Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Aug 19.
Published in final edited form as: Transl Med Aging. 2020 Jul 21;4:103–116.

The microbiome: an emerging key player in aging and longevity

Minhoo Kim 1, Bérénice A Benayoun 1,2,3,4,§
PMCID: PMC7437988  NIHMSID: NIHMS1613770  PMID: 32832742

Abstract

Revolutionary advancements of high-throughput sequencing and metagenomic tools have provided new insights to microbiome function, including a bidirectional relationship between the microbiome and host aging. The intestinal tract is the largest surface in the human body that directly interacts with foreign antigens – it is covered with extremely complex and diverse community of microorganisms, known as the gut microbiome. In a healthy gut, microbial communities maintain a homeostatic metabolism and reside within the host in a state of immune tolerance. Abnormal shifts in the gut microbiome, however, have been implicated in the pathogenesis of age-related chronic diseases, including obesity, cardiovascular diseases and neurodegenerative diseases. The gut microbiome is emerging as a key factor in the aging process. In this review, we describe studies of humans and model organisms that suggest a direct causal role of the gut microbiome on host aging. Additionally, we also discuss sex-dimorphism in the gut microbiome and its possible roles in age-related sex-dimorphic phenotypes. We also provide an overview of widely used microbiome analysis methods and tools which could be used to explore the impact of microbiome remodeling on aging.

1. Introduction

Over a century ago, Elie Metchnikoff proposed that age-related dysfunction could result from increased colon permeability-driven chronic system inflammation [1]. Recent advances in DNA sequencing technologies have allowed investigation of the composition and functional dynamics of complex microbial communities with great resolution and without the need for cultivation [2]. During the past two decades, microbiome research thrived to establish a causal relationship between the microbiome and host aging (Reviewed in [36]).

The microbiota consists of all the microbes (i.e. bacteria, archea, viruses, protozoa and fungi) and a distinct profile of microbiota is found on all host surfaces that are in direct contact with the outside environment (e.g. gut, skin, mouth, vagina, etc.) [715]. The microbiota performs essential functions that contribute to the physiology of the host through a symbiotic relationship [16, 17]. Consequently, perturbations of microbiomes have been proposed to exert negative effects on the host organism. The gut microbiome remains the most extensively studied microbiome and will be the main focus of our review.

Intestinal mucosa is the largest surface of the body that directly interacts with environmental antigens. Thus, the intestinal mucosal immune system monitors the gut environment through a variety of pattern-recognition receptors and is in active communication with the systemic immune system via the local mesenteric lymph nodes [18, 19]. The adult human gut microbiome, composed of approximately 1013 to 1014 micro-organisms, plays various essential roles in the host including degradation of food, lipid storage and metabolism, vitamin synthesis, suppression of harmful microbial species and maintenance of intestinal barrier integrity [2025]. Dysbiosis of the gut microbiome is associated with defects in gut barrier integrity and enhanced pro-inflammatory cytokines [26, 27]. Thus, aberrant alterations of the gut microbiome have been attributed to pathogenesis of various metabolic diseases including adiposity, insulin resistance, atherosclerosis and cardiovascular disease, as well as multiple sclerosis, depression and anxiety [2832].

In this review, we describe changes to the gut microbiome throughout lifespan of the host and key findings that implicate a central role of the gut microbiome in host aging. In addition, we discuss microbiome-relevant biological factors (e.g. sex) that may contribute to aging. Finally, we provide an overview of microbiome collection considerations, data analysis pipelines and potential confounding factors, that must be considered when analyzing and interpreting microbiome data.

2. The microbiome in response to aging and pro-longevity interventions

a. The aging gut microbiome: the human side

The microbiota co-evolves with its host and thus the composition of the microbial community within the intestinal tract fluctuates throughout lifespan, in response to genetic and environmental stimuli [3, 3335]. Based on recent studies that reported the presence of bacteria in the placenta, amniotic cavity and umbilical cord, microbial colonization may initiate as early as in-utero [3638]. During infancy, the gut microbiome undergoes significant fluctuations, which is namely driven by factors including delivery method, feeding, antibiotic exposure, maternal diet and environmental factors [3941]. Colonization of microbial species in the gastrointestinal tract during early stages of life is reported to affect later health of the host organism [42]. Nonetheless, the microbiome composition reaches a stable structure after the first three years, its profile resembling that of an “adult-like” microbiome [4345]. After stable recolonization of the microbiome, diet becomes a major force shaping the microbiome composition of the host throughout early adulthood [46, 47].

In general, healthy adults are reported to present with high levels of bacteria from the Bacteroidetes and Firmicutes phyla, and relatively lower proportions of the Proteobacteria, Actinobacteria, Fusobacteria and Verrucomicrobia phyla [8, 4850]. Bacteroidetes found in the gut mainly functions in polysaccharide metabolism and calorie absorption, whereas Firmicutes are important for production of Short-Chain Fatty Acids [SCFAs] [5155]. The Firmicutes/Bacteroidetes [F/B] ratio is reported to increase from birth to adulthood and studies show that high F/B ratios are associated to a dysbiotic microbiome [34, 56]. Studies have shown that the F/B ratio can be used as an important indicator of gut microbiome state and thus host health [5760].

Clinical studies have reported significant differences in microbial composition between young and elderly human subjects [6, 61]. A key transition from healthy adult to elderly microbiota is characterized by a decrease in microbial diversity. Reduced microbiota diversity in aged individuals have been suggested to result in the expansion of distinct groups of bacteria which has been implicated on the development of age-associated type 1 diabetes mellitus, rheumatoid arthritis and colitis [6264]. However, whether reduced microbiota diversity directly impacts host aging, or is a mere bystander, remains poorly understood.

Generally, in aged individuals, a decrease in Bifidobacterium and Lactobacillus, and increase in Enterobacteriaceae are observed [39, 65, 66]. Such changes in the microbiome structure are believed to result from changed lifestyle, dietary pattern, reduced mobility, weakened immune strength, reduced intestinal functionality, changes in gut morphology, use of medication, recurrent infections and more [27, 34, 35, 39, 61, 65, 67]. However, it is important to note that these generalizations do not apply to certain aged groups from different geological locations or genetic backgrounds [67, 68]. Interestingly, in centenarians and supercentenarians, health-associated bacteria genera, including Bifidobacteria and Christensenella, are especially abundant [69, 70]. Although these observations are correlative, studies in model organisms support pro-longevity and pro-health effects of these microbes [7173]. For example, supplementation of Bifidobacteria to C. elegans resulted in reduced accumulation of lipofuscin, a marker of aging, improved locomotor function and increased longevity [72]. Additionally, transplantation of Christensenella to germ-free mice has been shown to amend obese-associated microbiome and reduce weight gain [73].

b. The aging microbiome in model organisms

Baseline microbial composition of the gut microbiota varies across species and taxa [26, 7476]. However, similar to what has been observed in humans, extensive remodeling of the gut microbiome during aging has also been observed in a number of model organisms, spanning Drosophila melanogaster, the African turquoise killifish Nothobranchius furzeri and mice [26, 7477].

In D. melanogaster, the aging gut microbiome is characterized by an expansion of Gammaproteobacteria [74, 75], and microbiota transplantation from aged donors to young flies leads to reduced longevity. Metagenomics analysis also showed that age-related changes in Drosophila microbial species were somewhat similar to that observed in human inflammatory disorder patients and aged human gastrointestinal tract [75]. For example, increased levels of Enterobacteriaceae, the most abundant family of Gammaproteobacteria, were also observed in aged humans and mouse model of colitis [78, 79]. In a study of the aging microbiome in the African turquoise killifish, the microbiota of young individuals was found to be more enriched in species from the Bacteroidetes, Firmicutes, and Actinobacteria phyla, whereas the aged microbiota was enriched for species from the Proteobacteria phylum [76]. Interestingly, similar to observations in humans, the aging killifish microbiome was also characterized by decreased diversity of the gut microbial community [76]. Additionally, transplantation of microbiome from young to middle-aged killifish improved locomotion and longevity of recipient subjects [76]. The mouse aging microbiome showed a number of shifts in relative abundance of bacteria phyla, including increased presence of Clostridium and decreased levels of Lactobacillaceae as observed with human aging [80]. Increased abundance of Clostridium was also observed in the aging gut microbiome of rats, although (contrary to humans) rats seemed to acquire increased microbial diversity throughout life [81]. Interestingly, the relative proportion of Firmicutes and Bacteroidetes is also altered with aging in the gut microbiome of aging mice [26]. A direct role of the microbiome in promoting overall health in mammals is also suggested by the fact that fecal microbiota transplants from wild-type mice can significantly improve the health and lifespan of progeroid mice [82]. Together, these studies provide a strong rationale for microbiome-based interventions against age-related decline and pathologies.

c. Effects of pro-longevity interventions on the aging microbiome

Modulation of the microbiota is emerging as a potential mechanism underlying pro-health and longevity effects of various interventions (Table 1). Interestingly, a number of pro-longevity interventions seem to have rejuvenating effects on the microbiome. A recurrent effect is the expansion of bacteria from the Lactobacillae taxa, which occurs in the context of independent interventions [83, 84]. Interestingly, a study reported that weight loss in the context of calorie restriction in mice seems to require an intact microbiome [83]. Thus, it will be important to determine whether microbial community remodeling in the context of pro-longevity interventions is a mere bystander, or an actual mediator of pro-health effects.

[Table 1].

Impact of pro-longevity interventions on the aging microbiome.

Pro-longevity intervention Organism Site Effect on microbiome Microbiome profiling Reference
Calorie restriction M. musculus Gut - Gut microbiota required for CR-induced weight loss
- Significant increases in Lactobacillus, Bifidobacterium
- Decreased B/F ratio
V4 16S
V3-V4 16S
[83, 84, 219]
R. norvegicus Gut - Increase of Lactobacillus
- Increased B/F ratio
- Changes in microbial SCFA production (Increased propionogenesis, decreased butyrogenesis and acetogenesis)
V4 16S
Full length 16S
Metaproteomics
[220, 221]
Dwarfism
(Ames)
M. musculus Gut - Increased B/F ratio
V4 16S
[219]
Metformin C. elegans Gut
(food source)
- Changes the E. coli metabolism of folate and methionine
- E. coli required for longevity extension
N/A [222]
H. sapiens Gut - Increased E. coli abundance
- Increased production of SCFAs
V4 16S
Metagenomics
[223225]
M. musculus Gut - Increased B/F ratio- Increased abundance of Lactobacillus V4 16S [226]
Rapamycin M. musculus Oral - Rejuvenation of the oral microbiome V4 16S [13]
M. musculus Gut - Increased prevalence of segmented filamentous bacteria
- Remodeling of specific OTUs
- No change in B/F ratio
- Renders microbiome more similar to that of HFD-treated mice
V4 16S
Full length 16S (Sequencing and PhyloChip)
[227229]
Resveratrol M. musculus Gut - No change in B/F ratio
- Reverses HFD-induced changes in bacterial abundances
Full length 16S [229]
Mediterranean diet H. sapiens Gut - Increased taxa associated to lower frailty in aged humans
- Predicted increase in microbial SCFA production
V3-V4 16S [230]
Acarbose M. musculus Gut - Increased abundance in Muribaculaceae
- Increase in microbial SCFAs, including propionate
V4 16S [231]

HFD: High-fat diet

B/F: Bacteroidetes/ Firmicutes

3. A bidirectional relationship between the gut microbiome and aging?

During the past two decades, studies have provided evidence that age-associated shifts in the gut microbiome contributes to increased predisposition of aged individuals to certain diseases, including cardiovascular diseases, cancer, obesity, cancers, diabetes and neurodegenerative diseases [3, 4, 8587]. Aging is a complicated process that affects physiological, metabolic and immunological functions of the organism and thus is accompanied by inflammation and metabolic dysfunctions [88]. The overall age-related increase in chronic inflammation and deterioration of systemic immune system led to coining the term “inflamm-aging” [89]. A direct causal role of the gut microbiome on host aging has been suggested by a number of studies using various experimental models [76, 90]. In this section, we discuss studies that suggest the existence of a bidirectional relationship between the gut microbiome and host aging.

a. Interaction between the host immune system and the gut microbiome

Through millions of years of evolution, the host and its surrounding microbial environment have co-evolved into a complex organism [17, 18, 91]. Microbes beneficial to the host are able to reside within the host in a state of immune tolerance, whereas those that exert a pathogenic effect activates robust immune responses of the host [17]. The symbiotic co-existence between the host and microbiota is feasible due to the anatomical separation of microbial species from the host by a physical barrier. The intestinal barrier is responsible for adjusting metabolic homeostasis and systemic antimicrobial responses by detecting microbial-cell components and metabolites through its extensive repertoire of innate immune receptors [9296]. For example, activation of pattern-recognition receptors (e.g. Toll-like receptors) by the gut microbe or its products induces the production of antimicrobial peptides and mucus [92]. Perturbations of such receptors have been reported to result in intestinal inflammation and susceptibility to enteric infections [97].

