Review - (2020) Volume 14, Issue 1
Role of Intestinal Bacteria and their Mechanisms on Host Health and Aging at
the Genetic Level
Song S1,
Slone J2 and
Huang T2*
*Correspondence:
Huang T, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229,
USA,
Tel: +5138039260,
Email:
1Human Aging Research Institute, Nanchang University, Nanchang 330031, P.R. China
2Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
Received: 05-Feb-2020
Published:
24-Feb-2020
Citation: Song S, Slone J, Huang T (2020) Role of Intestinal Bacteria and their
Mechanisms on Host Health and Aging at the Genetic Level. J Mol Genet Med
14: 441
Copyright: © 2020 Song S, et al. This is an open-access article distributed under
the terms of the Creative Commons Attribution License, which permits unrestricted
use, distribution, and reproduction in any medium, provided the original author and
source are credited
Abstract
Researcher have paid more and more attention to the mechanisms by which intestinal flora affect the health and
wellbeing of the host, and to what extent intestinal flora contribute to illness in the general population. Researchers
also intend to determine the changes of intestinal flora over time, and the consequences of those changes for aging.
This review will focus on the role that intestinal bacteria play in health and aging by providing a landscape of the
intestinal flora with age, as well as the clinical signs/symptoms and mechanisms by which intestinal flora may affect
aging. We anticipate that by exploring the relationship between intestinal flora and the host, it may ultimately be
possible to identify particular classes of intestinal flora that affect aging.
Keywords
Gut bacteria; Aging; Probiotics
Introduction
There are a great variety of microbial groups that coexist within
the human body. The colon has about 1014 bacteria, far greater than
the number present on the skin 1012 [1]. Every individual has a distinct
gut bacteria composition, which is subject to change with age, with
the highest differences occurring as we get older [2,3]. Gut microbials
play a very important role in human health; for example, some of their
more important activities include helping to break down hard-todigest
proteins and fibers, and producing short-chain fatty acids such
as acetic acid, propionic acid and butyric acid [4,5]. These short-chain
fatty acids can have a significant effect on the T cells of the host, alter
the regulation of inflammation in the intestine, and can even have an
impact on the host's metabolism [6-9]. All of these suggest that our
daily diet and/or pharmaceutical usage will affect the flora in the gut,
which will in turn affect our health [10]. The gut microbiome plays
an important part in the gut environment and influences the host's
physiology and pathology [11]. The gut microbiome also affects our
endocrine performance directly or indirectly through their metabolites
[12]. The gut microbiome can also influence our neurological functions
via the so-called “gut-brain axis” or “liver-gut axis” [13,14]. The gut
flora have a core microbiome “enterotype” that is different for each
individual [15]. Researchers have identified three enterotypes based on
a variation of Bacteroides (enterotype 1), Prevotella (enterotype 2) and
Ruminococcus (enterotype 3) [16]. When we age, the abundance of core
microbes has a tendency to occur [17]. This change can be contributed
by many factors, such changes in the composition of food [18,19].
The nature of the broader society can also have a significant effect, as
the composition of gut microbes are different in industrial and nonindustrial
countries due to environment and diet [20,21].
Literature Review
The study of intestinal flora began with the study of the rDNAs of
intestinal bacteria; Subsequently, 16S rDNA that was PCR amplified
from human feces was used as a reference [22-24]. Since many
microbiomes have distinctive functions, understanding the precise
composition of the intestinal microbiome may yield valuable insights
as to the health status of an individual patient [25]. Thus, improvements
in bioinformatics technology and high-throughput sequencing
technology may allow us to process large quantities of data, helping
to advance the field in a rapid and meaningful way (Figure 1) [26,27].
Alterations in the intestinal flora with age
Recently, some groups have analyzed the relationship between
intestinal bacteria and aging, even to the point of being able to predict
age from microbiome composition [28]. The diversity of the gut flora
appears to be negatively correlated with age, raising the possibility
that the maintenance or improvement of gut flora diversity maybe
help counteract or slow the aging process [29]. It is also important to
note that microbiota diversity appears to be associated only with the
biological age of an individual, and not with their chronological age [30].
Newborns
Previous studies have shown that the unique microbes present in
the placenta suspected to be the first microbes to colonize the intestinal
flora during fetal development, and at least partially associated with the
adverse reactions of pregnancy or premature birth [31,32]. However,
it has also been shown that the microbial population in the placenta is
very small, and that even more of the adverse reactions of pregnancy
could result from vaginal microorganisms [33,34]. This putative effect
of vaginal microbes on the reactions of pregnancy and preterm birth
has been verified in multiple publications [35,36]. The delivery mode in
particular appears to have a significant effect on the gut flora; vaginallydelivered
children are more likely to have the mother’s vaginal
microbiota, while infants born by C-section are more likely to be
colonized by Lactobacillus, Bifidobacterium, and Bacteroides [37]. This
can have impacts on the incidence of childhood obesity, inflammatory
bowel disease, food allergies, asthma, and diabetes mellitus [38- 40]. The gut flora can also be affected by many other factors such as
breastfeeding. Breastfeeding can compensate for the colonization of B. bifidum and L. gasseri in C-section infants, resulting in flora more similar to those observed in infants who underwent vaginal delivery
[41,42]. Breast milk can increase Bifidobacterium and help promote the
health of the baby. As children grow, the consumption of breast milk
in the diet decreases, and the abundance of Bifidobacterium decreases
[43-46]. Through careful monitoring of the growth environment, food,
and use of antibiotics, parents can also have a significant impact on
their babies’ intestinal flora [47].
School children
The bacteria that initially colonize a newborn are determined by
the delivery mode. Subsequently, the environment dramatically alters
that newborn’s intestinal flora. For example, it has been demonstrated
that a baby’s intestinal bacteria moves closer to those observed in its
cohabiting family members [48]. Treatment with antibiotics can also
significantly reduce the abundance of intestinal flora [49]. Breastfeeding
is perhaps the biggest factor affecting the intestinal flora of a newborn [50,51]. However, introducing solid food produces a dramatic
switch of the microflora. One of the most critical changes involves
Bifidobacteria and Lactobacillus, as these two genera of bacteria are
known to be lower in adults compared to younger individuals [52].
Bifidobacteria are the most abundant gut flora in a breast milk-fed
infant, and formula milk and dairy milk can change the abundance
of the Bifidobacteria in the first 3 months after birth [53,54]. The
introduction of solid foods migrates the baby's gut flora to a more adult
composition [55,56]. The flora obtained from the mother gradually
shifts to the acquired flora as the child gets older. Dietary structure is
one of the most important factors. Malnourished children tend to have
abnormal microbial composition, which may lead to increased risk of
various diseases or even death [57].
Teenagers
Puberty represents a period of transition to independence, during which major lifestyle changes take place; it is a critical period for future
adult behavior. The differences in life span between males and females
is well-studied [58,59]. The male sex hormone testosterone has been
shown to correlate with increased pro-inflammatory cytokine release,
while estrogens can reduce mitochondrial free-radical production
[60]. In mice, the gut microbiota begins diverging between males and
females in puberty [61]. Sex hormones potentially impact on the gut
microbiome and its diversification in a gender-specific manner [61- 64]. Recent research has shown that the timing of androgen exposure
can affect a mouse’s gut microbiome [65]. Since microbial exposures,
sex hormones or both could have an influence on the age of human
beings, the question is whether hormonal changes in puberty initiate
gut microbiome shifts, or if changes in the microbiome influence
sexual maturation and growth.
