Barszcz M, Taciak M and Skomial J. The effects of inulin, dried Jerusalem artichoke tuber and a multispecies probiotic preparation on microbiota ecology and immune status of the large intestine in young pigs. Arch Anim Nutr 2016; 70: 278–292. [DOI] [PubMed] [Google Scholar]
- 130.Riboulet-Bisson E, Sturme MH, Jeffery IB, et al. Effect of Lactobacillus salivarius bacteriocin Abp118 on the mouse and pig intestinal microbiota. PloS one 2012; 7: e31113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Liu H, Ivarsson E, Dicksved J, et al. Inclusion of chicory (Cichorium intybus L.) in pigs’ diets affects the intestinal microenvironment and the gut microbiota. Appl Environ Microbiol 2012; 78: 4102–4109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Heinritz SN, Mosenthin R and Weiss E. Use of pigs as a potential model for research into dietary modulation of the human gut microbiota. Nutrition Res Rev 2013; 26: 191–209. [DOI] [PubMed] [Google Scholar]
- 133.Abecia L, Fondevila M, Balcells J, et al. The effect of lactating rabbit does on the development of the caecal microbial community in the pups they nurture. J Applied Microbiol 2007; 103: 557–564. [DOI] [PubMed] [Google Scholar]
- 134.Monteils V, Cauquil L, Combes S, et al. Potential core species and satellite species in the bacterial community within the rabbit caecum. FEMS Microbiol Ecol 2008; 66: 620–629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Combes S, Michelland RJ, Monteils V, et al. Postnatal development of the rabbit caecal microbiota composition and activity. FEMS Microbiol Ecol 2011; 77: 680–689. [DOI] [PubMed] [Google Scholar]
- 136.Combes S, Gidenne T, Cauquil L, et al. Coprophagous behavior of rabbit pups affects implantation of cecal microbiota and health status. J Animal Sci 2014; 92: 652–665. [DOI] [PubMed] [Google Scholar]
- 137.Yoshida T, Pleasants JR, Reddy BS, et al. Efficiency of digestion in germ-free and conventional rabbits. British J Nutrition 1968; 22: 723–727. [DOI] [PubMed] [Google Scholar]
- 138.Coates ME and O’Donoghue PN. Milk allergy in infant germ-free rabbits. Nature 1967; 213: 307–308. [DOI] [PubMed] [Google Scholar]
- 139.Owen EC, West DW and Coates ME. Metabolism of riboflavine in germ-free and conventional rabbits. British J Nutrition 1970; 24: 259–267. [DOI] [PubMed] [Google Scholar]
- 140.Lanning D, Sethupathi P, Rhee KJ, et al. Intestinal microflora and diversification of the rabbit antibody repertoire. J Immunol 2000; 165: 2012–2019. [DOI] [PubMed] [Google Scholar]
- 141.Schousboe LP, Rasmussen LM and Ovesen T. Induction of mucin and adhesion molecules in middle ear mucosa. Acta Otolaryngol 2001; 121: 596–601. [DOI] [PubMed] [Google Scholar]
- 142.Pleasants JR. History of germfree animal research at Lobund Laboratory, Biology Department, University of Notre Dame, 1928–1965. Proceed Indiana Acad Sci 1965; 75: 220–226. [Google Scholar]
- 143.De SN and Chatterje DN. An experimental study of the mechanism of action of Vibriod cholerae on the intestinal mucous membrane. J Pathol Bacteriol 1953; 66: 559–562. [DOI] [PubMed] [Google Scholar]
- 144.Taylor J, Maltby MP and Payne JM. Factors influencing the response of ligated rabbit-gut segments to injected Escherichia coli. J Pathol Bacteriol 1958; 76: 491–499. [DOI] [PubMed] [Google Scholar]
- 145.Duncan CL, Sugiyama H and Strong DH. Rabbit ileal loop response to strains of Clostridium perfringens. J Bacteriol 1968; 95: 1560–1566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Arm HG, Floyd TM, Faber JE, et al. Use of ligated segments of rabbit small intestine in experimental shigellosis. J Bacteriol 1965; 89: 803–809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Sanyal SC, Singh SJ and Sen PC. Enteropathogenicity of Aeromonas hydrophila and Plesiomonas shigelloides. J Med Microbiol 1975; 8: 195–198. [DOI] [PubMed] [Google Scholar]
- 148.Melling J, Capel BJ, Turnbull PC, et al. Identification of a novel enterotoxigenic activity associated with Bacillus cereus. J Clin Pathol 1976; 29: 938–940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Rhee KJ, Sethupathi P, Driks A, et al. Role of commensal bacteria in development of gut-associated lymphoid tissues and preimmune antibody repertoire. J Immunol 2004; 172: 1118–1124. [DOI] [PubMed] [Google Scholar]
- 150.Shanmugam M, Sethupathi P, Rhee KJ, et al. Bacterial-induced inflammation in germ-free rabbit appendix. Inflamm Bowel Dis 2005; 11: 992–996. [DOI] [PubMed] [Google Scholar]
- 151.Hanson NB and Lanning DK. Microbial induction of B and T cell areas in rabbit appendix. Dev Comp Immunol 2008; 32: 980–991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Cho DY, Mackey C, Van Der Pol WJ, et al. Sinus microanatomy and microbiota in a rabbit model of rhi-nosinusitis. Front Cell Infect Microbiol 2017; 7: 540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Jain R, Kim R, Waldvogel-Thurlow S, et al. The effects of topical agents on paranasal sinus mucosa healing: a rabbit study. Int Forum Allergy Rhinol 2015; 5: 310–317. [DOI] [PubMed] [Google Scholar]
- 154.Zenobia C, Hasturk H, Nguyen D, et al. Porphyromonas gingivalis lipid A phosphatase activity is critical for colonization and increasing the commensal load in the rabbit ligature model. Infect Immun 2014; 82: 650–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Kantarci A, Hasturk H and Van Dyke TE. Animal models for periodontal regeneration and peri-implant responses. Periodontol 2000 2015; 68: 66–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Jyonouchi H, Sun S, Kennedy CA, et al. Localized sinus inflammation in a rabbit sinusitis model induced by Bacteroides fragilis is accompanied by rigorous immune responses. Otolaryngol Head Neck Surg 1999; 120: 869–875. [DOI] [PubMed] [Google Scholar]