Relevant to aging, decline of the immune system in the aged intestinal epithelium have been suggested to contribute to age-onset dysbiosis [98, 99]. An important characteristic of age-onset dysbiosis is reduced microbiota diversity, which is suggested to lead to an expansion of distinct groups of bacteria [39, 100, 101]. Concurrently, bacteria that is reported to be involved in maintenance of immune tolerance in the gut, such as Bifidobacteria and Lactobacilli, are found in reduced level in aged groups, whereas those that are found in increased levels, such as Enterobacteriaceae and Clostridium, are involved in infection and intestinal inflammation stimulation [27, 66, 102, 103]. Together, these studies suggest that the host immune system shapes not only the host’s immune response to microbiome changes, but also the structure of the microbiome itself [104].

Cumulative evidence has implicated a close functional relationship between the immune system of the host and the microbiome, to an extent that the gut microbiome is important for proper development and expansion of intestinal mucosal and systemic immune system [105, 106]. Supporting the notion that the microbiome can directly shape the immune states of the host, the transcriptomic profile of African turquoise killifish guts derived from animals that received young or old gut microbiota transplants showed clear differences, especially in expression of immune-related genes [76]. Interestingly, studies using germ-free mice models also suggest a bidirectional relationship between the host immune system and the gut microbiome. Germ-free mice showed significant alterations in innate immune system composition compared to classical Specific-Pathogen Free [SPF] mice, including deficiencies in macrophage, monocyte and neutrophil populations [107]. Such alterations of the immune system in germ-free mice were partially rescued when mice were treated with specific bacteria and/or bacterial components (i.e. bacterial polysaccharide), demonstrating a direct role of the gut microbiome on immune system establishment of the host [108]. Interestingly, experiments using germ-free mice also revealed that, in addition to regulating the abundance of immune cells, the microbiome may also regulate bactericide properties of macrophages [26].

b. Increased intestinal barrier permeability with age

Increased permeability of the intestinal barrier with age has been described across animal species, including worms, flies, mice and rats [26, 74, 75, 109111]. Age-related deterioration of intestinal barrier function has been proposed to result in leakage of gut microbes into the systemic circulation, and ultimately lead to increased antigenic load and systemic immune activation [112, 113] (Figure 1). For example, age-associated remodeling of the gut microbiome in mice was shown to result in increased production of pro-inflammatory cytokines and intestinal barrier failure [26]. Consistently, the blood of aged mice contained increased levels of muramyl-dipeptide, a component of Gram-positive and Gram-negative bacteria cell wall [26]. Additionally, in a clinical study on aged type 2 diabetes patients, live gut bacteria were found to translocate into the blood stream, suggesting perturbations of the intestinal barrier integrity [114]. In Drosophila, the age-related increase in Gammaproteobacteria was suggested to lead to increased intestinal permeability, inflammation and mortality [74, 75]. The study showed that regardless of chronological age, intestinal dysbiosis serves as an indicator of age-onset mortality in flies [75].

[Figure 1]. The bidirectional relationship between the gut microbiome and aging.

[Figure 1]

(Left panel) In a healthy gut, balanced microbial composition and intestinal barrier integrity maintains gut homeostasis and contains the microbiota in the intestinal lumen. Microbiota-derived metabolites, including SCFAs, participate in a feedback mechanism with the host immune system to fortify the barrier function, produce mucus and promote intestinal stem cell proliferation. An efficient immune system tolerates the host immune responses to avoid excessive activation. (Right panel) In gut dysbiosis (such as with aging), declined intestinal barrier integrity results in translocation of microbes and microbial particles through the intestinal epithelial cell lining. Reduced microbiota diversity leads to overgrowth of distinct microbes and metabolism instability. Aberrant levels of microbiota-derived metabolites instigate abnormal immune responses resulting in chronic inflammation. SCFA: Short-chain fatty acid.

A number of molecular mechanisms has been suggested to underlie intestinal barrier permeability with age. Mouse studies suggest that host cytokine signaling may play a key role in barrier function breakdown [26]. Indeed, TNF-α signaling was found to play a role in age-related intestinal barrier breakdown, as (i) Tnfa knock-out mice did not accumulate bacteria byproducts in their blood with aging, and (ii) anti- TNF-α therapy led to significant remodeling of the gut microbiota [26]. Mechanistically, age-associated epithelial tight-junctions permeability and declined function of Paneth cells of the intestinal mucosa have been speculated to result in intestinal barrier permeability [115, 116]. However, further research will be needed to fully understand the mechanisms of age-associated increase in gut permeability.

c. Changes in production of microbiome-derived metabolites with age

The gut microbiome plays various essential roles in the host including degradation of food, lipid storage and metabolism, vitamin synthesis, suppression of harmful microbial species and maintenance of intestinal barrier integrity [25]. Microbiome-derived SCFAs, including butyrate, propionate, acetate and valerate, are important energy source for the epithelium and ultimately affects hypoxia-inducible factor-mediated fortification of the epithelial barrier [117]. Interestingly, a decline in SCFA levels, including that of butyrate, were observed in aged humans, whereas centenarians presented with a rearrangement in the population of specific butyrate-producing bacteria [70, 118]. Additionally, the blood and intestine of germ-free mice presented with significantly lower levels of SCFAs compared to conventionally raised mice, supporting a role of the microbiota in regulating host SCFA levels [119121]. For example, studies have shown that administration of butyrate restores the observed abnormal absorptive colonic motor activity and blood-brain barrier permeability in germ-free mice [122, 123]. Microbiota-derived metabolites has also been reported to play a role in intestinal epithelial stem cell proliferation [4]. For example, butyrate and nicotinic acid, both by-products of the gut microbiota, are involved in suppression and promotion of stem cell proliferation in the colon, respectively [124]. In addition, microbiota-derived neurostimulators, including serotonin, glutamate, gamma-aminobutyric acid, have been reported to regulate proliferation of intestinal epithelial stem cells through the enteric nervous system [125]. Collectively, functional products of the microbiota have been implicated in the regulation of intestinal barrier integrity and function.

Other microbiota-derived metabolites have been shown to directly affect numerous systems of the host, although their functions in relation to host aging is in need of further investigation [126]. For example, Trimethylamine N-Oxide [TMAO], a byproduct of microbial metabolism, is associated with cardiometabolic diseases, such as atherosclerosis and type 2 diabetes [127, 128]. Interestingly, microbial metabolites may also affect the host through epigenetic alterations: indeed, microbiota-derived butyrate can affect the immune response of colonic macrophages through the inhibition of histone deacetylases [129, 130]. The microbiome has also been shown to contribute to various neurological conditions through the so-called “microbiota-gut-brain axis” [131]. It is noteworthy that the microbiome can influence behavioral aspects of the host – the level of SCFAs is reported to affect feeding behavior of the host and thus energy homeostasis [132]. Given that diet is a key factor in remodeling of the microbiome structure, an integrative assessment of various physiological conditions of the host is required to precisely understand the functions and effects of the microbiome.

4. Sex-dimorphism in the gut microbiome, and possible impact on aging

Although aging is a conserved process across species and biological sex, accumulating evidence has shown that many age-related phenotypes are sex-dimorphic, and may thus modify aspects of aging between animals of opposite sex [133]. Concurrently, disparities between the sexes are observed in manifestation of certain age-associated diseases, including obesity, multiple sclerosis and Alzheimer’s disease [134, 135]. However, due to experimental pragmatism, still few studies systematically evaluate how sex interacts with aging phenotypes, including age-related microbial dysbiosis. Fundamentally, key phenotypic sex differences are driven by genetic and/or hormonal mechanisms of the host [136]. Intriguingly, recent studies suggest that there may be substantial involvement of the gut microbiome in tuning sex-dimorphic phenotypes.

a. Sex-dimorphism in the gut microbiome

Mouse model studies have shown that the composition of the microbiome starts to diverge between male and female individuals after the onset of puberty [137, 138]. As described above, the gut microbial composition of healthy human adults is reported to consist of high levels of Bacteroidetes and Firmicutes [8, 48, 49]. Interestingly, studies have shown females present with higher F/B ratio compared to that of males [16, 139]. Additionally, Proteobacteria, Veillonella and Blautia are found in higher levels in females compared to males, but in much lower proportions [16, 140]. Although increased F/B ratio is associated with gut dysbiosis, a systematic analysis of such disparities between the two sexes and understanding of its physiological implications are still lacking [34, 56].

Interesting sexually dimorphic phenotypes have been described in studies using germ-free mice models. For example, Non-Obese Diabetes [NOD] model female mice are more prone to spontaneously develop type 1 diabetes compared to NOD model male mice [137]. However, such difference between sexes disappeared when mice were raised in germ-free conditions [141]. In support of this finding, microbiota transplantation of conventionally raised NOD male mice microbiota to germ-free NOD female mice reduced the rate of type 1 diabetes incidence in the recipient mice [137].

b. Sex-dimorphism in host-microbiome communication?

In addition to observed sex differences in microbial communities’ composition, emerging evidence is suggesting that the microbiome may potentiate the expression of sex-dimorphic phenotypes in the hosts. For instance, a recent study utilizing germ-free mice suggested that the microbiome is required to establish sex-dimorphic gene expression patterns in the liver [142]. Another study, also comparing germ-free to SPF mice, found that presence of microbes was required for sex-dimorphic regulation of lipid metabolism in the small intestine of mice [143]. Consistent with sex-dimorphic modulation of the immune system by the microbiome, the transcriptional response of adult microglia – the resident macrophages of the brain – to chronic (i.e. germ-free vs. SPF husbandry) or acute (i.e. antibiotic treatment) microbiota depletion was found to be sex-dimorphic [144]. Intriguingly, a recent study showed that microbiota depletion through antibiotic treatment rescued a number of brain phenotypes only in males in a mouse model of Alzheimer’s disease [145]. Reestablishment of the microbiota reversed the rescue, supporting a direct implication of the microbiota in this phenomenon [145]. Thus, host responses to commensal microbes can be sex-dimorphic, revealing that the microbiome interacts with the biological sex of the host. However, how these sex-dimorphic interactions are modulated during aging remains largely unknown. Future studies investigating the impact of the microbiome on the aging process should systematically include sex as a variable to address this complex question.

c. Interactions between the microbiome and sex-steroid metabolism

The gut microbiome has been proposed to drive estrogen metabolism and regulate the proportions of recirculated and excreted estrogens and estrogen metabolites in the host organism [146148]. The term “estrobolome” has been coined to define “the gene repertoire of the microbiota of the gut capable of metabolizing estrogens” [149, 150]. Indeed, the human gut microbiome is able to hydrolyze estrogen sulfate and glucuronide conjugates [151]. Thus, through manipulation of the gut microbiome, circulating estrogen levels can be shifted in a dosage-dependent manner [148]. Consistently, in a recent study, germ-free female mice presented with significantly lowered levels of 17-β estradiol, the major form of estrogens in females, compared to conventionally raised mice [142]. In the same study, transcriptome analysis of sexual development marker genes and histological studies of follicle development in germ-free female mice indicated that sexual maturation is perturbed in microbiota-depleted mice [142].

Interestingly, estrogens have been shown to impact gut microbiome structure and contribute in gut homeostasis maintenance [152]. In a metabolic syndrome study, the microbiome structure of males and ovariectomized [OVX] females were observed to share similar profiles [153]. When the two test groups were supplemented with 17-β estradiol, both males and OVX females showed alteration of the gut microbiome and suppression of Western diet-induced obesity phenotypes. Collectively, these findings demonstrate a close bidirectional relationship between the gut microbiome and female sex hormones in affecting host health.

5. Microbiome data analysis: experimental and analytical “omics” pipelines

Traditionally, research on microbial interactions was focused on single pathogenic organisms through culture-based methods that capture only a small proportion of the bacterial microbiota [154]. However, recent findings suggest that disease pathogenesis is dependent not only on single pathogens, but also on global changes in the host microbiome [155, 156]. Advancements in next-generation sequencing techniques have enabled culture-independent analyses to capture the global changes in the microbiome. In addition, the advent of various model organisms and experimental tools, including germ-free rodent models and microbiota transplantation methods, have helped characterize microbial communities as key factors in not only dietary metabolism and host nutrition, but also in the pathogenesis of a number of chronic age-associated disease, including diabetes, cardiovascular diseases and neurodegenerative disorders [157161].

As the field expands, microbiome analysis methods and standards are rapidly advancing to allow accurate characterization and interpretation of the microbiome data. This section will provide a primer on microbial sample collection guidelines and widely used microbiome data analysis methods: marker gene, metagenomics and metatranscriptomics analysis (Table 2).

[Table 2].

Pros and cons of marker gene analysis, metagenomics and metatranscriptomics.