Adolescence is a pivotal period for brain maturation [66]. The
lifestyle and physiological changes that occur during this period can
be stressful, and may cause long-lasting detrimental changes [67-70].
Intestinal microbes susceptible to environmental influence may affect
the development of the nervous system through the Microbiota-Gut-
Brain Axis [71]. The details of the Microbiota-Gut-Brain Axis and
mental illness will be further discussed below.
Adults
As human intestinal flora continues to be exposed to diet, antibiotics
and other environmental factors, the composition of flora continues to
evolve and become more and more complex, until it reaches its peak
in adulthood [72-74]. In general, children have a higher number of
enterobacteria than adults [52]. On the other hand, the intestinal flora
of adults is very stable, but can be affected by many factors, including
diet, antibiotics, geographical location, and seasonal/temperature
conditions [75-83]. Even oxygen availability in high altitudes can affect
the adult intestinal flora [84].
Interestingly, pregnancy can reset gut flora in adulthood [61,73]. In
the early stages of pregnancy, some unique microbes can be detected in
women who subsequently have preterm delivery [85]. Proteobacteria and Actinobacteria have also been shown to be increased in the third
trimester of pregnancy relative to the first [86]. This shift is very similar
to what is observed in obese populations [87]. Bacteroides-Prevotella group bacteria are also higher in overweight mothers [88]. The dietary
consumption of pregnant women, particularly as it relates to probiotics,
can have a significant impact on their blood glucose regulation [89]. Sex
hormones can also affect pregnant women’s emotional state through
the brain-gut axis [90].
Aged people
The intestinal flora of the elderly has been widely studied in an
attempt to identify any correlations between aging and intestinal flora
[91-94]. Thus far, there has been no clear conclusion on this topic [95].
It appears that there is little difference in the gut flora between young
adults and the elderly, while there is a much greater difference in the
gut flora of the elderly when compared with centenarians [96]. With
advancing age, the elderly suffer a variety of increasingly serious and
complex diseases, and the gut microbiota in the elderly is a reflection of
this reality, showing an extreme variability in composition not usually
observed in younger individuals [3]. Long-term hospitalized patients
have a higher Bacteroidetes and Firmicutes composition (and thus, less
microbial complexity) than other individuals, and higher microbial
complexity is associated with resistance to vulnerability in the elderly
[97,98]. Bifidobacterium and Lactobacillus in core microbiota was found to be quite different in the elderly as compared with younger
adults [99,100]. The loss of Lactobacillus and Faecalibacterium and
the increased abundance of the Oscillibacter and Alistipes genera
and Eubacteriaceae family were found in older populations, while
the ratio of Firmicutes/Bacteroidetes first rises and then falls with age
[101]. The genus Alistipes is also overrepresented in aged mice [102].
The proportion of the families Ruminococcaceae, Lachnospiraceae,
and Bacteroidaceae decreases with age, and the aged population also
has a decreased presence of the beneficial bacteria of A. muciniphila,
F. prausnitzii, lactobacilli, and bifidobacterial, along with increased
presences of Clostridia, enterobacteria, Streptococci, Staphylococci,
yeast and facultative anaerobes in gut microbiota [103,104]. This
changed composition of flora may be largely associated with diet [105].
It is possible that gut microbiota interventions may extend
lifespan and improve age-related diseases [106]. In traditional Chinese
medicine (TCM), fecal microbiota transplantation was used some
Chinese medicine to treat diseases, including Gegen Qinlian Decoction
(GQD), Rhizoma Coptis, and yellow soup [107-110]. It has also been
reported that fecal microbiota transplantation can extend the lifespan
of mice and fish [111,112]. Have study showed that fecal microbiota
transplantation can affects expression of host genes associated with
the TOR-pathway (DEPTOR) and with cell adhesion and extracellular
matrix composition (DSCAM) [113]. Other studies find that fecal
microbiota transplantation prolongs life is considered to be associated
with the suppression of chronic low-grade inflammation resulting
from the intestinal luminal environment and tissue or produce shortchain
fatty acids (SCFAs) [114-116]. In addition, dietary management
to control the composition of intestinal flora could be beneficial for
counteracting the aging process [117,118].
The variation of microbiota in different age groups
Bifidobacterium, Eubacterium, Bacteroides and Lactobacillus are
higher in younger individuals than in older individuals. In contrast,
Ruminococcus is more common in older individuals. Bacteroides,
enterococci, enterobacteria and clostridia do not change at all during
aging [119,120].
The others factors influencing flora
The intestinal flora can change for a variety of reasons [15]. Longterm
changes in diet and the use of drugs can affect the flora [10,99,121- 126]. On the dietary side, fructo-oligosaccharides (FOS), vitamins,
creatine, bile acids, NO and colanic acid (CA) are among some of these
factors [102,127-131]. On the pharmaceutical side, more than 75% of
people age 65 or older take at least one prescription medication, which
may affect the intestinal flora and even lifespan [119,122,132]. Several
medicines showed attenuating or reversal effects on the aging process
in animal models, and may also extend lifespan in humans [133].
Rapamycin, metformin, acarbose and NAD precursors have been
particularly well-studied [134-138]. Rapamycin has long been known
as a modulator of extended aging in yeast, and affected the life of the
animal model such as Drosophila and mice with a way that is not fully
understood [139-145]. Rapamycin plays a mechanistic role in longevity
in mammals by targeting mTOR and regulating important cellular
processes [146]. Short-term rapamycin treatment can improve health
in mice, and down-regulation of the TOR pathway with RNAi supports
the roles of nutrients and the mTOR pathway in lifespan [147,148].
Interestingly, the mTOR pathway has greater impact on females than
males [149-151]. In animals, short-term rapamycin treatment can
improve learning and memory as observed in the case of a low-calorie
diet or caloric restriction (CR) [152-154]. T. Lactobacillus plantarum can affect lifespan in Drosophila through a mechanism whereby lactic
acid triggers ROS via the intestinal NADPH oxidase Nox [155]. SKN-
1/Nrf and DAF-16/FoxO-mediated transcription regulates protective
genes when TORC1 is inhibited with rapamycin [143]. There is also
a feedback loop in the TOR regulatory pathway, as SKN-1 increases
the transcription of TORC1 pathway genes when TORC1 is inhibited,
and the IIS and TOR pathways each influence aging by regulating
SKN-1 and DAF-16 [143]. In addition, the most common used
medicine in elder include acetylcholinesterase inhibitors (AchEIs),
multivitamins, HMG-CoA reductase inhibitors (statins), analgesics
(NSAIDs, weak and strong opioids), antihypertensives, antiplatelet
agents (aspirin) and metformin [156-163]. In the above drugs,
metformin can indirectly regulate the lifespan of the host by affecting
the bacteria [164]. Metformin also extends lifespan through mTOR
signaling, and it has been shown that the mucin-degrading species Akkermansia muciniphila is more abundant in mice which have been
metformin treated [165]. The mechanism by which metformin inhibits
mTORC1 is by activating AMP-activated protein kinase (AMPK) or
the Ragulator complex (Rag GTPase), or by REDD1 upregulation
[166,167]. Previous studies have speculated that metformin affects
host longevity by changing the production of secondary metabolites,
and by affecting level of folate metabolism in the host intestinal flora.