Method Cost Analysis Use in aging research Pros Cons
Marker gene analysis Low Simple +++++ - General overview of microbial communities
- Large public datasets available
- Cannot discriminate among live, dead or active features
- Low resolution (genus level)
- Primer choice significantly affect data
Metagenomics High Complex ++ - High resolution (species and strain level)
- Direct measure of functional microbial genes
- Assemble novel microbial genomes
- Cannot discriminate among live, dead or active features
- Prone to host contamination-derived errors
Meta-transcriptomics Medium Complex + - Measure actively transcribing microbial genes
- Directly observe microbial activity changes
- Prone to host mRNA and rRNA contamination
- May fail to detect rare species or low expressed microbial genes

a. Guidelines for microbiome sample collection

For human studies, oral, skin and vaginal samples are generally collected by a physician during a clinic visit – microbial samples can be collected by swabbing the appropriate area using a sterile soft cotton tip or nylon swab [162]. For model organisms, the same type of swab can be used for sample collection. Samples should be immediately flash-frozen and stored at −80°C until further processing [162]. Among various microbial samples, fecal sample collection for gut microbiome analysis presents with the most challenges because on demand collection of fecal samples is difficult. For human fecal samples, various transportation kits, including the Fisherbrand™ Commode Specimen Collection System (Fisher Scientific), OMNIgene Gut kit (DNA Genotek) and Cary Blair Transport Medium (Remel), have been developed in order to preserve microbial composition during shipping from site of sample collection to laboratories for further analysis [163165].

For mouse studies, gut microbiome samples can be collected by picking freshly defecated fecal pellets or extracting fecal pellets from the distal colon after euthanasia. When collecting from the distal colon, fecal pellets need to be homogenized in order to ensure even distribution of microbial species of the colon. Immediate freezing of fecal samples is crucial – storage of microbiome samples at room or higher temperature for extended times results in expansion of specific microbes, such as aerobic microbes but not anaerobes, introducing bias to the data [166]. Studies have shown that microbiome samples are stable for 2 years after being frozen at −80°C [162, 167]. Additionally, it is important to avoid multiple freeze-thaw cycles as it has been shown to affect microbial sample stability [168].

Due to the nature of the microbiome, microbiome data can be significantly affected by external factors, such as lifestyle, diet, medication and physiology. For example, for mice, housing conditions (single- or group-housed), time of cage/bedding change and fasting prior to sample collection can have significant effects on the microbial composition [169]. Additionally, technical variability is a critical issue in microbiome data analysis. Indeed, technical aspects, from DNA extraction to the choice of sequencing platform, have been found/ to substantially affect data reproducibility [170, 171]. Studies have also reported that the choice of DNA extraction kits, contaminants from carriers and storage methods may contribute to data variability [172175]. Thus, standards and controls must be carefully chosen, and complete metadata should be provided along with the raw microbiome sequencing data in order to promote reproducibility and translatability of microbiome research.

b. Microbiome data analysis pipelines: Marker gene, metagenomics and metatranscriptomics analysis

Development of various microbiome-related experimental protocols and analytical tools have provided great opportunities in age-related microbiome research. In this section, we provide a general overview of widely used microbiome data analysis pipelines: marker gene, metagenomics and metatranscriptomics analysis (Table 2). Additionally, we discuss age-related microbiome studies that utilize each analytical method.

Marker gene analysis: high-level, low-resolution overview of microbial composition

Marker genes are conserved genes that contain a highly variable region, flanked by highly conserved regions that serve as primer binding sites, that can be used for detailed identification of microbial species: 16S rDNA PCR amplification is commonly used for bacteria and archaea, and ITS (internal transcribed spacer) for fungi. Marker gene analysis is well-tested, fast and cost-effective. Consequently, a significant proportion of microbiome research, not limited to age-related studies, is based on marker gene analysis data [176]. Additionally, its quantification generally correlates well with genomic content of microbial species [177179]. However, it is important to note that marker gene data is susceptible to biases rising from variable region selection, amplicon size and number of PCR cycles [180, 181]. Thus, choice of amplicon primer will have significant effect on the resolution of data [182], and it is highly recommended to review primers used in the Earth Microbiome Project [183]. Marker gene analysis has been extensively reviewed elsewhere [184]. PhyloChip is another 16S rRNA gene-based method for tracking microbial communities – this microarray-based technique analyzes all nine variable regions of the 16S rRNA gene [185]. In terms of aging research, PhyloChip was used in a study to analyze age-associated changes in the microbial composition in hoatzin, a South American strict folivorous bird [186].

Typically, similar sequences detected from the marker gene analyses are clustered together into Operational Taxonomic Units [OTUs]. This process, called OTU picking, consolidates similar sequences into single features and thus, merges sequence variants and may lose subtle - but real - biological sequence variants. Recent studies have started to prefer the oligotyping method to capture position-specific information from marker gene sequencing data [187]. In this method, exact sequence variants are used to distinguish between closely related, but distinct, taxa. Widely used algorithms such as Deblur and DADA2 implements this method to allow detection of subtle variations between sequences and thus enable greater sensitivity in microbiome analysis from the marker gene method [188, 189]. Marker gene data can also be used to infer putative biological functions of the identified microbial community through predictive functional profiling [179, 190, 191]. This analysis method links the feature-abundance data from the marker gene analysis with available microbial genomes to predict the metagenome content and biological functions. A variety of open source microbial genome references are available, including Silva, Greengenes, and iMGMC [192194]. It is important to note that different microbial genome references are reported to show varying degrees of sensitivity towards different microbial composition arising from specific host organism and/or sampling site [194, 195]. Thus, choice of genome reference can have substantial differences on the final result (Figure 2).

[Figure 2]. General workflow for marker gene analysis.

[Figure 2]

For general marker gene analysis, after sample and metadata collection, choice of primer set should be carefully considered for library preparation. Quality control of sequenced raw data, such as primer and barcode sequence removal, is strongly recommended for accurate data analysis. Features (exact sequence variants) are denoised/clustered to produce a feature-abundance matrix. Feature-abundance matrix can be used for high-level analysis, including taxonomy classification, diversity and differential abundance analysis and predictive functional profiling.

Metagenomics: high-resolution with genome-level information

Metagenomics is used to sequence all the microbial genomes within a given sample. This technique captures all the DNA molecules present in the sample, spanning not only bacteria, but viral and eukaryotic DNA (including that of the host). Compared to marker gene analysis, metagenomics data yields more detailed genomic information and taxonomic resolution. Additionally, this technique allows detection of microbial species to strain level and enables de novo metagenome assembly using short DNA sequence reads, if desired [196, 197]. In addition, metagenomic studies directly enable the detection of the actual gene products present in the sample, thus giving a true window into the biological functions that the microbiome may perform [198, 199]. However, it is substantially more expensive than marker gene analysis, and thus more challenging to scale up to bigger comparative studies. For a thorough review of metagenomics analysis, refer to [200, 201].

Relevant to aging, metagenomic profiling of gut microbiome from young and elderly individuals and centenarians revealed distinct characteristics of microbiome structure and function of the different age groups, identifying 116 microbial genes that significantly correlated with aging [202]. For example, the study showed that a key feature of the gut microbiome profile of centenarians is the overall increase in Proteobacteria and a re-arrangement in Firmicutes compared to young adults [202]. Proteobacteria has been reported to contribute to systemic inflammation [203, 204]. In support of these findings, high levels of plasma interleukin-6 and interleukin-8 were detected in centenarians, although a possible pro-longevity effect of the abundant Proteobacteria in centenarians needs further investigation [70]. Additionally, a study by Pasolli E. et al. conducted a large-scale metagenomic analysis of human microbiome data from 9,316 metagenomes spanning 46 datasets from various populations, body sites, including oral cavity, gut, skin and vagina, and host ages, spanning ages of less than 1 to over 65 [205]. Utilization of such resources will provide new insights and comprehensive understanding of the functional relationship between the microbiome and host aging.

Metatranscriptomics: characterization of microbial gene expression

Metatranscriptomics uses RNA-sequencing technique to profile transcription of microbiomes present in a sample. Metatranscriptomics has been argued to best represent functionality of live microbiome and thus can provide unique insights of the sample [206]. When preparing metatranscriptomics libraries, a number of considerations are required due to host RNA contamination, such as from abundant host rRNA, and preservation of RNA quality. To note, metatranscriptomics may miss the activity of rare species in the samples, due to relatively lower gene expression. For a more thorough review, refer to [207, 208].

To our knowledge, there are only a limited number of metatranscriptomics studies that have been performed in the context of aging. In 2018, a large-scale investigation on 372 human male fecal metatranscriptomics was published [209]. The study involved male subjects from varying age groups, from 18 to 81 of age. Interestingly, the study revealed noticeable differences between metatranscriptomic and metagenomic data [209]. Such finding suggests the importance of multi-omics in microbiome research to accurately characterize taxonomic profiles and interpret physiological effects.

c. Multi-omics and multi-site analysis of the microbiome

Characterization and analysis of the microbiome have revealed immense taxonomic and genomic diversity of the microbiome and implicate more important functions of the microbiome to be revealed in future studies. However, due to the complex functional interaction between the host and the microbiome, establishment of a causal function of the microbiome should be done with caution [210]. A multi-omics approach involving analyses of transcriptomes, proteomes, metabolomes, and immunomes along with microbiome data analysis pipelines discussed above will provide unique insights in characterizing and understanding the roles of the microbiome. Additionally, a comprehensive analysis of different microbiomes, such as skin, oral and vaginal, will be fundamental in elucidating causal functions of the microbiome on host health and longevity.

The human skin is reported to possess a distinctive microbial composition and is estimated to inhabit approximately one billion bacteria per square centimeter of skin [12, 211]. Similar to other microbiomes, the skin microbiome has been shown to undergo various changes in composition throughout lifespan of the host [212]. For example, in a study of cheek microbiomes, specific genera of the Bacteroidetes and Firmicutes phyla were found on the young and specific genera of the Actinobacteria phyla were found only in the older age group [212]. Interestingly, Shibagaki, N. et al. suggested that the age-associated changes in the skin microbiome is largely influenced by the oral bacteria [213]. More specifically, microbial species that were found in greater abundance in the older age group, and thus contributed to differentiate the skin microbiomes of the different age groups, were identified as bacteria frequently found in the oral cavity, including Streptococcus, Rothia and Veillonella [213]. The oral microbiome has been shown to affect the whole body of the host and has been associated with a number of systemic diseases [214]. As an initiation point of digestion, the oral microbiome has been shown to impact the gut microbial composition [215]. Interestingly, the gut microbiome has been shown to affect appetite and feeding behavior of the host through the release of SCFAs [132]. Consequently, in term, the gut microbiome can affect microbial composition of the oral cavity. Together, these studies indicate a functional network among the different microbiomes on a single host and emphasize the importance of systematic investigation in microbiome research.

6. Summary and Perspective

Recent availability of methodological and analytical tools has prompted researchers around the world to investigate the functions of the microbiome and their effects on the well-being of its host. For example, a recently published study developed a human gut microbiome aging clock based on a gut metagenomics data-trained deep learning model [216]. The model was shown to achieve the mean absolute error of 5.91 years, demonstrating that generalizable indicators of age can be derived from microbiome data [216]. With increasing understanding of the importance of the gut microbiome in host longevity, it is anticipated that we will be able to identify and predict risk factors of age-onset gut dysbiosis in the near future.

In this review, we described general changes in the microbiome with age and key findings that implicate a bidirectional relationship between the host and the microbiome. We also discussed sexual dimorphism and various confounding technical factors that must be considered when analyzing and interpreting microbiome data. Over a century ago, Elie Metchnikoff hypothesized that frailty might be delayed by manipulating the gut microbiome with host-friendly bacteria found in yogurt [217]. Intriguingly, a study of yogurt consumption in Japanese individuals observed significant disparities between the two sexes in terms of induced changes to the gut microbiome, emphasizing widespread sex-dimorphism in host physiology [218]. Although it is certain that rigorous and reproducible characterization of microbiome data will offer great opportunities to develop new diagnostic biomarker in aging, key confounding (or biologically-relevant) factors, including sex-dimorphism, will need to be carefully considered. Finally, a comprehensive analysis of different microbiomes (e.g. oral, skin) and how they interact with each other, will be fundamental in elucidating causal functions of the microbiome on host health and longevity.

Acknowledgements

We apologize to authors whose work couldn’t be cited due to space constraints.

Funding

B.A.B is supported by NIA grants R00 AG049934 and R21 AG063739, an innovator grant from the Rose Hills foundation, and a seed grant from the NAVIGAGE foundation.

Footnotes

Conflict of interest

The authors declare that they have no conflict of interest to disclose.