However, there is no evidence to support that the lifespan of the host
is affected when the level of folate in the host is changed [15,144,168].
In a recent study, metformin was found to extend the host lifespan by
affecting host lipid metabolism with metformin-bacterial interactions,
and there is another research finding that Bacterial colanic acid (CA) is
also known to promote longevity in C. elegans [169,170].
Intestinal bacteria and aging
Bifidobacteria has been recognized as a probiotic that plays an
important role in human development [99]. Similarly, changes in
gut flora composition can also affect human aging. For example, agerelated
changes in the gut microbiome have implicated in decreases
in immunity, gastrointestinal dysfunction, increased risk of infectious
diseases, increased risk of cardiovascular diseases, liver diseases,
alopecia, decreased ability to exercise and even Alzheimer's disease
[94,171-178].
Obesity and gut flora
Obesity is a common and increasingly severe problem across the
world [179]. Inappropriate diet may adversely impact our health by
effecting microbial composition, with reverberating effects throughout
the gut-liver-adipose tissue axis and intestine-islet axis [180,181].
Obese individuals are known to have specific microbial populations
within their faeces, and even gastric bypass surgery (Roux-en-Y gastric
bypass, or RYGB) can affect the composition of the gut microbiota
[182,183]. RYGB is a treatment option for morbid obesity, and after
surgical treatment with RYGB, obese individuals will undergo unique
changes in their intestinal microbial community lead to further
improvements in long-term weight loss, improved metabolic status
and extended lifespan [184-186]. Fecal transplantation could also be
a promising approach for treating the morbidly obese [187,188]. The
species present in the gut microbiota can be affected by many factors,
such as energy balance, delivery mode, antibiotics, diet, neural signals,
genetics, health status, environment, lifestyle and endocrine factors
[189-195]. Gut microbiota can influence adipose tissue metabolism
through gut peptides (e.g. glucagon-like peptide-1 and -2) in the ECB
system or by changing the short-chain fatty acids to affect metabolic
diseases [196]. In mice, there is a well-studied interrelation between diet and the species composition of the gut microbial community [188].
For instance, Methanobrevibacter and Bacteroides thetaiotaomicron have been shown to lead to weight gain in gnotobiotic mice, and levels
of Bacteroidetes and Firmicutes are significantly different between
lean and obese mice [197-199]. Most intriguingly, Akkermansia
muciniphila-induced obesity in mice can be cured by treatment with
the prebiotic oligofructose, raising the possibility that similar dietary
interventions may help reverse obesity in humans as well [200].
In humans, a number of studies have compared the gut flora of
obese and normal-weight people, resulting in a number of insights and
correlations [189]. For instance, Lactobacillus and Bifidobacterium are
known to affect obesity or lean status in the gut [201]. Additionally, B.
eggerthii is observed at higher levels in obese schoolchildren, and the
abundance of Enterobacteriaceae is also higher in overweight children
[202,203]. The Firmicutes/Bacteroidetes ratio may also be associated
with degree of the obesity in schoolchildren [204]. In contrast, Akkermansia muciniphila-like bacteria and Desulfovibrio are more
abundant in normally-weighted children [205].
Athletic ability and gut flora
As we age, loss of skeletal muscle mass and the probability of
sarcopenia increase [177,206,207]. Exercise also plays a beneficial role
in neurocognition by protecting the damage of hippocampal nerves
through microbial flora [66,208]. Many studies have analyzed the gut
flora of athletes. These efforts have shown that Low Carbohydrate High
Fat (LCHF) diet can decrease the relative abundance of Faecalibacterium spp. and increase Bacteroides and Dorea spp. in the stool microbiota of
race walkers [209]. A greater abundance of Akkermansia and Prevotella was also found in the gut microbiomes of sportsmen; the latter genus is
particularly interesting, being mucin-degrading bacteria that can affect
the biosynthesis of branched-chain amino acid (BCAA) [210,211]. On
the other hand, the genus Veillonella utilizes exercise-induced lactate as
its sole carbon source to make propionate, and fecal metabolites such as
short-chain fatty acids (SCFAs), acetate, propionate, and butyrate are
known to be increased in athletes as compared to sedentary individuals
[212,213]. Veillonella metabolizes lactic acid through Methylmalonyl-
CoA-related gene expression during exercise [212,214]. The shortchain
fatty acids (SCFAs) converted from metabolic lactic acid by Veillonella can increase athletic ability by optimizing lipid oxidation,
heart rate and maximum oxygen consumption [215-217].
Inflammation and intestinal flora
Gut dysbiosis is closely related to inflammation in the intestinal
tract [172]. The immune dysfunctions seen in senescence-associated
phenotype are associated with increased ROS [218,219]. The microbial
changes associated with aging have a broad correlation with the
occurrence of intestinal inflammatory disorders; in particular, the
proportions of Proteobacteria and Firmicutes appear to be positively
correlated with such disorders [220]. The number of anaerobic bacteria
is also increased in the gut of the elderly [221]. Ruminococcus gnavus is increased in patients with Crohn's disease (CD), while the levels of Dialister invisus, Faecalibacterium prausnitzii and Bifidobacterium
adolescentis are decreased [222,223]. Increased Actinobacteria and Proteobacteria and decreased Bacteroidetes and Lachnospiraceae are associated with inflammatory bowel disease (IBD) [224,225]. Akkermansia, TM7 bacteria, and Proteobacteria cause inflammation,
as shown in the case of microbiota transferred from aged mice into
young GF mice [226]. In particular, Akkermansia muciniphila has
been found to have many roles in human health and disease [201,227- 229]. Diet is very important for patients with inflammatory bowel disease, particularly as it relates to probiotic intake [230]. Probiotics
in the gut flora protect the host through the immune response system
in the intestinal epithelial cells and help develop biofilms in the gut
(such as Bacillus subtilis) [175,231-233]. All of these factors may also be
affected by sex hormones [234].
The influence of gut flora on the brain
The gut flora is associated with the mental health of the host [71].
Therefore, the influence of the intestinal flora on the host nervous
system has been widely studied, with some intriguing insights. For
instance, it has been demonstrated that the microbes in the human gut
can affect the nerves system by production neurochemicals [235,236].
Probiotics have also been shown to affect neurotransmitter levels and
chronic inflammation that are associated neurodegenerative disease
[237]. The change in gut motility, acidity and neurochemicals can
also alter the microbial composition of the gut flora [182]. Together,
this reciprocal interaction between the gut flora and the brain is
referred to as the “Gut Microbiota-brain Axis”, and it is clear that
disruptions of the microbial composition in the human gut can
result in neurological disease by this axis [238-242]. For instance, the
microbial composition of patients with depression has been shown
to have a higher abundance of Enterobacteriaceae and Alistipes and a
lower abundance of Faecalibacterium than observed in individuals not
diagnosed with depression, and Faecalibacterium has been shown to
protect against depression via microbiota transplantation [243]. Gut
microbes have also been implicated in PD progression by producing
SCFA to act on the enteroendocrine cells (EECs) [244]. Parkinson's
disease in the elderly is often affected by the gut microbiota-brain axis,
and intestinal infection can also trigger the occurrence of Parkinson's
disease [245,246]. Together, these results would seem to suggest that
fecal microbiota transplantation may provide treatment (or at least
some level of preventative power) for many forms of neurological
diseases.
Conclusion
Here, we provide a brief overview of the effects of gut microbiota
on host health and aging. It seems clear that intestinal flora play an
important role in human aging and age-related diseases. Of course,
the intestinal flora is affected by the host's diet, antibiotic use, exercise,
the external environment and the effects of the host's immune system.
There are many ways to improve the health and healthy aging of the
host by manipulating gut flora. The development of such treatments
may provide a major opportunity for improving human health.
References
- Sender R, Fuchs S, Milo R (2016) Revised estimates for the number of human and bacteria cells in the body. PLoS Biol 14: e1002533.
- OToole PW, Jeffery IB (2015) Gut microbiota and aging. Science 350: 1214- 1215.
- Claesson MJ (2011) Composition, variability, and temporal stability of the intestinal microbiota of the elderly. Proc Natl Acad Sci USA 108: 4586-4591.
- Wong JMW, Souza R, Kendall CWC, Emam A, Jenkins DJA (2006) Colonic health: Fermentation and short chain fatty acids. 40: 235-243.
- Louis P, Flint HJ (2017) Formation of propionate and butyrate by the human colonic microbiota. Environ Microbiol 19: 29-41.
- Smith PM (2013) The microbial metabolites, short-chain fatty acids, regulate colonic treg cell homeostasis. Science 341: 569-73.
- Tedelind S, Westberg F, Kjerrulf M, Vidal A (2007) Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: A study with relevance to inflammatory bowel disease. World J Gastroenterol 13: 2826-2832.
- Vinolo MAR, Rodrigues HG, Nachbar RTN, Curi R (2011) Regulation of inflammation by short chain fatty acids. 3: 858-876.
- Vinolo MAR, Rodrigues HG, Nachbar RTN, Curi R (2011) Suppressive effect of short-chain fatty acids on production of proinflammatory mediators by neutrophils. 22: 849-855.
- Claesson MJ (2012) Gut microbiota composition correlates with diet and health in the elderly. Nature 488: 178-184.
- Guarner F, Malagelada JR (2003) Gut flora in health and disease. Lancet 361:
- Clarke G, Stilling RM, Kennedy PJ, Stanton C, Cryan JF, et al. (2014) Gut microbiota: The neglected endocrine organ. 28: 1221-1238.
- Grenham S (2011) Brain-gut-microbe communication in health and disease. Front Physiol 2: 94.
- Minemura M, Shimizu Y (2015) Gut microbiota and liver diseases. World J Gastroenterol 21: 1691-1702.
- Kim S, Jazwinski SM (2018) The gut microbiota and healthy aging: A mini- minireview. Gerontol 64: 513-520.
- Anne M (2011) Enterotypes of the human gut microbiome. 473: 174-180.
- Biagi E, Franceschi C, Rampelli S, Severgnini M, Ostan R, et al. (2016) Gut microbiota and extreme longevity. 26: 1480-1485.
- Costea PI, Hildebrand F, Arumugam M, Bäckhed F, Blaser MJ (2018) Enterotypes in the landscape of gut microbial community composition. 3: 8-16.
- Wu GD (2011) Linking long-term dietary patterns with gut microbial enterotypes. Science 334: 105-108.
- Segata N (2015) Gut Microbiome: Westernization and the disappearance of intestinal diversity. Curr Biol 25: R611-613.
- Martinez I (2015) The gut microbiota of rural papua new guineans: Composition, diversity patterns, and ecological processes. Cell Rep 11: 527-538.
- Wilson KH, Blitchington RB (1996) Human colonic biota studied by ribosomal DNA sequence analysis. Appl Environ Microbiol 62: 2273-2278.
- Suau A (1999) Direct analysis of genes encoding 16S rRNA from complex communities reveals many novel molecular species within the human gut. Appl Environ Microbiol 65: 4799-807.
- Yang YW (2015) Use of 16S rRNA Gene-targeted group-specific primers for real-time pcr analysis of predominant bacteria in mouse feces. Appl Environ Microbiol 81: 6749-6756.
- Gill SR (2006) Metagenomic analysis of the human distal gut microbiome. Science 312: 1355-1359.
- Qin J (2010) A human gut microbial gene catalogue established by metagenomic sequencing. Nature 464: 59-65.
- Caporaso JG (2010) QIIME allows analysis of high-throughput community sequencing data. Nat Methods 7: 335-336.
- Galkin F (2018) Human microbiome aging clocks based on deep learning and tandem of permutation feature importance and accumulated local effects. Cold Spring Harbor Lab 2: 2-6
- Jackson MA (2016) Signatures of early frailty in the gut microbiota. Genome Med 8: 8.
- Maffei VJ (2017) Biological aging and the human gut microbiota. Genome Med 72: 1474-1482.
- Collado MC (2016) Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid. Genome Med 6: 23129.
- Antony KM (2015) The pre-term placental microbiome varies in association with excess maternal gestational weight gain. Genome Med 212: 653.e1-16.
- Goffau MC (2019) Human placenta has no microbiome but can contain potential pathogens. Genome Med 572: 329-334.
- Aagaard K (2014) The placenta harbors a unique microbiome. Genome Med 6: 237ra65.
- Serrano MG (2019) Racioethnic diversity in the dynamics of the vaginal microbiome during pregnancy. Nat Med 25: 1001-1011.
- Fettweis JM (2019) The vaginal microbiome and preterm birth. Nat Med 25: 1012-1021.
- Dominguez-Bello MG (2010) Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci USA 107: 11971-11975.
- Cardwell CR (2008) Caesarean section is associated with an increased risk of childhood-onset type 1 diabetes mellitus: A meta-analysis of observational studies. Nat Med 51: 726-735.
- Thavagnanam S (2008) A meta-analysis of the association between caesarean section and childhood asthma. Nat Med 38: 629-633.
- Lim ES (2015) Early life dynamics of the human gut virome and bacterial microbiome in infants. Nat Med 21: 1228-1234.
- Martin R (2016) Early-life events, including mode of delivery and type of feeding, siblings and gender, shape the developing gut microbiota. Nat Med 11: e0158498.
- Shigeno Y (2018) Gut microbiota development in mice is affected by hydrogen peroxide produced from amino acid metabolism during lactation. FASEB J 1: 2-4.
- Milani C (2017) The first microbial colonizers of the human gut: Composition, activities, and health implications of the infant gut microbiota. Genome Med 81: 2-4.
- Munyaka PM, Khafipour E (2014) External influence of early childhood establishment of gut microbiota and subsequent health implications. Front Pediatrics 2: 109.
- Stewart CJ (2018) Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature 562: 583-588.
- Bäckhed F (2015) Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe 17: 690-703.
- Hanski I (2012) Environmental biodiversity, human microbiota, and allergy are interrelated. Proc Nat Acad Sci 109: 8334-8339.