References

  • 1.Metchnikoff II, The prolongation of life: optimistic studies. 2004: Springer Publishing Company. [Google Scholar]
  • 2.Integrative, H.M.P.R.N.C., The Integrative Human Microbiome Project. Nature, 2019. 569 (7758) 641–648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Nagpal R, Mainali R, Ahmadi S, Wang S, Singh R, Kavanagh K, Kitzman DW, Kushugulova A, Marotta F, and Yadav H, Gut microbiome and aging: Physiological and mechanistic insights. Nutr Healthy Aging, 2018. 4 (4) 267–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bana B and Cabreiro F, The Microbiome and Aging. Annu Rev Genet, 2019. 53 239–261. [DOI] [PubMed] [Google Scholar]
  • 5.Aleman FDD and Valenzano DR, Microbiome evolution during host aging. PLoS Pathog, 2019. 15 (7) e1007727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Xu C, Zhu H, and Qiu P, Aging progression of human gut microbiota. BMC Microbiol, 2019. 19 (1) 236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lederberg J and McCray AT, Ome SweetOmics--A genealogical treasury of words. The Scientist, 2001. 15 (7) 8–8. [Google Scholar]
  • 8.Human Microbiome Project, C., Structure, function and diversity of the healthy human microbiome. Nature, 2012. 486 (7402) 207–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Nash AK, Auchtung TA, Wong MC, Smith DP, Gesell JR, Ross MC, Stewart CJ, Metcalf GA, Muzny DM, Gibbs RA, Ajami NJ, and Petrosino JF, The gut mycobiome of the Human Microbiome Project healthy cohort. Microbiome, 2017. 5 (1) 153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Gilbert JA, Blaser MJ, Caporaso JG, Jansson JK, Lynch SV, and Knight R, Current understanding of the human microbiome. Nat Med, 2018. 24 (4) 392–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cresci GA and Bawden E, Gut Microbiome: What We Do and Don’t Know. Nutr Clin Pract, 2015. 30 (6) 734–746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Byrd AL, Belkaid Y, and Segre JA, The human skin microbiome. Nat Rev Microbiol, 2018. 16 (3) 143–155. [DOI] [PubMed] [Google Scholar]
  • 13.An JY, Kerns KA, Ouellette A, Robinson L, Morris HD, Kaczorowski C, Park SI, Mekvanich T, Kang A, McLean JS, Cox TC, and Kaeberlein M, Rapamycin rejuvenates oral health in aging mice. Elife, 2020. 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Curtis MA, Diaz PI, and Van Dyke TE, The role of the microbiota in periodontal disease. Periodontol 2000, 2020. 83 (1) 14–25. [DOI] [PubMed] [Google Scholar]
  • 15.Buchta V, Vaginal microbiome. Ceska Gynekol. 83 (5) 371–379. [PubMed] [Google Scholar]
  • 16.Li M, Wang B, Zhang M, Rantalainen M, Wang S, Zhou H, Zhang Y, Shen J, Pang X, Zhang M, Wei H, Chen Y, Lu H, Zuo J, Su M, Qiu Y, Jia W, Xiao C, Smith LM, Yang S, Holmes E, Tang H, Zhao G, Nicholson JK, Li L, and Zhao L, Symbiotic gut microbes modulate human metabolic phenotypes. Proc Natl Acad Sci U S A, 2008. 105 (6) 2117–2122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Eloe-Fadrosh EA and Rasko DA, The human microbiome: from symbiosis to pathogenesis. Annu Rev Med, 2013. 64 145–163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Backhed F, Ley RE, Sonnenburg JL, Peterson DA, and Gordon JI, Host-bacterial mutualism in the human intestine. Science, 2005. 307 (5717) 1915–1920. [DOI] [PubMed] [Google Scholar]
  • 19.Dethlefsen L, McFall-Ngai M, and Relman DA, An ecological and evolutionary perspective on human-microbe mutualism and disease. Nature, 2007. 449 (7164) 811–818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Bianconi E, Piovesan A, Facchin F, Beraudi A, Casadei R, Frabetti F, Vitale L, Pelleri MC, Tassani S, Piva F, Perez-Amodio S, Strippoli P, and Canaider S, An estimation of the number of cells in the human body. Ann Hum Biol, 2013. 40 (6) 463–471. [DOI] [PubMed] [Google Scholar]
  • 21.Savage DC, Microbial ecology of the gastrointestinal tract. Annu Rev Microbiol, 1977. 31 107–133. [DOI] [PubMed] [Google Scholar]
  • 22.Kumarappah A and Senderovich H, Therapeutic Touch in the Management of Responsive Behavior in Patients With Dementia. Adv Mind Body Med, 2016. 30 (4) 8–13. [PubMed] [Google Scholar]
  • 23.Sender R, Fuchs S, and Milo R, Are We Really Vastly Outnumbered? Revisiting the Ratio of Bacterial to Host Cells in Humans. Cell, 2016. 164 (3) 337–340. [DOI] [PubMed] [Google Scholar]
  • 24.Dobbins JJ, Prescott’s microbiology. Journal of Microbiology & Biology Education: JMBE, 2010. 11 (1) 64. [Google Scholar]
  • 25.Lakshminarayanan B, Stanton C, O’Toole PW, and Ross RP, Compositional dynamics of the human intestinal microbiota with aging: implications for health. J Nutr Health Aging, 2014. 18 (9) 773–786. [DOI] [PubMed] [Google Scholar]
  • 26.Thevaranjan N, Puchta A, Schulz C, Naidoo A, Szamosi JC, Verschoor CP, Loukov D, Schenck LP, Jury J, Foley KP, Schertzer JD, Larche MJ, Davidson DJ, Verdu EF, Surette MG, and Bowdish DME, Age-Associated Microbial Dysbiosis Promotes Intestinal Permeability, Systemic Inflammation, and Macrophage Dysfunction. Cell Host Microbe, 2017. 21 (4) 455–466 e454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Claesson MJ, Jeffery IB, Conde S, Power SE, O’Connor EM, Cusack S, Harris HM, Coakley M, Lakshminarayanan B, O’Sullivan O, Fitzgerald GF, Deane J, O’Connor M, Harnedy N, O’Connor K, O’Mahony D, van Sinderen D, Wallace M, Brennan L, Stanton C, Marchesi JR, Fitzgerald AP, Shanahan F, Hill C, Ross RP, and O’Toole PW, Gut microbiota composition correlates with diet and health in the elderly. Nature, 2012. 488 (7410) 178–184. [DOI] [PubMed] [Google Scholar]
  • 28.Zapata HJ and Quagliarello VJ, The microbiota and microbiome in aging: potential implications in health and age-related diseases. J Am Geriatr Soc, 2015. 63 (4) 776–781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, and Pettersson S, Host-gut microbiota metabolic interactions. Science, 2012. 336 (6086) 1262–1267. [DOI] [PubMed] [Google Scholar]
  • 30.Burcelin R, Serino M, Chabo C, Blasco-Baque V, and Amar J, Gut microbiota and diabetes: from pathogenesis to therapeutic perspective. Acta Diabetol, 2011. 48 (4) 257–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Collins SM, Surette M, and Bercik P, The interplay between the intestinal microbiota and the brain. Nat Rev Microbiol, 2012. 10 (11) 735–742. [DOI] [PubMed] [Google Scholar]
  • 32.Luna RA and Foster JA, Gut brain axis: diet microbiota interactions and implications for modulation of anxiety and depression. Curr Opin Biotechnol, 2015. 32 35–41. [DOI] [PubMed] [Google Scholar]
  • 33.Salazar N, Valdes-Varela L, Gonzalez S, Gueimonde M, and de Los Reyes-Gavilan CG, Nutrition and the gut microbiome in the elderly. Gut Microbes, 2017. 8 (2) 82–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Mariat D, Firmesse O, Levenez F, Guimaraes V, Sokol H, Dore J, Corthier G, and Furet JP, The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol, 2009. 9 123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hopkins MJ and Macfarlane GT, Changes in predominant bacterial populations in human faeces with age and with Clostridium difficile infection. J Med Microbiol, 2002. 51 (5) 448–454. [DOI] [PubMed] [Google Scholar]
  • 36.Jimenez E, Fernandez L, Marin ML, Martin R, Odriozola JM, Nueno-Palop C, Narbad A, Olivares M, Xaus J, and Rodriguez JM, Isolation of commensal bacteria from umbilical cord blood of healthy neonates born by cesarean section. Curr Microbiol, 2005. 51 (4) 270–274. [DOI] [PubMed] [Google Scholar]
  • 37.Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, and Versalovic J, The placenta harbors a unique microbiome. Sci Transl Med, 2014. 6 (237) 237ra265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Collado MC, Rautava S, Aakko J, Isolauri E, and Salminen S, Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid. Sci Rep, 2016. 6 23129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Odamaki T, Kato K, Sugahara H, Hashikura N, Takahashi S, Xiao JZ, Abe F, and Osawa R, Age-related changes in gut microbiota composition from newborn to centenarian: a cross-sectional study. BMC Microbiol, 2016. 16 90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Bokulich NA, Chung J, Battaglia T, Henderson N, Jay M, Li H, Wu DLA,F, Perez-Perez GI, Chen Y, Schweizer W, Zheng X, Contreras M, Dominguez-Bello MG, and Blaser MJ, Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci Transl Med, 2016. 8 (343) 343ra382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Backhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P, Li Y, Xia Y, Xie H, Zhong H, Khan MT, Zhang J, Li J, Xiao L, Al-Aama J, Zhang D, Lee YS, Kotowska D, Colding C, Tremaroli V, Yin Y, Bergman S, Xu X, Madsen L, Kristiansen K, Dahlgren J, and Wang J, Dynamics and Stabilization of the Human Gut Microbiome during the First Year of Life. Cell Host Microbe, 2015. 17 (6) 852. [DOI] [PubMed] [Google Scholar]
  • 42.Tanaka M and Nakayama J, Development of the gut microbiota in infancy and its impact on health in later life. Allergol Int, 2017. 66 (4) 515–522. [DOI] [PubMed] [Google Scholar]
  • 43.Nagpal R, Kurakawa T, Tsuji H, Takahashi T, Kawashima K, Nagata S, Nomoto K, and Yamashiro Y, Evolution of gut Bifidobacterium population in healthy Japanese infants over the first three years of life: a quantitative assessment. Sci Rep, 2017. 7 (1) 10097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Nagpal R, Tsuji H, Takahashi T, Nomoto K, Kawashima K, Nagata S, and Yamashiro Y, Ontogenesis of the Gut Microbiota Composition in Healthy, Full-Term, Vaginally Born and Breast-Fed Infants over the First 3 Years of Life: A Quantitative Bird’s-Eye View. Front Microbiol, 2017. 8 1388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP, Heath AC, Warner B, Reeder J, Kuczynski J, Caporaso JG, Lozupone CA, Lauber C, Clemente JC, Knights D, Knight R, and Gordon JI, Human gut microbiome viewed across age and geography. Nature, 2012. 486 (7402) 222–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S, Collini S, Pieraccini G, and Lionetti P, Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A, 2010. 107 (33) 14691–14696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, Biddinger SB, Dutton RJ, and Turnbaugh PJ, Diet rapidly and reproducibly alters the human gut microbiome. Nature, 2014. 505 (7484) 559–563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hollister EB, Gao C, and Versalovic J, Compositional and functional features of the gastrointestinal microbiome and their effects on human health. Gastroenterology, 2014. 146 (6) 1449–1458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, Gill SR, Nelson KE, and Relman DA, Diversity of the human intestinal microbial flora. Science, 2005. 308 (5728) 1635–1638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Quigley EM, Gut bacteria in health and disease. Gastroenterol Hepatol (N Y), 2013. 9 (9) 560–569. [PMC free article] [PubMed] [Google Scholar]
  • 51.Wexler HM, Bacteroides: the good, the bad, and the nitty-gritty. Clin Microbiol Rev, 2007. 20 (4) 593–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud D-J, and Bakker BM, The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. Journal of lipid research, 2013. 54 (9) 2325–2340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Louis P, Duncan SH, McCrae SI, Millar J, Jackson MS, and Flint HJ, Restricted distribution of the butyrate kinase pathway among butyrate-producing bacteria from the human colon. J Bacteriol, 2004. 186 (7) 2099–2106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Louis P, Young P, Holtrop G, and Flint HJ, Diversity of human colonic butyrate-producing bacteria revealed by analysis of the butyryl-CoA:acetate CoA-transferase gene. Environ Microbiol, 2010. 12 (2) 304–314. [DOI] [PubMed] [Google Scholar]