- Yatsunenko T (2012) Human gut microbiome viewed across age and geography. Nature 486: 222-227.
- Koenig JE (2011) Succession of microbial consortia in the developing infant gut microbiome. Proc Natl Acad Sci USA 108: 4578-4585.
- Vaishampayan PA (2010) Comparative metagenomics and population dynamics of the gut microbiota in mother and infant. Genome Biol Evol 2: 53-66.
- Martin R (2007) Cultivation-independent assessment of the bacterial diversity of breast milk among healthy women. Res Microbiol 158: 31-37.
- Hopkins MJ (2001) Gut, age and disease related changes in intestinal bacterial populations assessed by cell culture, 16S rRNA abundance, and community cellular fatty acid profiles. Gut 48: 198-205.
- Sela DA (2008) The genome sequence of Bifidobacterium longum subsp. infantis reveals adaptations for milk utilization within the infant microbiome. Proc Nat Acad Sci 105: 18964-18969.
- Favier CF (2002) Molecular monitoring of succession of bacterial communities in human neonates. Appl Environ Microbiol 68: 219-226.
- Palmer C (2007) Development of the human infant intestinal microbiota. Plos Biol 5: e177.
- Kurokawa K (2007) Comparative metagenomics revealed commonly enriched gene sets in human gut microbiomes. DNA Res 14: 169-181.
- Smith MI (2013) Gut microbiomes of Malawian twin pairs discordant for kwashiorkor. Science 339: 548-554.
- Tower J (2009) The genetics of gender and life span. Biology 8: 38. Autoimmun Rev 11: A377-385.
- Nussinovitch U (2012) The role of gender and organ specific autoimmunity.
- Pan Z (2012) Gender and the regulation of longevity: Implications for autoimmunity. Autoimmun Rev 11: A393-403.
- Markle JGM (2013) Sex differences in the gut microbiome drive hormonedependent regulation of autoimmunity. Science 339: 1084-1088.
- Hollister EB (2015) Structure and function of the healthy pre-adolescent pediatric gut microbiome. Microbiome 3: 36.
- Agans R (2011) Distal gut microbiota of adolescent children is different from that of adults. FEMS Microbiol Ecol 77: 404-412.
- Yurkovetskiy L (2013) Gender bias in autoimmunity is influenced by microbiota. Immun 39: 400-412.
- Torres PJ (2019) Letrozole treatment of adult female mice results in a similar reproductive phenotype but distinct changes in metabolism and the gut microbiome compared to pubertal mice. BMC Microbiol 19: 57.
- Hueston CM (2017) Stress and adolescent hippocampal neurogenesis: Diet and exercise as cognitive modulators. Transl Psych 7: e1081.
- Kempermann G (2008) The contribution of failing adult hippocampal neurogenesis to psychiatric disorders. Curr Opinion Psych 21: 290-295.
- Toro C (2007) Adult neurogenesis and schizophrenia: A window on abnormal early brain development? Schizo Res 90: 1-14.
- Nixon K (2010) Adolescence as a critical window for developing an alcohol use disorder: current findings in neuroscience. Curr Opinion Psych 23: 227-232.
- Lee TH (2012) Effects of ADHD therapeutic agents, methylphenidate and atomoxetine, on hippocampal neurogenesis in the adolescent mouse dentate gyrus. Neurosci Letters 524: 84-88.
- Neufeld KA (2016) Reframing the teenage wasteland: Adolescent microbiota- gut-brain axis. Canadian J Pysch 61: 214-221.
- Borre YE (2014) Microbiota and neurodevelopmental windows: Implications for brain disorders. Trends Mol Med 20: 509-518.
- Kundu P (2017) Our gut microbiome: The evolving inner self. Cell 171: 1481- 1493.
- Turnbaugh PJ, Gordon JI (2009) The core gut microbiome, energy balance and obesity. J Physiol 587: 4153-4158.
- Bonder MJ (2016) The effect of host genetics on the gut microbiome. Nat Genet 48: 1407-1412.
- Korem T (2017) Bread affects clinical parameters and induces gut microbiome- associated personal glycemic responses. Cell Metab 25: 1243-1253.e5.
- David LA (2013) Diet rapidly and reproducibly alters the human gut microbiome. Nature 505: 559-563.
- Rajilić-Stojanović M (2013) Long-term monitoring of the human intestinal microbiota composition. Environ Microbiol 15: 1146-1159.
- Gibson MK, Crofts TS, Dantas G (2015) Antibiotics and the developing infant gut microbiota and resistome. Curr Opinion Microbiol 27: 51-56.
- Modi SR, Collins JJ, Relman DA (2014) Antibiotics and the gut microbiota. J Clin Invest 124: 4212-4218.
- De Filippo C (2010) Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci USA 107: 14691-14696.
- Chevalier C (2015) Gut microbiota orchestrates energy homeostasis during cold. Cell 163: 1360-1374.
- Maurice CF (2015) Marked seasonal variation in the wild mouse gut microbiota. ISME J 9: 2423-2434.
- Zhang Z (2016) Convergent evolution of rumen microbiomes in high-altitude mammals. Curr Biol 26: 1873-1879.
- DiGiulio DB (2015) Temporal and spatial variation of the human microbiota during pregnancy. Proceed National Acad Scienc 112: 11060-11065.
- Koren O (2012) Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell 150: 470-480.
- Collado MC (2008) Distinct composition of gut microbiota during pregnancy in overweight and normal-weight women. Am J Clin Nutr 88: 894-899.
- Collado MC (2010) Effect of mother's weight on infant's microbiota acquisition, composition, and activity during early infancy: a prospective follow-up study initiated in early pregnancy. Am J Clin Nutr 92: 1023-1030.
- Laitinen K, Poussa T, Isolauri E (2009) Probiotics and dietary counselling contribute to glucose regulation during and after pregnancy: a randomised controlled trial. Br J Nutr 101: 1679-1687.
- Valles-Colomer M (2019) The neuroactive potential of the human gut microbiota in quality of life and depression. Nat Microbiol
- Rampelli S (2013) Functional metagenomic profiling of intestinal microbiome in extreme ageing. Aging (Albany NY) 5: 902-912.
- Woodmansey EJ (2004) Comparison of compositions and metabolic activities of fecal microbiotas in young adults and in antibiotic-treated and non-antibiotic- treated elderly subjects. Appl Environ Microbiol 70: 6113-6122.
- Jeffery IB, Lynch DB, O'Toole PW (2016) Composition and temporal stability of the gut microbiota in older persons. ISME J 10: 170-182.
- Odamaki T (2016) Age-related changes in gut microbiota composition from newborn to centenarian: A cross-sectional study. BMC Microbiol 16: 90.
- Biagi E (2012) Aging of the human metaorganism: The microbial counterpart. Age 34: 247-267.
- Biagi E (2010) Through ageing, and beyond: Gut microbiota and inflammatory status in seniors and centenarians. PLoS One 5: e10667.
- Zapata HJ, Quagliarello VJ (2015) The microbiota and microbiome in aging: Potential implications in health and age-related diseases. J Am Geriatr Soc 63: 776-781.
- Kong F (2016) Gut microbiota signatures of longevity. Current Biol 26: R832-R833.