  • 55.Hooper LV, Midtvedt T, and Gordon JI, How host-microbial interactions shape the nutrient environment of the mammalian intestine. Annu Rev Nutr, 2002. 22 283–307. [DOI] [PubMed] [Google Scholar]
  • 56.Razavi AC, Potts KS, Kelly TN, and Bazzano LA, Sex, gut microbiome, and cardiovascular disease risk. Biol Sex Differ, 2019. 10 (1) 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Indiani C, Rizzardi KF, Castelo PM, Ferraz LFC, Darrieux M, and Parisotto TM, Childhood Obesity and Firmicutes/Bacteroidetes Ratio in the Gut Microbiota: A Systematic Review. Child Obes, 2018. 14 (8) 501–509. [DOI] [PubMed] [Google Scholar]
  • 58.Ley RE, Turnbaugh PJ, Klein S, and Gordon JI, Microbial ecology: human gut microbes associated with obesity. Nature, 2006. 444 (7122) 1022–1023. [DOI] [PubMed] [Google Scholar]
  • 59.Rinninella E, Raoul P, Cintoni M, Franceschi F, Miggiano GAD, Gasbarrini A, and Mele MC, What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms, 2019. 7 (1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Koliada A, Syzenko G, Moseiko V, Budovska L, Puchkov K, Perederiy V, Gavalko Y, Dorofeyev A, Romanenko M, Tkach S, Sineok L, Lushchak O, and Vaiserman A, Association between body mass index and Firmicutes/Bacteroidetes ratio in an adult Ukrainian population. BMC Microbiol, 2017. 17 (1) 120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Claesson MJ, Cusack S, O’Sullivan O, Greene-Diniz R, de Weerd H, Flannery E, Marchesi JR, Falush D, Dinan T, Fitzgerald G, Stanton C, van Sinderen D, O’Connor M, Harnedy N, O’Connor K, Henry C, O’Mahony D, Fitzgerald AP, Shanahan F, Twomey C, Hill C, Ross RP, and O’Toole PW, Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc Natl Acad Sci U S A, 2011. 108 Suppl 1 4586–4591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kostic AD, Gevers D, Siljander H, Vatanen T, Hyotylainen T, Hamalainen AM, Peet A, Tillmann V, Poho P, Mattila I, Lahdesmaki H, Franzosa EA, Vaarala O, de Goffau M, Harmsen H, Ilonen J, Virtanen SM, Clish CB, Oresic M, Huttenhower C, Knip M, D.S. Group, and Xavier RJ, The dynamics of the human infant gut microbiome in development and in progression toward type 1 diabetes. Cell Host Microbe, 2015. 17 (2) 260–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Scher JU, Sczesnak A, Longman RS, Segata N, Ubeda C, Bielski C, Rostron T, Cerundolo V, Pamer EG, Abramson SB, Huttenhower C, and Littman DR, Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife, 2013. 2 e01202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Moschen AR, Gerner RR, Wang J, Klepsch V, Adolph TE, Reider SJ, Hackl H, Pfister A, Schilling J, Moser PL, Kempster SL, Swidsinski A, Orth Holler D, Weiss G, Baines JF, Kaser A, and Tilg H, Lipocalin 2 Protects from Inflammation and Tumorigenesis Associated with Gut Microbiota Alterations. Cell Host Microbe, 2016. 19 (4) 455–469. [DOI] [PubMed] [Google Scholar]
  • 65.O’Toole PW and Jeffery IB, Gut microbiota and aging. Science, 2015. 350 (6265) 1214–1215. [DOI] [PubMed] [Google Scholar]
  • 66.Gavini F, Cayuela C, Antoine J-M, Lecoq C, Lefebvre B, Membré J-M, and Neut C, Differences in the distribution of bifidobacterial and enterobacterial species in human faecal microflora of three different (children, adults, elderly) age groups. Microbial ecology in health and disease, 2001. 13 (1) 40–45. [Google Scholar]
  • 67.Mueller S, Saunier K, Hanisch C, Norin E, Alm L, Midtvedt T, Cresci A, Silvi S, Orpianesi C, Verdenelli MC, Clavel T, Koebnick C, Zunft HJ, Dore J, and Blaut M, Differences in fecal microbiota in different European study populations in relation to age, gender, and country: a cross-sectional study. Appl Environ Microbiol, 2006. 72 (2) 1027–1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Benno Y, Endo K, Mizutani T, Namba Y, Komori T, and Mitsuoka T, Comparison of fecal microflora of elderly persons in rural and urban areas of Japan. Appl Environ Microbiol, 1989. 55 (5) 1100–1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Biagi E, Franceschi C, Rampelli S, Severgnini M, Ostan R, Turroni S, Consolandi C, Quercia S, Scurti M, Monti D, Capri M, Brigidi P, and Candela M, Gut Microbiota and Extreme Longevity. Curr Biol, 2016. 26 (11) 1480–1485. [DOI] [PubMed] [Google Scholar]
  • 70.Biagi E, Nylund L, Candela M, Ostan R, Bucci L, Pini E, Nikkila J, Monti D, Satokari R, Franceschi C, Brigidi P, and De Vos W, Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians. PLoS One, 2010. 5 (5) e10667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Komura T, Yasui C, Miyamoto H, and Nishikawa Y, Caenorhabditis elegans as an alternative model host for legionella pneumophila, and protective effects of Bifidobacterium infantis. Appl Environ Microbiol, 2010. 76 (12) 4105–4108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Komura T, Ikeda T, Yasui C, Saeki S, and Nishikawa Y, Mechanism underlying prolongevity induced by bifidobacteria in Caenorhabditis elegans. Biogerontology, 2013. 14 (1) 73–87. [DOI] [PubMed] [Google Scholar]
  • 73.Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren O, Blekhman R, Beaumont M, Van Treuren W, Knight R, Bell JT, Spector TD, Clark AG, and Ley RE, Human genetics shape the gut microbiome. Cell, 2014. 159 (4) 789–799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Clark RI, Salazar A, Yamada R, Fitz-Gibbon S, Morselli M, Alcaraz J, Rana A, Rera M, Pellegrini M, Ja WW, and Walker DW, Distinct Shifts in Microbiota Composition during Drosophila Aging Impair Intestinal Function and Drive Mortality. Cell Rep, 2015. 12 (10) 1656–1667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Rera M, Clark RI, and Walker DW, Intestinal barrier dysfunction links metabolic and inflammatory markers of aging to death in Drosophila. Proc Natl Acad Sci U S A, 2012. 109 (52) 21528–21533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Smith P, Willemsen D, Popkes M, Metge F, Gandiwa E, Reichard M, and Valenzano DR, Regulation of life span by the gut microbiota in the short-lived African turquoise killifish. Elife, 2017. 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Cabreiro F and Gems D, Worms need microbes too: microbiota, health and aging in Caenorhabditis elegans. EMBO Mol Med, 2013. 5 (9) 1300–1310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.van Tongeren SP, Slaets JP, Harmsen HJ, and Welling GW, Fecal microbiota composition and frailty. Appl Environ Microbiol, 2005. 71 (10) 6438–6442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Carvalho FA, Koren O, Goodrich JK, Johansson ME, Nalbantoglu I, Aitken JD, Su Y, Chassaing B, Walters WA, Gonzalez A, Clemente JC, Cullender TC, Barnich N, Darfeuille-Michaud A, Vijay-Kumar M, Knight R, Ley RE, and Gewirtz AT, Transient inability to manage proteobacteria promotes chronic gut inflammation in TLR5-deficient mice. Cell Host Microbe, 2012. 12 (2) 139–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Langille MG, Meehan CJ, Koenig JE, Dhanani AS, Rose RA, Howlett SE, and Beiko RG, Microbial shifts in the aging mouse gut. Microbiome, 2014. 2 (1) 50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Flemer B, Gaci N, Borrel G, Sanderson IR, Chaudhary PP, Tottey W, O’Toole PW, and Brugere JF, Fecal microbiota variation across the lifespan of the healthy laboratory rat. Gut Microbes, 2017. 8 (5) 428–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Barcena C, Valdes-Mas R, Mayoral P, Garabaya C, Durand S, Rodriguez F, Fernandez-Garcia MT, Salazar N, Nogacka AM, Garatachea N, Bossut N, Aprahamian F, Lucia A, Kroemer G, Freije JMP, Quiros PM, and Lopez-Otin C, Healthspan and lifespan extension by fecal microbiota transplantation into progeroid mice. Nat Med, 2019. 25 (8) 1234–1242. [DOI] [PubMed] [Google Scholar]
  • 83.Wang S, Huang M, You X, Zhao J, Chen L, Wang L, Luo Y, and Chen Y, Gut microbiota mediates the anti-obesity effect of calorie restriction in mice. Sci Rep, 2018. 8 (1) 13037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Pan F, Zhang L, Li M, Hu Y, Zeng B, Yuan H, Zhao L, and Zhang C, Predominant gut Lactobacillus murinus strain mediates anti-inflammaging effects in calorie-restricted mice. Microbiome, 2018. 6 (1) 54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Blumberg R and Powrie F, Microbiota, disease, and back to health: a metastable journey. Sci Transl Med, 2012. 4 (137) 137rv137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Carding S, Verbeke K, Vipond DT, Corfe BM, and Owen LJ, Dysbiosis of the gut microbiota in disease. Microb Ecol Health Dis, 2015. 26 26191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Khan MT, Nieuwdorp M, and Backhed F, Microbial modulation of insulin sensitivity. Cell Metab, 2014. 20 (5) 753–760. [DOI] [PubMed] [Google Scholar]
  • 88.Franceschi C and Campisi J, Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci, 2014. 69 Suppl 1 S4–9. [DOI] [PubMed] [Google Scholar]
  • 89.Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca M, Ottaviani E, and De Benedictis G, Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci, 2000. 908 244–254. [DOI] [PubMed] [Google Scholar]
  • 90.Macpherson AJ and Harris NL, Interactions between commensal intestinal bacteria and the immune system. Nature Reviews Immunology, 2004. 4 (6) 478–485. [DOI] [PubMed] [Google Scholar]
  • 91.Van den Abbeele P, Van de Wiele T, Verstraete W, and Possemiers S, The host selects mucosal and luminal associations of coevolved gut microorganisms: a novel concept. FEMS Microbiol Rev, 2011. 35 (4) 681–704. [DOI] [PubMed] [Google Scholar]
  • 92.Turner JR, Intestinal mucosal barrier function in health and disease. Nat Rev Immunol, 2009. 9 (11) 799–809. [DOI] [PubMed] [Google Scholar]
  • 93.Pott J and Hornef M, Innate immune signalling at the intestinal epithelium in homeostasis and disease. EMBO Rep, 2012. 13 (8) 684–698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Belkaid Y and Naik S, Compartmentalized and systemic control of tissue immunity by commensals. Nat Immunol, 2013. 14 (7) 646–653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Abt MC, Osborne LC, Monticelli LA, Doering TA, Alenghat T, Sonnenberg GF, Paley MA, Antenus M, Williams KL, Erikson J, Wherry EJ, and Artis D, Commensal bacteria calibrate the activation threshold of innate antiviral immunity. Immunity, 2012. 37 (1) 158–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Yoo BB and Mazmanian SK, The Enteric Network: Interactions between the Immune and Nervous Systems of the Gut. Immunity, 2017. 46 (6) 910–926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Man SM, Zhu Q, Zhu L, Liu Z, Karki R, Malik A, Sharma D, Li L, Malireddi RK, Gurung P, Neale G, Olsen SR, Carter RA, McGoldrick DJ, Wu G, Finkelstein D, Vogel P, Gilbertson RJ, and Kanneganti TD, Critical Role for the DNA Sensor AIM2 in Stem Cell Proliferation and Cancer. Cell, 2015. 162 (1) 45–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Guo L, Karpac J, Tran SL, and Jasper H, PGRP-SC2 promotes gut immune homeostasis to limit commensal dysbiosis and extend lifespan. Cell, 2014. 156 (1–2) 109–122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Levy M, Kolodziejczyk AA, Thaiss CA, and Elinav E, Dysbiosis and the immune system. Nat Rev Immunol, 2017. 17 (4) 219–232. [DOI] [PubMed] [Google Scholar]
  • 100.Bartosch S, Fite A, Macfarlane GT, and McMurdo ME, Characterization of bacterial communities in feces from healthy elderly volunteers and hospitalized elderly patients by using real-time PCR and effects of antibiotic treatment on the fecal microbiota. Appl Environ Microbiol, 2004. 70 (6) 3575–3581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Jackson MA, Jeffery IB, Beaumont M, Bell JT, Clark AG, Ley RE, O’Toole PW, Spector TD, and Steves CJ, Signatures of early frailty in the gut microbiota. Genome Med, 2016. 8 (1) 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Kumar A, Wu H, Collier-Hyams LS, Hansen JM, Li T, Yamoah K, Pan ZQ, Jones DP, and Neish AS, Commensal bacteria modulate cullin-dependent signaling via generation of reactive oxygen species. EMBO J, 2007. 26 (21) 4457–4466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Pamer EG, Immune responses to commensal and environmental microbes. Nat Immunol, 2007. 8 (11) 1173–1178. [DOI] [PubMed] [Google Scholar]