- Arboleya S (2016) Gut Bifidobacteria populations in human health and aging. Front Microbiol 2016. 7: 1204.
- Ewaschuk J (2007) Probiotic bacteria prevent hepatic damage and maintain colonic barrier function in a mouse model of sepsis. Hepatol 46: 841-850.
- Mariat D (2009) The firmicutes/bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol 9: 123.
- Langille MG (2014) Microbial shifts in the aging mouse gut. Microbiome 2: 50.
- Biagi E (2016) Gut microbiota and extreme longevity. Current Biol 26: 1480- 1485.
- Biragyn A, Ferrucci L (2018) Gut dysbiosis: A potential link between increased cancer risk in ageing and inflammaging. Lancet Oncol 19: e295-e304.
- O'Toole PW, Jeffery IB (2015) Gut microbiota and aging. Science 350: 1214- 1215.
- Ottaviani E (2011) Gut microbiota as a candidate for lifespan extension: An ecological/evolutionary perspective targeted on living organisms as metaorganisms. Biogerontol 12: 599-609.
- Zhang F (2018) Microbiota transplantation: Concept, methodology and strategy for its modernization. Protein Cell 9: 462-473.
- Xu J (2015) Structural modulation of gut microbiota during alleviation of type 2 diabetes with a Chinese herbal formula. ISME J 9: 552-562.
- Xie W (2011) Effects and action mechanisms of berberine and Rhizoma coptidis on gut microbes and obesity in high-fat diet-fed C57BL/6J mice. PLoS One 6: e24520.
- Zhang X (2012) Structural changes of gut microbiota during berberine- mediated prevention of obesity and insulin resistance in high-fat diet-fed rats. PLoS One 7: e42529.
- Barcena C (2019) Health span and lifespan extension by fecal microbiota transplantation into progeroid mice. Nat Med 25: 1234-1242.
- Callaway E (2017) 'Young poo' makes aged fish live longer. Nature 544: 147.
- Smith P (2017) Regulation of life span by the gut microbiota in the short-lived African turquoise killifish. ELife 6: 1-2.
- Matsumoto M (2011) Longevity in mice is promoted by probiotic-induced suppression of colonic senescence dependent on upregulation of gut bacterial polyamine production. PLoS One 6: e23652.
- Shen ZH (2018) Relationship between intestinal microbiota and ulcerative colitis: Mechanisms and clinical application of probiotics and fecal microbiota transplantation. World J Gastroenterol 24: 5-14.
- Seekatz AM (2018) Restoration of short chain fatty acid and bile acid metabolism following fecal microbiota transplantation in patients with recurrent Clostridium difficile infection. Anaerobe 53: 64-73.
- Bhatnagar D (2015) Gut flora, diet and intestinal metabolism on cardiovascular risk. Curr Opin Lipidol 26: 148-149.
- Velasquez MT (2018) Altered gut microbiota: A link between diet and the metabolic syndrome. Metab Syndr Relat Disord 16: 321-328.
- Tiihonen K, Ouwehand AC, Rautonen N (2010) Human intestinal microbiota and healthy ageing. Ageing Res Rev 9: 107-116.
- Harmsen HJ (2000) Development of 16S rRNA-based probes for the Coriobacterium group and the Atopobium cluster and their application for enumeration of Coriobacteriaceae in human feces from volunteers of different age groups. App Environ Microbiol 66: 4523-4527.
- Magrone T, Jirillo E (2013) The interaction between gut microbiota and age-related changes in immune function and inflammation. Immun Ageing 10: 31.
- Weng Y (2019) Diet-microbiota-metabolite interaction networks reveal key players in inflammatory bowel disease. J Dig Dis 4: 2-4.
- Gehrig JL (2019) Effects of microbiota-directed foods in gnotobiotic animals and undernourished children. Science 365: eaau4732.
- Fontana L, Partridge L (2015) Promoting health and longevity through diet: From model organisms to humans. Cell 161: 106-118.
- Jandhyala SM (2015) Role of the normal gut microbiota. World J Gastroenterol 21: 8787-8803.
- Jourova L, Anzenbacher P, Anzenbacherova E (2016) Human gut microbiota plays a role in the metabolism of drugs. Biomed Pap Med 160: 317-326.
- Mao B (2015) Metagenomic insights into the effects of fructo-oligosaccharides (FOS) on the composition of fecal microbiota in mice. J Agric Food Chem 63: 856-863.
- O'Keefe SJ (2009) Products of the colonic microbiota mediate the effects of diet on colon cancer risk. J Nutr 139: 2044-2048.
- Kaddurah-Daouk R (2011) Enteric microbiome metabolites correlate with response to simvastatin treatment. PLoS One 6: e25482.
- Gusarov I (2013) Bacterial nitric oxide extends the lifespan of C. elegan. Cell 152: 818-830.
- Han B (2017) Microbial genetic composition tunes host longevity. Cell 169: 1249-1262 e13.
- Wakita Y (2018) Taxonomic classification for microbiome analysis, which correlates well with the metabolite milieu of the gut. BMC Microbiol 18: 188.
- Kaeberlein M, Rabinovitch PS, Martin GM (2015) Healthy aging: The ultimate preventative medicine. Science 350: 1191-1193.
- Mannick JB (2014) mTOR inhibition improves immune function in the elderly. Sci Transl Med 6: 268ra179.
- De Haes W (2014) Metformin promotes lifespan through mitohormesis via the peroxiredoxin PRDX-2. Proc Natl Acad Sci USA 111: E2501-2509.
- Harrison DE (2014) Acarbose, 17-alpha-estradiol, and nordihydroguaiaretic acid extend mouse lifespan preferentially in males. Aging Cell 13: 273-282.
- Verdin E (2014) NAD(+) in aging, metabolism, and neurodegeneration. Science 350: 1208-1213.
- Mitchell SJ (2014) The SIRT1 activator SRT1720 extends lifespan and improves health of mice fed a standard diet. Cell Rep 6: 836-843.
- Medvedik O (2007) MSN2 and MSN4 link calorie restriction and TOR to sirtuin-mediated lifespan extension in Saccharomyces cerevisiae. PLoS Biol 5: e261.
- Powers RW (2006) Extension of chronological life span in yeast by decreased TOR pathway signaling. Genes Dev 20: 174-184.
- Anisimov VN (2011) Rapamycin increases lifespan and inhibits spontaneous tumorigenesis in inbred female mice. Cell Cycle 10: 4230-4236.
- Bjedov I (2010) Mechanisms of life span extension by rapamycin in the fruit fly Drosophila melanogaster. Cell Metab 11: 35-46.
- Robida-Stubbs S (2012) TOR signaling and rapamycin influence longevity by regulating SKN-1/Nrf and DAF-16/FoxO. Cell Metab 15: 713-724.
- Cabreiro F (2013) Metformin retards aging in C. elegans by altering microbial folate and methionine metabolism. Cell 153: 228-239.
- Johnson SC, Rabinovitch PS, Kaeberlein M (2013) mTOR is a key modulatorof ageing and age-related disease. Nature 493: 338-345.
- Hurez V (2015) 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 14: 945-956.