  • 104.Karczewski J, Poniedzialek B, Adamski Z, and Rzymski P, The effects of the microbiota on the host immune system. Autoimmunity, 2014. 47 (8) 494–504. [DOI] [PubMed] [Google Scholar]
  • 105.Belkaid Y and Hand TW, Role of the microbiota in immunity and inflammation. Cell, 2014. 157 (1) 121–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, and Medzhitov R, Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell, 2004. 118 (2) 229–241. [DOI] [PubMed] [Google Scholar]
  • 107.Khosravi A, Yanez A, Price JG, Chow A, Merad M, Goodridge HS, and Mazmanian SK, Gut microbiota promote hematopoiesis to control bacterial infection. Cell Host Microbe, 2014. 15 (3) 374–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Mazmanian SK, Liu CH, Tzianabos AO, and Kasper DL, An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell, 2005. 122 (1) 107–118. [DOI] [PubMed] [Google Scholar]
  • 109.Mullin JM, Valenzano MC, Verrecchio JJ, and Kothari R, Age- and diet-related increase in transepithelial colon permeability of Fischer 344 rats. Dig Dis Sci, 2002. 47 (10) 2262–2270. [DOI] [PubMed] [Google Scholar]
  • 110.Annaert P, Brouwers J, Bijnens A, Lammert F, Tack J, and Augustijns P, Ex vivo permeability experiments in excised rat intestinal tissue and in vitro solubility measurements in aspirated human intestinal fluids support age-dependent oral drug absorption. Eur J Pharm Sci, 2010. 39 (1–3) 15–22. [DOI] [PubMed] [Google Scholar]
  • 111.Gelino S, Chang JT, Kumsta C, She X, Davis A, Nguyen C, Panowski S, and Hansen M, Intestinal Autophagy Improves Healthspan and Longevity in C. elegans during Dietary Restriction. PLoS Genet, 2016. 12 (7) e1006135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Amar J, Chabo C, Waget A, Klopp P, Vachoux C, Bermudez-Humaran LG, Smirnova N, Berge M, Sulpice T, Lahtinen S, Ouwehand A, Langella P, Rautonen N, Sansonetti PJ, and Burcelin R, Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: molecular mechanisms and probiotic treatment. EMBO Mol Med, 2011. 3 (9) 559–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Raybould HE, Gut microbiota, epithelial function and derangements in obesity. J Physiol, 2012. 590 (3) 441–446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Sato J, Kanazawa A, Ikeda F, Yoshihara T, Goto H, Abe H, Komiya K, Kawaguchi M, Shimizu T, Ogihara T, Tamura Y, Sakurai Y, Yamamoto R, Mita T, Fujitani Y, Fukuda H, Nomoto K, Takahashi T, Asahara T, Hirose T, Nagata S, Yamashiro Y, and Watada H, Gut dysbiosis and detection of “live gut bacteria” in blood of Japanese patients with type 2 diabetes. Diabetes Care, 2014. 37 (8) 2343–2350. [DOI] [PubMed] [Google Scholar]
  • 115.Al-Sadi RM and Ma TY, IL-1beta causes an increase in intestinal epithelial tight junction permeability. J Immunol, 2007. 178 (7) 4641–4649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Man AL, Gicheva N, and Nicoletti C, The impact of ageing on the intestinal epithelial barrier and immune system. Cell Immunol, 2014. 289 (1–2) 112–118. [DOI] [PubMed] [Google Scholar]
  • 117.Kelly CJ, Zheng L, Campbell EL, Saeedi B, Scholz CC, Bayless AJ, Wilson KE, Glover LE, Kominsky DJ, Magnuson A, Weir TL, Ehrentraut SF, Pickel C, Kuhn KA, Lanis JM, Nguyen V, Taylor CT, and Colgan SP, Crosstalk between Microbiota-Derived Short-Chain Fatty Acids and Intestinal Epithelial HIF Augments Tissue Barrier Function. Cell Host Microbe, 2015. 17 (5) 662–671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Tiihonen K, Ouwehand AC, and Rautonen N, Human intestinal microbiota and healthy ageing. Ageing Res Rev, 2010. 9 (2) 107–116. [DOI] [PubMed] [Google Scholar]
  • 119.Hoverstad T and Midtvedt T, Short-chain fatty acids in germfree mice and rats. J Nutr, 1986. 116 (9) 1772–1776. [DOI] [PubMed] [Google Scholar]
  • 120.Fachi JL, Felipe JS, Pral LP, da Silva BK, Correa RO, de Andrade MCP, da Fonseca DM, Basso PJ, Camara NOS, de Sales ESEL, Dos Santos Martins F, Guima SES, Thomas AM, Setubal JC, Magalhaes YT, Forti FL, Candreva T, Rodrigues HG, de Jesus MB, Consonni SR, Farias ADS, Varga-Weisz P, and Vinolo MAR, Butyrate Protects Mice from Clostridium difficile-Induced Colitis through an HIF-1-Dependent Mechanism. Cell Rep, 2019. 27 (3) 750–761 e757. [DOI] [PubMed] [Google Scholar]
  • 121.Erny D, Hrabe de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, Keren-Shaul H, Mahlakoiv T, Jakobshagen K, Buch T, Schwierzeck V, Utermohlen O, Chun E, Garrett WS, McCoy KD, Diefenbach A, Staeheli P, Stecher B, Amit I, and Prinz M, Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci, 2015. 18 (7) 965–977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Vincent AD, Wang XY, Parsons SP, Khan WI, and Huizinga JD, Abnormal absorptive colonic motor activity in germ-free mice is rectified by butyrate, an effect possibly mediated by mucosal serotonin. Am J Physiol Gastrointest Liver Physiol, 2018. 315 (5) G896–G907. [DOI] [PubMed] [Google Scholar]
  • 123.Braniste V, Al-Asmakh M, Kowal C, Anuar F, Abbaspour A, Toth M, Korecka A, Bakocevic N, Ng LG, Kundu P, Gulyas B, Halldin C, Hultenby K, Nilsson H, Hebert H, Volpe BT, Diamond B, and Pettersson S, The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med, 2014. 6 (263) 263ra158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Kaiko GE, Ryu SH, Koues OI, Collins PL, Solnica-Krezel L, Pearce EJ, Pearce EL, Oltz EM, and Stappenbeck TS, The Colonic Crypt Protects Stem Cells from Microbiota-Derived Metabolites. Cell, 2016. 167 (4) 1137. [DOI] [PubMed] [Google Scholar]
  • 125.Peck BCE, Shanahan MT, Singh AP, and Sethupathy P, Gut Microbial Influences on the Mammalian Intestinal Stem Cell Niche. Stem Cells Int, 2017. 2017 5604727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Koh A and Backhed F, From Association to Causality: the Role of the Gut Microbiota and Its Functional Products on Host Metabolism. Mol Cell, 2020. 78 (4) 584–596. [DOI] [PubMed] [Google Scholar]
  • 127.Shan Z, Sun T, Huang H, Chen S, Chen L, Luo C, Yang W, Yang X, Yao P, and Cheng J, Association between microbiota-dependent metabolite trimethylamine-N-oxide and type 2 diabetes. The American journal of clinical nutrition, 2017. 106 (3) 888–894. [DOI] [PubMed] [Google Scholar]
  • 128.Tang WH, Kitai T, and Hazen SL, Gut Microbiota in Cardiovascular Health and Disease. Circ Res, 2017. 120 (7) 1183–1196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Chang PV, Hao L, Offermanns S, and Medzhitov R, The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc Natl Acad Sci U S A, 2014. 111 (6) 2247–2252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Qin Y and Wade PA, Crosstalk between the microbiome and epigenome: messages from bugs. The Journal of Biochemistry, 2018. 163 (2) 105–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Jameson KG, Olson CA, Kazmi SA, and Hsiao EY, Toward Understanding Microbiome-Neuronal Signaling. Mol Cell, 2020. 78 (4) 577–583. [DOI] [PubMed] [Google Scholar]
  • 132.Byrne CS, Chambers ES, Morrison DJ, and Frost G, The role of short chain fatty acids in appetite regulation and energy homeostasis. Int J Obes (Lond), 2015. 39 (9) 1331–1338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Sampathkumar NK, Bravo JI, Chen Y, Danthi PS, Donahue EK, Lai RW, Lu R, Randall LT, Vinson N, and Benayoun BA, Widespread sex dimorphism in aging and age-related diseases. Hum Genet, 2020. 139 (3) 333–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Ostan R, Monti D, Gueresi P, Bussolotto M, Franceschi C, and Baggio G, Gender, aging and longevity in humans: an update of an intriguing/neglected scenario paving the way to a gender-specific medicine. Clin Sci (Lond), 2016. 130 (19) 1711–1725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Manolopoulos KN, Karpe F, and Frayn KN, Gluteofemoral body fat as a determinant of metabolic health. Int J Obes (Lond), 2010. 34 (6) 949–959. [DOI] [PubMed] [Google Scholar]
  • 136.Rose E, Gedela M, Miller N, and Carpenter PL, Pregnancy-Related Spontaneous Coronary Artery Dissection: A Case Series and Literature Review. J Emerg Med, 2017. 52 (6) 867–874. [DOI] [PubMed] [Google Scholar]
  • 137.Markle JG, Frank DN, Mortin-Toth S, Robertson CE, Feazel LM, Rolle-Kampczyk U, von Bergen M, McCoy KD, Macpherson AJ, and Danska JS, Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science, 2013. 339 (6123) 1084–1088. [DOI] [PubMed] [Google Scholar]
  • 138.Steegenga WT, Mischke M, Lute C, Boekschoten MV, Pruis MG, Lendvai A, Verkade HJ, Boekhorst J, Timmerman HM, Plosch T, and Muller M, Sexually dimorphic characteristics of the small intestine and colon of prepubescent C57BL/6 mice. Biol Sex Differ, 2014. 5 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Dominianni C, Sinha R, Goedert JJ, Pei Z, Yang L, Hayes RB, and Ahn J, Sex, body mass index, and dietary fiber intake influence the human gut microbiome. PLoS One, 2015. 10 (4) e0124599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Haro C, Rangel-Zuniga OA, Alcala-Diaz JF, Gomez-Delgado F, Perez-Martinez P, Delgado-Lista J, Quintana-Navarro GM, Landa BB, Navas-Cortes JA, Tena-Sempere M, Clemente JC, Lopez-Miranda J, Perez-Jimenez F, and Camargo A, Intestinal Microbiota Is Influenced by Gender and Body Mass Index. PLoS One, 2016. 11 (5) e0154090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Yurkovetskiy L, Burrows M, Khan AA, Graham L, Volchkov P, Becker L, Antonopoulos D, Umesaki Y, and Chervonsky AV, Gender bias in autoimmunity is influenced by microbiota. Immunity, 2013. 39 (2) 400–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Weger BD, Gobet C, Yeung J, Martin E, Jimenez S, Betrisey B, Foata F, Berger B, Balvay A, Foussier A, Charpagne A, Boizet-Bonhoure B, Chou CJ, Naef F, and Gachon F, The Mouse Microbiome Is Required for Sex-Specific Diurnal Rhythms of Gene Expression and Metabolism. Cell Metab, 2019. 29 (2) 362–382 e368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Baars A, Oosting A, Lohuis M, Koehorst M, El Aidy S, Hugenholtz F, Smidt H, Mischke M, Boekschoten MV, Verkade HJ, Garssen J, van der Beek EM, Knol J, de Vos P, van Bergenhenegouwen J, and Fransen F, Sex differences in lipid metabolism are affected by presence of the gut microbiota. Sci Rep, 2018. 8 (1) 13426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Thion MS, Low D, Silvin A, Chen J, Grisel P, Schulte-Schrepping J, Blecher R, Ulas T, Squarzoni P, Hoeffel G, Coulpier F, Siopi E, David FS, Scholz C, Shihui F, Lum J, Amoyo AA, Larbi A, Poidinger M, Buttgereit A, Lledo PM, Greter M, Chan JKY, Amit I, Beyer M, Schultze JL, Schlitzer A, Pettersson S, Ginhoux F, and Garel S, Microbiome Influences Prenatal and Adult Microglia in a Sex-Specific Manner. Cell, 2018. 172 (3) 500–516 >e516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Dodiya HB, Kuntz T, Shaik SM, Baufeld C, Leibowitz J, Zhang X, Gottel N, Zhang X, Butovsky O, Gilbert JA, and Sisodia SS, Sex-specific effects of microbiome perturbations on cerebral Abeta amyloidosis and microglia phenotypes. J Exp Med, 2019. 216 (7) 1542–1560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Adlercreutz H, Martin F, Jarvenpaa P, and Fotsis T, Steroid absorption and enterohepatic recycling. Contraception, 1979. 20 (3) 201–223. [DOI] [PubMed] [Google Scholar]
  • 147.Eriksson H and Gustafsson JA, Steroids in germfree and conventional rats. Distribution and excretion of labelled pregnenolone and corticosterone in male and female rats. Eur J Biochem, 1970. 15 (1) 132–139. [DOI] [PubMed] [Google Scholar]
  • 148.Lombardi P, Goldin B, Boutin E, and Gorbach SL, Metabolism of androgens and estrogens by human fecal microorganisms. J Steroid Biochem, 1978. 9 (8) 795–801. [DOI] [PubMed] [Google Scholar]
  • 149.Plottel CS and Blaser MJ, Microbiome and malignancy . Cell Host Microbe, 2011. 10 (4) 324–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Baker JM, Al-Nakkash L, and Herbst-Kralovetz MM, Estrogen-gut microbiome axis: Physiological and clinical implications. Maturitas, 2017. 103 45–53. [DOI] [PubMed] [Google Scholar]
  • 151.Ervin SM, Li H, Lim L, Roberts LR, Liang X, Mani S, and Redinbo MR, Gut microbial beta-glucuronidases reactivate estrogens as components of the estrobolome that reactivate estrogens. J Biol Chem, 2019. 294 (49) 18586–18599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Vemuri R, Sylvia KE, Klein SL, Forster SC, Plebanski M, Eri R, and Flanagan KL, The microgenderome revealed: sex differences in bidirectional interactions between the microbiota, hormones, immunity and disease susceptibility. Semin Immunopathol, 2019. 41 (2) 265–275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Kaliannan K, Robertson RC, Murphy K, Stanton C, Kang C, Wang B, Hao L, Bhan AK, and Kang JX, Estrogen-mediated gut microbiome alterations influence sexual dimorphism in metabolic syndrome in mice. Microbiome, 2018. 6 (1) 205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Fraher MH, O’Toole PW, and Quigley EM, Techniques used to characterize the gut microbiota: a guide for the clinician. Nat Rev Gastroenterol Hepatol, 2012. 9 (6) 312–322. [DOI] [PubMed] [Google Scholar]
  • 155.Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, and Gordon JI, An obesity-associated gut microbiome with increased capacity for energy harvest. Nature, 2006. 444 (7122) 1027–1031. [DOI] [PubMed] [Google Scholar]
  • 156.Smith MI, Yatsunenko T, Manary MJ, Trehan I, Mkakosya R, Cheng J, Kau AL, Rich SS, Concannon P, Mychaleckyj JC, Liu J, Houpt E, Li JV, Holmes E, Nicholson J, Knights D, Ursell LK, Knight R, and Gordon JI, Gut microbiomes of Malawian twin pairs discordant for kwashiorkor. Science, 2013. 339 (6119) 548–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Gustafsson B, Germ-free rearing of rats. Acta Anat (Basel), 1946. 2 (3–4) 376–391. [DOI] [PubMed] [Google Scholar]
  • 158.Singer-Englar T, Barlow G, and Mathur R, Obesity, diabetes, and the gut microbiome: an updated review. Expert Rev Gastroenterol Hepatol, 2019. 13 (1) 3–15. [DOI] [PubMed] [Google Scholar]
  • 159.Tang WH and Hazen SL, The Gut Microbiome and Its Role in Cardiovascular Diseases. Circulation, 2017. 135 (11) 1008–1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Elinav E, Garrett WS, Trinchieri G, and Wargo J, The cancer microbiome. Nat Rev Cancer, 2019. 19 (7) 371–376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Harach T, Marungruang N, Duthilleul N, Cheatham V, Mc Coy KD, Frisoni G, Neher JJ, Fak F, Jucker M, Lasser T, and Bolmont T, Reduction of Abeta amyloid pathology in APPPS1 transgenic mice in the absence of gut microbiota. Sci Rep, 2017. 7 41802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Kumar R, Eipers P, Little RB, Crowley M, Crossman DK, Lefkowitz EJ, and Morrow CD, Getting started with microbiome analysis: sample acquisition to bioinformatics. Curr Protoc Hum Genet, 2014. 82 18 18 11–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Human Microbiome Project C, A framework for human microbiome research. Nature, 2012. 486 (7402) 215–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Vogtmann E, Chen J, Kibriya MG, Chen Y, Islam T, Eunes M, Ahmed A, Naher J, Rahman A, Amir A, Shi J, Abnet CC, Nelson H, Knight R, Chia N, Ahsan H, and Sinha R, Comparison of Fecal Collection Methods for Microbiota Studies in Bangladesh. Appl Environ Microbiol, 2017. 83 (10). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Wasfy M, Oyofo B, Elgindy A, and Churilla A, Comparison of preservation media for storage of stool samples. J Clin Microbiol, 1995. 33 (8) 2176–2178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Nechvatal JM, Ram JL, Basson MD, Namprachan P, Niec SR, Badsha KZ, Matherly LH, Majumdar AP, and Kato I, Fecal collection, ambient preservation, and DNA extraction for PCR amplification of bacterial and human markers from human feces. J Microbiol Methods, 2008. 72 (2) 124–132. [DOI] [PubMed] [Google Scholar]
  • 167.Shaw AG, Sim K, Powell E, Cornwell E, Cramer T, McClure ZE, Li MS, and Kroll JS, Latitude in sample handling and storage for infant faecal microbiota studies: the elephant in the room? Microbiome, 2016. 4 (1) 40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Thomas V, Clark J, and Dore J, Fecal microbiota analysis: an overview of sample collection methods and sequencing strategies. Future Microbiol, 2015. 10 (9) 1485–1504. [DOI] [PubMed] [Google Scholar]
  • 169.Ericsson AC, Gagliardi J, Bouhan D, Spollen WG, Givan SA, and Franklin CL, The influence of caging, bedding, and diet on the composition of the microbiota in different regions of the mouse gut. Sci Rep, 2018. 8 (1) 4065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Sinha R, Abu-Ali G, Vogtmann E, Fodor AA, Ren B, Amir A, Schwager E, Crabtree J, Ma S, Microbiome C Quality Control Project, Abnet CC, Knight R, White O, and Huttenhower C, Assessment of variation in microbial community amplicon sequencing by the Microbiome Quality Control (MBQC) project consortium. Nat Biotechnol, 2017. 35 (11) 1077–1086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Costea PI, Zeller G, Sunagawa S, Pelletier E, Alberti A, Levenez F, Tramontano M, Driessen M, Hercog R, Jung FE, Kultima JR, Hayward MR, Coelho LP, Allen-Vercoe E, Bertrand L, Blaut M, Brown JRM, Carton T, Cools-Portier S, Daigneault M, Derrien M, Druesne A, de Vos WM, Finlay BB, Flint HJ, Guarner F, Hattori M, Heilig H, Luna RA, van Hylckama Vlieg J, Junick J, Klymiuk I, Langella P, Le Chatelier E, Mai V, Manichanh C, Martin JC, Mery C, Morita H, O’Toole PW, Orvain C, Patil KR, Penders J, Persson S, Pons N, Popova M, Salonen A, Saulnier D, Scott KP, Singh B, Slezak K, Veiga P, Versalovic J, Zhao L, Zoetendal EG, Ehrlich SD, Dore J, and Bork P, Towards standards for human fecal sample processing in metagenomic studies. Nat Biotechnol, 2017. 35 (11) 1069–1076. [DOI] [PubMed] [Google Scholar]
  • 172.Salter SJ, Cox MJ, Turek EM, Calus ST, Cookson WO, Moffatt MF, Turner P, Parkhill J, Loman NJ, and Walker AW, Reagent and laboratory contamination can critically impact sequence-based microbiome analyses. BMC Biol, 2014. 12 87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Amir A, McDonald D, Navas-Molina JA, Debelius J, Morton JT, Hyde E, Robbins-Pianka A, and Knight R, Correcting for Microbial Blooms in Fecal Samples during Room-Temperature Shipping. mSystems, 2017. 2 (2). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Fouhy F, Deane J, Rea MC, O’Sullivan O, Ross RP, O’Callaghan G, Plant BJ, and Stanton C, The effects of freezing on faecal microbiota as determined using MiSeq sequencing and culture-based investigations. PLoS One, 2015. 10 (3) e0119355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Panek M, Cipcic Paljetak H, Baresic A, Peric M, Matijasic M, Lojkic I, Vranesic Bender D, Krznaric Z, and Verbanac D, Methodology challenges in studying human gut microbiota - effects of collection, storage, DNA extraction and next generation sequencing technologies. Sci Rep, 2018. 8 (1) 5143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Team, N.I.H.H.M.P.A., A review of 10 years of human microbiome research activities at the US National Institutes of Health, Fiscal Years 2007–2016. Microbiome, 2019. 7 (1) 31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Zaneveld JR, Lozupone C, Gordon JI, and Knight R, Ribosomal RNA diversity predicts genome diversity in gut bacteria and their relatives. Nucleic Acids Res, 2010. 38 (12) 3869–3879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Okuda S, Tsuchiya Y, Kiriyama C, Itoh M, and Morisaki H, Virtual metagenome reconstruction from 16S rRNA gene sequences. Nat Commun, 2012. 3 1203. [DOI] [PubMed] [Google Scholar]
  • 179.Langille MG, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, Clemente JC, Burkepile DE, Vega Thurber RL, Knight R, Beiko RG, and Huttenhower C, Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotechnol, 2013. 31 (9) 814–821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Walker AW, Martin JC, Scott P, Parkhill J, Flint HJ, and Scott KP, 16S rRNA gene-based profiling of the human infant gut microbiota is strongly influenced by sample processing and PCR primer choice. Microbiome, 2015. 3 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Bonnet R, Suau A, Dore J, Gibson GR, and Collins MD, Differences in rDNA libraries of faecal bacteria derived from 10- and 25-cycle PCRs. Int J Syst Evol Microbiol, 2002. 52 (Pt 3) 757–763. [DOI] [PubMed] [Google Scholar]
  • 182.Walters WA, Caporaso JG, Lauber CL, Berg-Lyons D, Fierer N, and Knight R, PrimerProspector: de novo design and taxonomic analysis of barcoded polymerase chain reaction primers. Bioinformatics, 2011. 27 (8) 1159–1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Thompson LR, Sanders JG, McDonald D, Amir A, Ladau J, Locey KJ, Prill RJ, Tripathi A, Gibbons SM, Ackermann G, Navas-Molina JA, Janssen S, Kopylova E, Vazquez-Baeza Y, Gonzalez A, Morton JT, Mirarab S, Zech Xu Z, Jiang L, Haroon MF, Kanbar J, Zhu Q, Jin Song S, Kosciolek T, Bokulich NA, Lefler J, Brislawn CJ, Humphrey G, Owens SM, Hampton-Marcell J, Berg-Lyons D, McKenzie V, Fierer N, Fuhrman JA, Clauset A, Stevens RL, Shade A, Pollard KS, Goodwin KD, Jansson JK, Gilbert JA, Knight R, and C. Earth Microbiome Project, A communal catalogue reveals Earth’s multiscale microbial diversity. Nature, 2017. 551 (7681) 457–463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Janda JM and Abbott SL, 16S rRNA gene sequencing for bacterial identification in the diagnostic laboratory: pluses, perils, and pitfalls. J Clin Microbiol, 2007. 45 (9) 2761–2764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Nelson TA, Holmes S, Alekseyenko AV, Shenoy M, Desantis T, Wu CH, Andersen GL, Winston J, Sonnenburg J, Pasricha PJ, and Spormann A, PhyloChip microarray analysis reveals altered gastrointestinal microbial communities in a rat model of colonic hypersensitivity. Neurogastroenterol Motil, 2011. 23 (2) 169–177, e141–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Godoy-Vitorino F, Goldfarb KC, Brodie EL, Garcia-Amado MA, Michelangeli F, and Dominguez-Bello MG, Developmental microbial ecology of the crop of the folivorous hoatzin. ISME J, 2010. 4 (5) 611–620. [DOI] [PubMed] [Google Scholar]