- Bitto A (2016) Transient rapamycin treatment can increase lifespan and healthspan in middle-aged mice. Elife 5: 1-2.
- Kapahi P, Zid B (2004) TOR pathway: Linking nutrient sensing to life span. Sci Aging Knowledge Environ 2004: Pe34.
- Harrison DE (2009) Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 460: 392-395.
- Ramos FJ (2014) Rapamycin reverses elevated mTORC1 signaling in lamin A/C-deficient mice, rescues cardiac and skeletal muscle function, and extends survival. Sci Transl Med 4: 144ra103.
- Miller RA (2011) Rapamycin, but not resveratrol or simvastatin, extends life span of genetically heterogeneous mice. J Gerontol A Biol Sci Med Sci 66: 191-201.
- Witte AV (2009) Caloric restriction improves memory in elderly humans. Proc Natl Acad Sci USA 106: 1255-1260.
- Halloran J (2012) Chronic inhibition of mammalian target of rapamycin by rapamycin modulates cognitive and non-cognitive components of behavior throughout lifespan in mice. Neurosci 223: 102-113.
- Majumder S (2012) Lifelong rapamycin administration ameliorates age- dependent cognitive deficits by reducing IL-1beta and enhancing NMDA signaling. Aging Cell 11: 326-335.
- Iatsenko I, Boquete JP, Lemaitre B (2018) Microbiota-derived lactate activates production of reactive oxygen species by the intestinal NADPH Oxidase Nox and shortens Drosophila lifespan. Immunity 49: 929-942.e5.
- Soysal P, Isik AT (2016) Effects of acetylcholinesterase inhibitors on nutritional status in elderly patients with dementia: A 6-month follow-up study. J Nutr Health Aging 20: 398-403.
- Rautiainen S (2017) Effect of baseline nutritional status on long-term multivitamin use and cardiovascular disease risk: A secondary analysis of the physicians' health study ii randomized clinical trial. JAMA Cardiol 2: 617-625.
- Nadkarni NK (2015) Statins and brain integrity in older adults: Secondary analysis of the Health ABC study. Alzheimers Dement 11: 1202-1211.
- Ingrasciotta Y (2019) Analgesic drug use in elderly persons: A population- based study in Southern Italy. PLoS One 14: e0222836.
- Hou Y (2016) The association between self-perceptions of aging and antihypertensive medication adherence in older Chinese adults. Aging Clin Exp Res 28: 1113-1120.
- Little MO (2018) Updates in nutrition and polypharmacy. Curr Opin Clin Nutr Metab Care 21: 4-9.
- Nusdeo G, Terrosu P, Parodi G (2018) Optimal antiplatelet therapy after an acute coronary syndrome in the elderly: An old issue. Int J Cardiol 259: 49-50.
- Schlender L (2017) Efficacy and safety of metformin in the management of type 2 diabetes mellitus in older adults: A systematic review for the development of recommendations to reduce potentially inappropriate prescribing. BMC Geriatr 17: 227.
- Pryor R (2019) Host-microbe-drug-nutrient screen identifies bacterial effectors of metformin therapy. Cell 178: 1299-1312.e29.
- Shin NR (2014) An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 63: 727-735.
- Jung MJ (2016) Chronic Repression of mTOR complex 2 induces changes in the gut microbiota of diet-induced obese mice. Sci Rep 6: 30887.
- Pierotti MA (2013) Targeting metabolism for cancer treatment and prevention: Metformin, an old drug with multi-faceted effects. Oncogene 32: 1475-1487.
- Virk B (2016) Folate acts in E. coli to accelerate C. elegans aging independently of bacterial biosynthesis. Cell Rep 14: 1611-1620.
- Han B (2017) Microbial genetic composition tunes host longevity. Cell 169: 1249-1262.e13.
- Gruber J, Kennedy BK (2017) Microbiome and longevity: Gut microbes send signals to host mitochondria. Cell 169: 1168-1169.
- Muller L, Pawelec G (2014) Aging and immunity - Impact of behavioral intervention. Brain Behav Immun 39: 8-22.
- Aan B, Ferrucci L, (2018) Gut dysbiosis: A potential link between increased cancer risk in ageing and inflammaging. Lancet Oncol 19: e295-e304.
- Li D (2016) The gut microbiota: A treasure for human health. Biotechnol Adv 34: 1210-1224.
- Sanna S (2019) Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat Genet 51: 600-605.
- Vogt CM (2018) Mouse intestinal microbiota reduction favors local intestinal immunity triggered by antigens displayed in Bacillus subtilis biofilm. Microb Cell Fact 17: 187.
- Hayashi A (2017) Intestinal dysbiosis and biotin deprivation induce alopecia through overgrowth of Lactobacillus murinus in mice. Cell Rep 20: 1513-1524.
- Dalbo VJ (2009) The effects of age on skeletal muscle and the phosphocreatine energy system: Can creatine supplementation help older adults. Dyn Med 8: 6.
- Lane CA, Hardy J, Schott JM (2018) Alzheimer's disease. Eur J Neurol 25: 59-70.
- Prospective Studies Collaboration (2009) Body-mass index and cause- specific mortality in 900 000 adults: Collaborative analyses of 57 prospective studies. Curr Biol 373: 1083-1096.
- Machado MV, Cortez-Pinto H (2016) Diet, microbiota, obesity, and NAFLD: A dangerous quartet. Int J Mol Sci 17: 481.
- Zhang Q (2019) Intestinal lysozyme liberates Nod1 ligands from microbes to direct insulin trafficking in pancreatic beta cells. Cell Res 29: 516-532.
- Gioia D (2014) Bifidobacteria: Their impact on gut microbiota composition and their applications as probiotics in infants. 98: 563-577.
- Gross M (2013) Does the gut microbiome hold clues to obesity and diabetes? Curr Biol 23: R359-362.
- Buchwald H (2014) Bariatric surgery: A systematic review and meta-analysis. 292: 1724-1737.
- Zhang H (2009) Human gut microbiota in obesity and after gastric bypass. Proc Nat Acad Sci 106: 2365-2370.
- Graessler J (2013) Metagenomic sequencing of the human gut microbiome before and after bariatric surgery in obese patients with type 2 diabetes: Correlation with inflammatory and metabolic parameters. Pharmacogenom J 13: 514-522.
- Chan YK (2013) Clinical consequences of diet-induced dysbiosis. Ann Nutri Metab 4: 28-40.
- Jumpertz R (2011) Energy-balance studies reveal associations between gut microbes, caloric load, and nutrient absorption in humans. Am J Clin Nutr 94: 58-65.
- Million M (2012) Comparative meta-analysis of the effect of Lactobacillus species on weight gain in humans and animals. Microb Patho 53: 100-108.
- Gérard P (2016) Gut microbiota and obesity. Cell Mol Lif Sci 73: 147-162.
- Turta O (2016) Antibiotics, obesity and the link to microbes - what are we doing to our children? Med 14: 57.
- Riva A (2017) Pediatric obesity is associated with an altered gut microbiota and discordant shifts in Firmicutes populations. Environ Microbiol 19: 95-105.
- Turnbaugh PJ (2008) Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Micro 3: 213-223.
- Pastor-Villaescusa B (2016) Evaluation of differential effects of metformin treatment in obese children according to pubertal stage and genetic variations: A study protocol for a randomized controlled trial. Trials 17: 323.
- Pastor-Villaescusa B (2016) Evaluation of differential effects of metformin treatment in obese children according to pubertal stage and genetic variations: A study protocol for a randomized controlled trial. Trials 17: 323.
- Cani PD (2012) Involvement of gut microbiota in the development of low-grade inflammation and type 2 diabetes associated with obesity. Gut Micro 3: 279-288.
- rmougom F (2009) Monitoring bacterial community of human gut microbiota reveals an increase in Lactobacillus in obese patients and Methanogens in anorexic patients. Plos One 4: e7125.
- Kim KA (2012) High fat diet-induced gut microbiota exacerbates inflammation and obesity in mice via the TLR4 signaling pathway. Plos One 7: e47713.
- Xu P (2012) Correlation of intestinal microbiota with overweight and obesity in Kazakh school children. Microbiol 12: 283.
- Everard A (2013) Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Nat Acad Sci 110: 9066-9071.
- Million M (2012) Obesity-associated gut microbiota is enriched in Lactobacillus reuteri and depleted in Bifidobacterium animalis and Methanobrevibacter smithii. Int J Obesity 36: 817-825.
- López-Contreras BE (2018) Composition of gut microbiota in obese and normal-weight Mexican school-age children and its association with metabolic traits. Ped Obesity 13: 381-388.
- Karlsson CL (2012) The microbiota of the gut in preschool children with normal and excessive body weight. Obesity 20: 2257-2261.
- Payne AN (2011) The metabolic activity of gut microbiota in obese children is increased compared with normal-weight children and exhibits more exhaustive substrate utilization. Nutr Diab 1: e12.
- Goyns MH (1998) Differential display analysis of gene expression indicates that age-related changes are restricted to a small cohort of genes. Mech Ageing Dev 101: 73-90.
- Giresi PG (2005) Identification of a molecular signature of sarcopenia. Physiol Genomics 21: 253-263.
- Mohle L (2016) Ly6C(hi) Monocytes provide a link between antibiotic-induced changes in gut microbiota and adult hippocampal neurogenesis. Cell Rep 15: 1945-1956.
- Murtaza N (2019) The effects of dietary pattern during intensified training on stool microbiota of elite race walkers. Nutrients 11: 261.
- Petersen LM (2017) Community characteristics of the gut microbiomes of competitive cyclists. Microbiome 5: 98.
- Clarke SF (2014) Exercise and associated dietary extremes impact on gut microbial diversity. 63: 1913-1920.
- Scheiman J (2019) Meta-omics analysis of elite athletes identifies a performance-enhancing microbe that functions via lactate metabolism. Nat Med 25: 1104-1109.
- Barton W (2018) The microbiome of professional athletes differs from that of more sedentary subjects in composition and particularly at the functional metabolic level. Gut 67: 625-633.
- Hamilton IR (1973) Carbon dioxide fixation by Veillonella parvula M 4 and its relation to propionic acid formation. Can J Microbiol 19: 715-723.
- Kimura I (2011) Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41). 108: 8030-8035.
- Pluznick J (2014) A novel SCFA receptor, the microbiota, and blood pressure regulation. 5: 202-207.
- Chambers ES (2018) Acute oral sodium propionate supplementation raises resting energy expenditure and lipid oxidation in fasted humans. 20: 1034- 1039.
- Saffrey MJ (2014) Aging of the mammalian gastrointestinal tract: A complex organ system. 36: 9603.
- Sato S, Kiyono H, Fujihashi K (2015) Mucosal immunosenescence in the gastrointestinal tract: A mini-review. Gerontol 61: 336-342.
- Clark RI (2015) Distinct shifts in microbiota composition during drosophila aging impair intestinal function and drive mortality. Cell Rep 12: 1656-1667.
- Woodmansey EJ (2007) Intestinal bacteria and ageing. J Appl Microbiol 102: 1178-1186.
- Kang SJ (2011) Dysbiosis of the faecal microbiota in patients with Crohn's disease and their unaffected relatives. Intes Res 60: 631-637.
- Sokol H (2008) Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci USA 105: 16731-16736.
- Frank DN (2007) Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Nat Acad Sci 104: 13780-13785.
- Vandeputte D (2017) Quantitative microbiome profiling links gut community variation to microbial load. Nature 551: 507-511.
- Fransen F (2017) Aged gut microbiota contributes to systemical inflammaging after transfer to germ-free mice. Front Immunol 8: 1385.
- Derrien M (2004) Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int J Sys Evo Microbiol 54: 1469-1476.
- Santoni M (2018) Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Eur Urol 359: 91-97.
- Blacher E (2019) Potential roles of gut microbiome and metabolites in modulating ALS in mice. Nature 572: 474-480.
- Owczarek D (2016) Diet and nutritional factors in inflammatory bowel diseases.World J Gastroenterol 22: 895-905.
- Chen G (2018) G protein-coupled receptor 109A and host microbiota modulate intestinal epithelial integrity during sepsis. Front Immunol 9: 2079.
- Littman DR (2011) Role of the commensal microbiota in normal and pathogenic host immune responses. Cell Host Microb 10: 311-323.
- Guang G (2018) Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Oncol Times 359: 97-103.
- Klein SL (2016) Immunology, sex differences in immune responses. Nat Rev Immunol 16: 626-638.
- Lyte M (2013) Microbial endocrinology in the microbiome-gut-brain axis: How bacterial production and utilization of neurochemicals influence behavior. PLoS Pathol 9: e1003726.
- Forsythe P, Kunze WA (2013) Cellular, and CMLS, voices from within: Gut microbes and the CNS. Cell Mol Lif Sci 70: 55-69.
- Westfall S (2017) Microbiome, probiotics and neurodegenerative diseases: Deciphering the gut brain axis. Cell Mol Lif Sci 74: 3769-3787.
- Kaelberer MM (2018) A gut-brain neural circuit for nutrient sensory transduction. Sci 361: 1-4.
- Wang HX (2016) Gut microbiota-brain axis. Chinese Med J 129: 2373-2380.
- Borre YE (2014) Microbiota and neurodevelopmental windows: Implications for brain disorders. Trends Mol Med 20: 509-518.
- Murray E (2019) Probiotic consumption during puberty mitigates LPS-induced immune responses and protects against stress-induced depression and anxiety-like behaviors in adulthood in a sex-specific manner. Brain Behav Immun 81: 198-212.
- Lang UE (2009) Immunosuppression using the mammalian target of rapamycin (mTOR) inhibitor everolimus: Pilot study shows significant cognitive and affective improvement. Transplant Proc 41: 4285-4288.
- Jiang H (2015) Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav Immun 48: 186-194.
- Chandra R (2017) α-Synuclein in gut endocrine cells and its implications for Parkinson's disease. JCI Insight 2: 1-2.
- Matheoud D (2019) Intestinal infection triggers Parkinson's disease-like symptoms in Pink1(-/) mice. Nature 571: 565-569.
- Sampson TR (2016) Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson's disease. Cell 167: 1469- 1480.e12.
- Mangiola F (2016) Gut microbiota in autism and mood disorders. World J Gastroenterol 22: 361-368.