  • 187.Eren AM, Maignien L, Sul WJ, Murphy LG, Grim SL, Morrison HG, and Sogin ML, Oligotyping: Differentiating between closely related microbial taxa using 16S rRNA gene data. Methods Ecol Evol, 2013. 4 (12). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Amir A, McDonald D, Navas-Molina JA, Kopylova E, Morton JT, Zech Xu Z, Kightley EP, Thompson LR, Hyde ER, Gonzalez A, and Knight R, Deblur Rapidly Resolves Single-Nucleotide Community Sequence Patterns. mSystems, 2017. 2 (2). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJ, and Holmes SP, DADA2: High-resolution sample inference from Illumina amplicon data. Nat Methods, 2016. 13 (7) 581–583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Asshauer KP, Wemheuer B, Daniel R, and Meinicke P, Tax4Fun: predicting functional profiles from metagenomic 16S rRNA data. Bioinformatics, 2015. 31 (17) 2882–2884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Jun SR, Robeson MS, Hauser LJ, Schadt CW, and Gorin AA, PanFP: pangenome-based functional profiles for microbial communities. BMC Res Notes, 2015. 8 479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Quast C, Pruesse E, Yilmaz P, Gerken J, Schweer T, Yarza P, Peplies J, and Glockner FO, The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res, 2013. 41 (Database issue) D590–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.McDonald D, Price MN, Goodrich J, Nawrocki EP, DeSantis TZ, Probst A, Andersen GL, Knight R, and Hugenholtz P, An improved Greengenes taxonomy with explicit ranks for ecological and evolutionary analyses of bacteria and archaea. ISME J, 2012. 6 (3) 610–618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Lesker TR, Durairaj AC, Galvez EJC, Lagkouvardos I, Baines JF, Clavel T, Sczyrba A, McHardy AC, and Strowig T, An Integrated Metagenome Catalog Reveals New Insights into the Murine Gut Microbiome. Cell Rep, 2020. 30 (9) 2909–2922 e2906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Balvociute M and Huson DH, SILVA, RDP, Greengenes, NCBI and OTT - how do these taxonomies compare? BMC Genomics, 2017. 18 (Suppl 2) 114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Scholz M, Ward DV, Pasolli E, Tolio T, Zolfo M, Asnicar F, Truong DT, Tett A, Morrow AL, and Segata N, Strain-level microbial epidemiology and population genomics from shotgun metagenomics. Nat Methods, 2016. 13 (5) 435–438. [DOI] [PubMed] [Google Scholar]
  • 197.Mukherjee S, Seshadri R, Varghese NJ, Eloe-Fadrosh EA, Meier-Kolthoff JP, Goker M, Coates RC, Hadjithomas M, Pavlopoulos GA, Paez-Espino D, Yoshikuni Y, Visel A, Whitman WB, Garrity GM, Eisen JA, Hugenholtz P, Pati A, Ivanova NN, Woyke T, Klenk HP, and Kyrpides NC, 1,003 reference genomes of bacterial and archaeal isolates expand coverage of the tree of life. Nat Biotechnol, 2017. 35 (7) 676–683. [DOI] [PubMed] [Google Scholar]
  • 198.Tringe SG, von Mering C, Kobayashi A, Salamov AA, Chen K, Chang HW, Podar M, Short JM, Mathur EJ, Detter JC, Bork P, Hugenholtz P, and Rubin EM, Comparative metagenomics of microbial communities. Science, 2005. 308 (5721) 554–557. [DOI] [PubMed] [Google Scholar]
  • 199.Tyson GW, Chapman J, Hugenholtz P, Allen EE, Ram RJ, Richardson PM, Solovyev VV, Rubin EM, Rokhsar DS, and Banfield JF, Community structure and metabolism through reconstruction of microbial genomes from the environment. Nature, 2004. 428 (6978) 37–43. [DOI] [PubMed] [Google Scholar]
  • 200.Quince C, Walker AW, Simpson JT, Loman NJ, and Segata N, Shotgun metagenomics, from sampling to analysis. Nat Biotechnol, 2017. 35 (9) 833–844. [DOI] [PubMed] [Google Scholar]
  • 201.Breitwieser FP, Lu J, and Salzberg SL, A review of methods and databases for metagenomic classification and assembly. Brief Bioinform, 2019. 20 (4) 1125–1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Rampelli S, Candela M, Turroni S, Biagi E, Collino S, Franceschi C, O’Toole PW, and Brigidi P, Functional metagenomic profiling of intestinal microbiome in extreme ageing. Aging (Albany NY), 2013. 5 (12) 902–912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Round JL and Mazmanian SK, The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol, 2009. 9 (5) 313–323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Guigoz Y, Dore J, and Schiffrin EJ, The inflammatory status of old age can be nurtured from the intestinal environment. Curr Opin Clin Nutr Metab Care, 2008. 11 (1) 13–20. [DOI] [PubMed] [Google Scholar]
  • 205.Pasolli E, Asnicar F, Manara S, Zolfo M, Karcher N, Armanini F, Beghini F, Manghi P, Tett A, Ghensi P, Collado MC, Rice BL, DuLong C, Morgan XC, Golden CD, Quince C, Huttenhower C, and Segata N, Extensive Unexplored Human Microbiome Diversity Revealed by Over 150,000 Genomes from Metagenomes Spanning Age, Geography, and Lifestyle. Cell, 2019. 176 (3) 649–662 e620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Emerson JB, Adams RI, Roman CMB, Brooks B, Coil DA, Dahlhausen K, Ganz HH, Hartmann EM, Hsu T, Justice NB, Paulino-Lima IG, Luongo JC, Lymperopoulou DS, Gomez-Silvan C, Rothschild-Mancinelli B, Balk M, Huttenhower C, Nocker A, Vaishampayan P, and Rothschild LJ, Schrodinger’s microbes: Tools for distinguishing the living from the dead in microbial ecosystems. Microbiome, 2017. 5 (1) 86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Bashiardes S, Zilberman-Schapira G, and Elinav E, Use of Metatranscriptomics in Microbiome Research. Bioinform Biol Insights, 2016. 10 19–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Shakya M, Lo CC, and Chain PSG, Advances and Challenges in Metatranscriptomic Analysis. Front Genet, 2019. 10 904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Abu-Ali GS, Mehta RS, Lloyd-Price J, Mallick H, Branck T, Ivey KL, Drew DA, DuLong C, Rimm E, Izard J, Chan AT, and Huttenhower C, Metatranscriptome of human faecal microbial communities in a cohort of adult men. Nat Microbiol, 2018. 3 (3) 356–366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Klassen JL, Defining microbiome function. Nat Microbiol, 2018. 3 (8) 864–869. [DOI] [PubMed] [Google Scholar]
  • 211.Grice EA, Kong HH, Renaud G, Young AC, Program NCS, Bouffard GG, Blakesley RW, Wolfsberg TG, Turner ML, and Segre JA, A diversity profile of the human skin microbiota. Genome Res, 2008. 18 (7) 1043–1050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Kim HJ, Kim JJ, Myeong NR, Kim T, Kim D, An S, Kim H, Park T, Jang SI, Yeon JH, Kwack I, and Sul WJ, Segregation of age-related skin microbiome characteristics by functionality. Sci Rep, 2019. 9 (1) 16748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Shibagaki N, Suda W, Clavaud C, Bastien P, Takayasu L, Iioka E, Kurokawa R, Yamashita N, Hattori Y, Shindo C, Breton L, and Hattori M, Aging-related changes in the diversity of women’s skin microbiomes associated with oral bacteria. Sci Rep, 2017. 7 (1) 10567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Gao L, Xu T, Huang G, Jiang S, Gu Y, and Chen F, Oral microbiomes: more and more importance in oral cavity and whole body. Protein Cell, 2018. 9 (5) 488–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Segata N, Haake SK, Mannon P, Lemon KP, Waldron L, Gevers D, Huttenhower C, and Izard J, Composition of the adult digestive tract bacterial microbiome based on seven mouth surfaces, tonsils, throat and stool samples. Genome Biol, 2012. 13 (6) R42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Galkin F, Mamoshina P, Aliper A, Putin E, Moskalev V, Gladyshev VN, and Zhavoronkov A, Human Gut Microbiome Aging Clock Based on Taxonomic Profiling and Deep Learning. iScience, 2020. 23 (6) 101199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Mackowiak PA, Recycling metchnikoff: probiotics, the intestinal microbiome and the quest for long life. Front Public Health, 2013. 1 52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Suzuki Y, Ikeda K, Sakuma K, Kawai S, Sawaki K, Asahara T, Takahashi T, Tsuji H, Nomoto K, Nagpal R, Wang C, Nagata S, and Yamashiro Y, Association between Yogurt Consumption and Intestinal Microbiota in Healthy Young Adults Differs by Host Gender. Front Microbiol, 2017. 8 847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Wiesenborn DS, Galvez EJC, Spinel L, Victoria B, Allen B, Schneider A, Gesing A, Al-Regaiey KA, Strowig T, Schafer KH, and Masternak MM, The Role of Ames Dwarfism and Calorie Restriction on Gut Microbiota. J Gerontol A Biol Sci Med Sci, 2020. 75 (7) e1–e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Fraumene C, Manghina V, Cadoni E, Marongiu F, Abbondio M, Serra M, Palomba A, Tanca A, Laconi E, and Uzzau S, Caloric restriction promotes rapid expansion and long-lasting increase of Lactobacillus in the rat fecal microbiota. Gut Microbes, 2018. 9 (2) 104–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Tanca A, Abbondio M, Palomba A, Fraumene C, Marongiu F, Serra M, Pagnozzi D, Laconi E, and Uzzau S, Caloric restriction promotes functional changes involving short-chain fatty acid biosynthesis in the rat gut microbiota. Sci Rep, 2018. 8 (1) 14778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Cabreiro F, Au C, Leung KY, Vergara-Irigaray N, Cocheme HM, Noori T, Weinkove D, Schuster E, Greene ND, and Gems D, Metformin retards aging in C. elegans by altering microbial folate and methionine metabolism. Cell, 2013. 153 (1) 228–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Zhernakova A, Kurilshikov A, Bonder MJ, Tigchelaar EF, Schirmer M, Vatanen T, Mujagic Z, Vila AV, Falony G, Vieira-Silva S, Wang J, Imhann F, Brandsma E, Jankipersadsing SA, Joossens M, Cenit MC, Deelen P, Swertz MA, s. LifeLines cohort, Weersma RK, Feskens EJ, Netea MG, Gevers D, Jonkers D, Franke L, Aulchenko YS, Huttenhower C, Raes J, Hofker MH, Xavier RJ, Wijmenga C, and Fu J , Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science, 2016. 352 (6285) 565–569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Forslund K, Hildebrand F, Nielsen T, Falony G, Le Chatelier E, Sunagawa S, Prifti E, Vieira-Silva S, Gudmundsdottir V, Pedersen HK, Arumugam M, Kristiansen K, Voigt AY, Vestergaard H, Hercog R, Costea PI, Kultima JR, Li J, Jorgensen T, Levenez F, Dore J, Meta H.I.T.c, Nielsen HB, Brunak S, Raes J, Hansen T, Wang J, Ehrlich SD, Bork P, and Pedersen O, Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature, 2015. 528 (7581) 262–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Bryrup T, Thomsen CW, Kern T, Allin KH, Brandslund I, Jorgensen NR, Vestergaard H, Hansen T, Hansen TH, Pedersen O, and Nielsen T, Metformin-induced changes of the gut microbiota in healthy young men: results of a non-blinded, one-armed intervention study. Diabetologia, 2019. 62 (6) 1024–1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Lee H, Lee Y, Kim J, An J, Lee S, Kong H, Song Y, Lee CK, and Kim K, Modulation of the gut microbiota by metformin improves metabolic profiles in aged obese mice. Gut Microbes, 2018. 9 (2) 155–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Bitto A, Ito TK, Pineda VV, LeTexier NJ, Huang HZ, Sutlief E, Tung H, Vizzini N, Chen B, and Smith K, Transient rapamycin treatment can increase lifespan and healthspan in middle-aged mice. elife, 2016. 5 e16351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Hurez V, Dao V, Liu A, Pandeswara S, Gelfond J, Sun L, Bergman M, Orihuela CJ, Galvan V, Padron A, Drerup J, Liu Y, Hasty P, Sharp ZD, and Curiel TJ, Chronic mTOR inhibition in mice with rapamycin alters T, B, myeloid, and innate lymphoid cells and gut flora and prolongs life of immune-deficient mice. Aging Cell, 2015. 14 (6) 945–956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Jung MJ, Lee J, Shin NR, Kim MS, Hyun DW, Yun JH, Kim PS, Whon TW, and Bae JW, Chronic Repression of mTOR Complex 2 Induces Changes in the Gut Microbiota of Diet-induced Obese Mice. Sci Rep, 2016. 6 30887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Ghosh TS, Rampelli S, Jeffery IB, Santoro A, Neto M, Capri M, Giampieri E, Jennings A, Candela M, Turroni S, Zoetendal EG, Hermes GDA, Elodie C, Meunier N, Brugere CM, Pujos-Guillot E, Berendsen AM, De Groot L, Feskins EJM, Kaluza J, Pietruszka B, Bielak MJ, Comte B, Maijo-Ferre M, Nicoletti C, De Vos WM, Fairweather-Tait S, Cassidy A, Brigidi P, Franceschi C, and O’Toole PW, Mediterranean diet intervention alters the gut microbiome in older people reducing frailty and improving health status: the NU-AGE 1-year dietary intervention across five European countries. Gut, 2020. 69 (7) 1218–1228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Smith BJ, Miller RA, Ericsson AC, Harrison DC, Strong R, and Schmidt TM, Changes in the gut microbiome and fermentation products concurrent with enhanced longevity in acarbose-treated mice. BMC Microbiol, 2019. 19 (1) 130. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES