You are on page 1of 27

Annual Review of Pharmacology and Toxicology

The Gut Microbiome,


Metformin, and Aging
Sri Nitya Reddy Induri,1,∗ Payalben Kansara,1,∗
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

Scott C. Thomas,1 Fangxi Xu,1 Deepak Saxena,1,2


Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

and Xin Li1


1
Department of Molecular Pathobiology, New York University College of Dentistry,
New York, NY 10010, USA; email: xl15@nyu.edu
2
Department of Surgery, New York University School of Medicine, New York, NY 10016, USA

Annu. Rev. Pharmacol. Toxicol. 2022. 62:85–108 Keywords


First published as a Review in Advance on
gut microbiome, aging, inflammaging, drug toxicity, metformin
August 24, 2021

The Annual Review of Pharmacology and Toxicology is Abstract


online at pharmtox.annualreviews.org
Metformin has been extensively used for the treatment of type 2 diabetes,
https://doi.org/10.1146/annurev-pharmtox-051920-
and it may also promote healthy aging. Despite its widespread use and
093829
versatility, metformin’s mechanisms of action remain elusive. The gut
Copyright © 2022 by Annual Reviews.
typically harbors thousands of bacterial species, and as the concentration of
All rights reserved
metformin is much higher in the gut as compared to plasma, it is plausible

These authors contributed equally to this article
that microbiome-drug-host interactions may influence the functions of
metformin. Detrimental perturbations in the aging gut microbiome lead
to the activation of the innate immune response concomitant with chronic
low-grade inflammation. With the effectiveness of metformin in diabetes
and antiaging varying among individuals, there is reason to believe that the
gut microbiome plays a role in the efficacy of metformin. Metformin has
been implicated in the promotion and maintenance of a healthy gut micro-
biome and reduces many age-related degenerative pathologies. Mechanistic
understanding of metformin in the promotion of a healthy gut microbiome
and aging will require a systems-level approach.

85
We cannot fathom the marvelous complexity of an organic being; but on the hypothesis here advanced
this complexity is much increased. Each living creature must be looked at as a microcosm—a little
universe, formed of a host of self-propagating organisms, inconceivably minute and as numerous as the
stars in heaven.

—Charles Darwin

THE MICROBIOME
The human body is inhabited by trillions of microbial cells from all domains of life (1, 2). The
microbial cells supported by the human body are estimated to outnumber somatic and germ
cells in our bodies by a factor of 10 (3). The collective ensemble of all the microbes, their genes,
and the environmental conditions found in and on the human body is referred to as the human
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

microbiome and creates the human ecosystem. Within the human ecosystem, the localized
differences in the taxa or types of microorganisms present are referred to as microbiota (4–6).
Each individual has a unique microbiome that becomes increasingly unique with age, likely
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

reflecting the culmination of the interactions throughout one’s life, including demographic and
environmental influences (7–10). These diverse microbes typically coexist harmoniously with the
host and can, in some cases, contribute to maintaining host health and immune function. The
composition of the microbiome and any disruption to its integrity can potentially adversely affect
health, disease susceptibility, and disease progression (10, 11). These observations have led to
the human microbiome being considered our so-called last organ (12) and to proposals that the
human body may be better conceptualized as a human microbial superorganism (13–16).
Human microbiome research has substantially expanded its reach in preceding decades—from
understanding the diversity of host microbiota and its influential role on human health, disease,
and evolution to technological advancements in personalized microbiome-based therapies in the
omics fields (e.g., metagenomics, proteomics, metabolomics).
Microorganisms colonize nearly every surface of the human body and can thrive in several or-
gans in our gastrointestinal, genitourinary, and respiratory tracts (13, 17–19). A typical human gut
microbiome is estimated to house at least 1,800 genera and approximately 15,000–36,000 bacte-
rial species that have evolved in a symbiotic relationship with the host, exhibiting temporal and
spatial differences in their distribution from the esophagus to the distal colon (7, 14, 20–22). The
human intestine, with an estimated surface area of 200–300 m2 (23), provides a nutrient-rich envi-
ronment to approximately 100 trillion microbes that encode for 100-fold more unique genes than
encode our genome (16, 24, 25). The healthy colon alone is host to the overwhelming majority
(up to 70%) of the microbial community (1011 –1012 cells/mL) (26), whereas the small intestine is
estimated to contain 103 –109 cells/mL of microbes (27). This homeostasis is mediated by the in-
terplay of a diversity of factors, including migration, translocation, elimination, competition, and
reproduction rates of microbiota, as determined by regional growth conditions (28, 29).
The microorganisms found in the gastrointestinal tract (GIT) are often categorized into au-
tochthonous flora (indigenous flora) and allochthonous flora (transient flora) (30). Autochthonous
flora are the resident microbes that colonize specific habitats in the GIT and that are thought
to be closely associated with the intestinal mucosa, whereas allochthonous flora cannot colonize
specific habitats—barring unusual circumstances—and are usually located in the central lumen
as part of the fecal stream (30, 31). The majority of the autochthonous gut microbiome is strictly
anaerobic and predominantly composed of two bacterial phyla, Firmicutes and Bacteroidetes,
yet members of the archaea can also be found in low relative abundance (e.g., Methanobrevibacter
smithii) (32, 33). Relatively less-represented taxa include Proteobacteria, Actinobacteria, Fusobac-
teria, and Verrucomicrobia (32). Autochthonous microbiota are currently hypothesized to have

86 Induri et al.
evolved symbiotically with the host, thereby developing specialized defense mechanisms against
endogenous antimicrobial peptides while being harmless to the host (34, 35).

Gut Microbiome in the Promotion of Health


The gut microbiome plays a crucial role in maintaining the host’s local and systemic physiology
(35). The spectrum of beneficial services provided by gut microbiota includes gut homeostasis, nu-
trient metabolism, immunomodulation of the host’s gastrointestinal (GI) system (development of
intestinal mucosa and systemic immune systems), maintenance of immune homeostasis, metabolic
activities, and protection against pathogenic microbiota.

Impact on structure and integrity of the intestine. Intestinal epithelial cells (IECs) line the
gut, forming a physicochemical barrier between host tissues and the luminal content, including
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

the gut microbiome. The integrity of this barrier is essential to the host’s health and appears to be
positively affected by a healthy microbiome (36). The cells of the immune system underlying IECs
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

showed altered microvilli morphology and decreased turnover rate in germ-free (GF) animals (37).
Normal gut bacteria also showed an influence on the glycosylation patterns of surface proteins in
the small intestinal epithelium (38). These findings indicate that the integrity and structure of the
GIT are dependent on conventional intestinal bacteria, whereas GF animals have a prolonged cell
cycle duration (39), reduced apoptosis (40), reduced intestinal surface area (up to 30%) (41), and
slower intestinal motility (42).

Nutrient metabolism. The gut microbiome enhances the host’s digestive efficacy and contains
an abundance of genes coding for enzymes involved in the metabolism of glycans and amino
acids as well as in methanogenesis and the biosynthesis of vitamins, isoprenoids, and xenobiotics
(16). Plant-based foods are rich in xylan, pectin, and arabinose-containing polysaccharides that
are indigestible by host enzymes (16). Autochthonous bacteria such as Bacteroides thetaiotaomicron
release glycosyl hydrolases that metabolize plant polysaccharides into simple carbohydrates for
easier absorption by the host (43). The primary resident microbiota can also process glycans,
including nondigestible carbohydrates, and can synthesize short-chain fatty acids (SCFAs) such as
acetate, butyrate, and propionate while also producing gases (e.g., H2 , CO2 ) (16, 44).
The distal gut microbiome is also involved in the synthesis of amino acids and vitamins such
as vitamins B1 (thiamine), B6, and K (16, 45). Polyphenolic compounds such as flavonoids, an-
thocyanidins, and tannins are hydrolyzed by enzymes of Bacteroides, Eubacterium, and Clostridium
species into final products that enter the portal vein (46). Through the microbially mediated trans-
formation of nondigestible carbon and energy sources into SCFAs and other nutrients accessible
to the host, the host gains access to additional caloric and nutrimental sources.

Immunomodulation and gut-immunological homeostasis. The human gut microbiome


is crucially involved in the training and development of the host immune system (47). The
immunomodulation of gut microbiota from gnotobiotic animal studies suggests the influence of
commensal intestinal bacteria on innate and adaptive immune systems. Compared with con-
ventionally housed animals, the mucosal immune system of GF animals is underdeveloped,
with hypoplastic Peyer’s patches with a few germinal centers (48), fewer mesenteric lymph
nodes (49), impaired and hypomature isolated lymphoid follicles (50), and significantly reduced
IgA-producing plasma cells and lamina propria CD4+ T cells (48). Signaling pathways involving
pattern recognition receptors such as Toll-like receptors (TLRs), nucleotide-binding oligomer-
ization domain–containing protein 1 (NOD1)-like receptor family, β-defensin-3, and chemokine

www.annualreviews.org • Gut Microbiome, Metformin, and Aging 87


(C–C motif ) ligand 20 (CCL20) mediated through C–C chemokine receptor 6 (CCR6) are
stimulated by peptidoglycan.
Microbial products induce lymphoid follicles, which play a pivotal role in intestinal immune
responses (50). Suzuki et al. (51) reported that bacterial products and retinoic acid stimulate fol-
licular dendritic cells (FDCs) in Peyer’s patches. Microbially mediated FDC stimulation resulted
in an increase in the expression of chemokine CXCL13, the survival factor BAFF, and chemicals
associated with the activation and secretion of cytokine transforming growth factor (TGF)-β1.
Reduced production of these molecules in GF animals is associated with deficiencies in TLR-
associated pathways (51); these pathways are crucial for microbial recognition and controlling
both innate and adaptive immune responses (52). To improve the tolerance state of the GIT im-
mune system to resident bacteria, dendritic cells in intestinal mucosa induce the differentiation of
naïve T cells into regulatory T cells—referred to as Treg cells—through the production of IL-10
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

(Th2 cytokines) and T cell anergy or depletion (53–56). Taken together, these results suggest that
the gut microbiome can interact with host gut cells in a manner that promotes B cell migration,
survival, and production of IgA, thereby promoting gut health (51).
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

Protection against pathogenic strains. A major function of indigenous gut microbiota is to


protect the host against infection from colonization and overgrowth of pathogenic microbiota
(i.e., pathobionts) (57, 58). A few strategies used by resident gut microbiota to achieve this are
competitive metabolic interactions, localization to certain physical spaces (30, 31), and modulation
of immune responses (59). Probiotic organisms/commensal microbiota such as nonpathogenic
Escherichia coli produce bacteriocin, a peptidic toxin that inhibits the growth of enterohemor-
rhagic E. coli (EHEC) (60, 61). Resident nonpathogenic E. coli and other microbiota help resist
EHEC colonization by competing for nutrients such as amino acids and organic acids, ultimately
leading to the starvation of pathogenic strains (62). Commensals also keep pathogenic microbe
colonization at bay by generating SCFAs, which alter the host’s environmental conditions and
also downregulate virulence genes, for example, those that encode for type III secretion system
(T3SS) proteins in Salmonella enterica serovar Enteritidis and Typhimurium (63).
When the composition of normal gut microbiota changes, some pathobionts are able to
thrive due to the lack of competition from endogenous microbiota. Clostridioides difficile (formerly
Clostridium) infections (CDIs) are a prime example of this dynamic. CDIs result from a dysbiosis
in normal gut microbiota, frequently from antibiotic use (especially broad-spectrum antibiotics)
(64), and the proliferation of C. difficile in the absence of competition from, and the metabolic
byproducts of, the normal microbiota (e.g., secondary bile acids) (58, 65). Of note, the restoration
of a diverse microbiome through fecal transplants from healthy donors has been shown to be an
effective treatment for CDIs (66, 67), demonstrating that a healthy gut microbiota can not only
prevent the colonization of pathogenic strains but also be used as treatment against previously
established pathobionts.

Gut Dysbiosis in the Promotion of Disease


Dysbiosis is a change in the normal or healthy composition of gut microbiota and is involved
in the pathogenesis of multiple disorders (68–70). Recent evidence suggests that dysbiosis of gut
bacteria engenders a myriad of intestinal and extraintestinal disorders (68–70).
Modulation of TLR2, a receptor that normally maintains gut barrier homeostasis, may lead
to inflammation from stress-induced damage and is considered to play a role in the patho-
genesis of inflammatory bowel disease, Crohn’s disease (CD), and ulcerative colitis (UC) (52).

88 Induri et al.
Significant differences in the composition of the gut microbiome have been noted between CD
and UC patients, mainly due to the depletion of the dominant resident phyla (i.e., Firmicutes and
Bacteroidetes) (20). An association between gut dysbiosis and celiac disease has also been reported
by comparing the IgA-coated fecal immunoglobulin levels between patients with a history of celiac
disease and healthy controls (71). Adenomas that are precursors to colorectal cancer have also been
studied to evaluate their association with bacterial community changes (72).
Dysbiosis is considered to be related to metabolic disorders such as obesity and diabetes
(73–75). This is supported by studies in animal models wherein fecal microbiome transplants
from obese mice to GF lean mice or vice versa yielded the phenotype of the microbiota donor
(56). Evidence from metagenome-wide association studies demonstrates a high correlation
between the intestinal microbiome and type 2 diabetes (76). The findings indicate that the
relative abundance of butyrate-producing bacteria such as Roseburia intestinalis, Faecalibacterium
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

prausnitzii, and Akkermansia muciniphila is lower in type 2 diabetics (76). It has also been suggested
that biguanides such as metformin alter the gut microbiota, increasing the relative abundance of a
few genera (e.g., Escherichia/Shigella spp. and Bilophila sp.) while decreasing the relative abundance
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

of others (e.g., Intestinibacter spp. and Clostridium spp.) (77).


Existing data suggest that gut bacteria play a role in central nervous system–related disorders
such as anxiety, depression (78, 79), autism spectrum disorder (80), schizophrenia (81), autoim-
mune diseases (82), epilepsy (83), lung disorders and infections (68), and aging (84, 85). These
observations suggest a direct relationship between the gut microbiota and the brain, corroborat-
ing the concept of a human microbial superorganism.

The Human Gut Microbiome and Drug Metabolism


The interplay between the human gut microbiome, drugs, and related xenobiotics is ex-
tremely complex and considered bidirectional (86). The gut microbiome is capable of directly
and indirectly affecting drug metabolism, disposition, availability, efficacy, and toxicity (87).
Some drugs can alter the composition and diversity of gut microbiota (88). Gut microbially medi-
ated drug metabolism includes biotransformations such as reductive metabolism, hydrolytic reac-
tions, demethylation, deamination, dehydroxylation, deacylation, decarboxylation, deconjugation,
and oxidation (87). Other direct effects include the expression of genetic elements involved in
drug activation, sequestering drugs from the site of action, and developing bacterial transporters
to change drug efficacy (89). Indirect effects of drug metabolism include competitive inhibition
and changes in the host’s metabolic enzymes (87).
Healthy gut bacteria are essential for detoxifying and eliminating a myriad of unwanted
metabolic waste products, but they can also complicate the efficacy of drugs. Clayton et al. (90)
proposed that a microbial metabolite, p-Cresol, has a direct effect on acetaminophen efficacy
through competitive inhibition by reducing the ability of the liver to metabolize acetaminophen.
The GI toxicity of chemotherapeutic CPT-11 (irinotecan) is dramatically increased by a bac-
terial sugar-scavenging β-glucuronidase (91). Studies on nonsteroidal anti-inflammatory drug
(NSAID)-induced enteropathy indicated that gut bacteria are determinants in the development of
GI ulcers in mice treated with NSAIDs (92, 93). Furthermore, secondary gut microbiomes derived
from bile acids contribute to predicting the magnitude of statin-induced low-density lipoprotein
(94). These results indicate a relationship between gut bacterial metabolism and therapeutic out-
comes of a diversity of drugs.
Many classes of drugs are associated with alterations in the composition of the gut microbiota
and their functions (95, 96). Drug-induced dysbiosis can impact the enteric and overall health

www.annualreviews.org • Gut Microbiome, Metformin, and Aging 89


of the host. Multidrug analysis suggested that a few classes of drugs, including antimicrobials,
proton pump inhibitors (PPIs), laxatives, and metformin, have the largest impact on the gut
microbiome (97). Metagenomic analysis of antibiotic-induced changes in gut bacteria showed
that administration of broad-spectrum antibiotics resulted in a tenfold reduction in the number
of gut bacteria, including temporal and spatial changes that cumulatively suggest a decrease in
the abundance of Firmicutes and Lactobacillus species (98). Different classes of antibiotics alter the
gut microbiome differently given their broad spectrum of activity and targets (99). Antibiotic
treatment for 7 days with clindamycin yielded a dramatic decline in the diversity of Bacteroides and
a long-lasting impact on gut microbiota composition that could last up to 2 years posttreatment
(100). A few antibiotics (e.g., vancomycin) have demonstrated eubiotic properties, for example,
in mice models given vancomycin from birth until weaning, it played a protective role against the
development of diabetes and facilitated dominant colonization by A. muciniphila, known for its
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

anti-inflammatory properties (101).


Clarithromycin use, which is given to patients with Helicobacter pylori infection, caused a sharp
decline in Actinobacteria, with the presence of macrolide resistance gene erm(B) being detectable
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

for up to 4 years after the therapeutic regimen (102). PPIs, which are also used in the treatment of
H. pylori infection, can also alter the gut microbiome by directly inhibiting certain endogenous gut
bacteria (e.g., Dorea spp., Ruminococcus spp.) and by modulating the pH of the gut by suppressing
acid production (103, 104). A meta-analysis of 23 studies showed that among 300,000 patients
using PPIs, a 65% increase was found in the incidence of C. difficile–associated diarrhea (105).
Metformin, a common antidiabetic medication used in type 2 diabetics, has been found to alter
the microbial community composition both in vivo and in vitro where it promoted the growth of
Bifidobacterium adolescentis and Bifidobacterium, respectively, whose growth has been suggested to
enhance the antidiabetic effect of metformin (106). Transfer of metformin-altered microbiota to
GF mice showed improved gluconeogenesis triggered by significant improvement in the produc-
tion of SCFAs (i.e., butyrate, propionate) (106, 107).

Microbiome and Aging


The microbiome begins to develop immediately upon birth (108). The gut microbiome is ex-
tremely diverse and complex, and it keeps changing during different stages of life (9, 84, 109–111).
The seven pillars of aging are inflammation, stem cell regeneration, macromolecular damage,
stress, proteostasis, metabolism, and epigenetics (112). Interestingly, the tightly networked aging
pillars converge on inflammation because the impairment of any one pillar stokes inflammation,
which then affects all other pillars (112). Though the precise etiology of inflammaging is not
completely understood, the involvement of the gut microbiome has been highlighted. The
composition of the gut microbiota dramatically changes during aging and is associated with host
health and life span (113–118). The stimulation of TLR4 by lipopolysaccharides from gut micro-
biota can accelerate age-dependent inflammation, a process that can impact early hematopoietic
development and terminal myeloid differentiation potential (119–121). Studies have reported that
age-related gut dysbiosis contributes to a chronic and global inflammatory state in humans (122).
In aged animal models, genera such as Parabacteroides, Mucispirillum, Clostridium, and Sarcina are
positively associated with the proinflammatory MCP-1, while Akkermansia, Oscillospira, Blautia,
and Lactobacillus are negatively correlated with MCP-1. This finding led to the conclusion that
changes in the animal gut microbiota through age are associated with inflammaging, specifically
through increased levels of pro-inflammatory molecules and cells, including IL-6, IL-10, tumor
necrosis factor α (TNF-α), TGF-β, p16, SAM domain and HD domain–containing protein 1
(SAMHD1), eotaxin, regulated upon activation, normal T cell expressed and presumably

90 Induri et al.
secreted (RANTES), T helper type (Th)1, Th2, and Treg; activation of TLR2, nuclear factor
kappa-light-chain-enhancer of activated B cells (NF-κB), and mechanistic target of rapamycin
(mTOR); and decreased levels of cyclin E and cyclin-dependent kinase 2 (CDK2) (122). Human
studies showed that Proteobacteria positively correlated with IL-6 and IL-8, while Ruminococcus
lactaris negatively correlated with IL-8 (122). In aged populations, a reduction in the number
of intestinal commensal bacteria that maintain immune tolerance in the gut has been observed
(122–124). Additionally, opportunistic bacteria that stimulate intestinal inflammation tend to
be more numerous at older ages (125, 126), and microbiota associated with anti-inflammatory
responses decrease (e.g., Bifidobacterium spp., F. prausnitzii) (127).
The precise etiology of inflammaging is not completely understood, but the role of the gut
microbiome and myelopoiesis has been emphasized (128). Myelopoiesis is tightly regulated by
transcription factors and extrinsic signals such as infection and inflammation to ensure homeo-
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

stasis, and dysregulated myelopoiesis may represent the initial step in inflammatory and stress
responses to gut dysbiosis (129, 130). Studies using model organisms indicate that age-related gut
dysbiosis may contribute to unhealthy aging and reduced longevity by triggering the innate im-
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

mune response and chronic low-grade inflammation, leading to many age-related degenerative
pathologies and unhealthy aging (131–138).

METFORMIN
Metformin History and Pharmacology
Metformin (dimethylbiguanide) and the related drug phenformin are derived from galegine, a
natural product from the herb Galega officinalis, which has been known since the 1920s to lower
blood glucose levels; in fact, accounts of the herb being used to treat type 2 diabetes date back
as early as the seventeenth century (139, 140). Phenformin and other derivatives proved to be
more toxic and were associated with lactic acidosis, leading to the preference for metformin (140).
Metformin is currently a preferred orally administered type 2 diabetes drug and holds the title of
the most-prescribed glucose-lowering drug worldwide (139, 141). Yet, despite its historical and
modern widespread use, the detailed mechanisms of metformin action are still unclear (140, 141).
In addition, metformin has proven useful in numerous other applications, ranging from use as a
cardiovascular protective agent to an antiaging agent (141).
Metformin is not bound to any plasma protein. Following intravenous administration, the vol-
ume of distribution has been reported at 63–276 L, and after oral administration of 200 mg daily it
is approximately 600 L; this elevated value designates significant uptake of metformin in the tissue.

Metformin Toxicity
Since its introduction, metformin has been widely consumed as a first-line treatment for
type 2 diabetes that can be used either alone or in combination with sulfonylurea, thiazolidine-
diones, sodium-glucose cotransporter-2 inhibitors, dipeptidyl peptidase-4 inhibitors, glucagon-
like peptide-1 receptor agonists, and insulin (142). The biguanide available in immediate-release
(IR) and extended-release formulations is begun at 500 mg/day and titrated to a maximum dose
of 2,000 mg/day (143).
The toxic markers of biguanide (metformin/phenformin) therapy are considered to be
hyperlactatemia, metabolic acidosis, an increase in the lactate:pyruvate ratio, and elevated
blood alanine concentration (144–146), which occur when metformin concentrations are over
5 μg/mL (147, 148). Metabolic acidosis associated with hyperlactatemia is due to rapid adenosine
triphosphate (ATP) turnover during mitochondrial respiratory complex I inhibition and to the

www.annualreviews.org • Gut Microbiome, Metformin, and Aging 91


insufficient oxidative phosphorylation for ATP hydrolysis (144). Metformin does not undergo
hepatic metabolism and is excreted renally (149, 150). Accumulation of metformin due to reduced
renal clearance or acute/chronic kidney disease results in metformin-associated lactic acidosis
(MALA) (151, 152); however, metformin accumulation does not heighten the risk of acute kidney
injury (153). Although MALA is not subject to a significant accumulation of metformin, it is
associated with comorbidities that contribute to pathological changes (144). The extent of MALA
is determined by blood pH and by concentrations of plasma metformin and lactate pH (150,
151). Metformin-induced lactic acidosis refers to the accumulation of metformin that is largely
responsible for lactic acidosis, without underlying comorbidities (144, 154).
Acute reversible effects of metformin IR involve adverse GI effects that occur in 5–20% of pa-
tients, with diarrhea present in 20% of patients and with less than 5% of patients exhibiting met-
formin intolerance (155). Metformin-associated diarrhea presented with electrolytic deficiencies
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

indicative of hypomagnesemia, hypocalcemia, and hypokalemia (156). These effects can be mini-
mized if it is administered with food, it is used in lower doses, and the dosage is increased slowly
(155). Case reports on metformin-induced hepatotoxicity, hepatitis, and cholestasis have been re-
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

ported in the literature as rare idiosyncratic reactions associated with elevated serum transaminases
markers (157–159). Most of the data on metformin-induced hepatotoxicity were associated with
concomitant intake of other drugs with hepatotoxic potential. A recent case report mentioned that
metformin alone could be responsible for elevated liver enzymes and liver damage in a patient with
no history of liver disease or toxic habits (160).
Some patients (10–30%) receiving long-term metformin therapy have shown evidence of re-
duced vitamin B12 absorption, low serum total vitamin B12, and low bioavailable B12 (holo-
TC II); however, the underlying mechanisms remain unclear (161, 162). Metformin impairs
calcium-dependent membrane activity in the ileum, in turn affecting the vitamin B12 intrinsic
factor complex (162). Metformin-induced vitamin B12 malabsorption is reversible and can be
abated with regular vitamin B12 supplementation (163), with studies currently underway to assess
the efficiency of oral calcium supplementation at treating this deficiency (162).
Management of metformin toxicity consists predominantly of supportive care, including elec-
trolyte replenishment and acid-base, respiratory, renal, and hemodynamic management. Adjunct
therapies such as serum alkalinization, metabolic rescue, extracorporeal techniques (hemodialysis,
hemoperfusion, plasma exchange, continuous renal replacement therapy), and GI decontamina-
tion can be used to manage metformin toxicity in a therapeutic setting (144). In general, the side
effects of metformin are rare and manageable. The data in the literature in the past decade have
suggested metformin’s beneficial effects on conditions beyond glucose control. These conditions,
including neoplasia, cardiovascular diseases, and others, are closely related to aging, which leads
to an intriguing question: Does metformin regulate the aging process, and if so, how?

Metformin and Aging


Life expectancy has been modulated by genetic, pharmacologic, and dietary involvement in
numerous model systems. Similar to the seven pillars of aging (112), Lopez-Otin et al. (164)
recognized and categorized the cellular and molecular hallmarks of aging into nine categories:
(a) genomic instability, (b) epigenetic alterations, (c) loss of proteostasis, (d) deregulated nutrient
sensing, (e) mitochondrial dysfunction, ( f ) cellular senescence, (g) stem cell exhaustion, (h) altered
intercellular communication, and (i) telomere attrition. These hallmarks can help evaluate and
prioritize appropriate interventions. Below, we summarize how metformin directly or indirectly
influences each hallmark of aging.

92 Induri et al.
Genomic instability. One common measure of aging is the accretion of genetic mutations
throughout the life span. Exogenous physical, chemical, and biological agents and endogenous
threats include hydrolytic reactions, DNA replication errors, and reactive oxygen species (ROS),
any of which can compromise the integrity and stability of DNA (165). Studies suggest that
metformin confers genome-defensive effects via reduction of both oxidative stress and DNA im-
pairment. AMP-activated protein kinase (AMPK) is a sensor of cellular energy that is expressed
in essentially all eukaryotic cells, implying that it evolved early during eukaryotic evolution.
Activation of AMPK by metformin can inhibit ROS production and activate p53-mediated
DNA repair (166, 167). A study by Dogan Turacli et al. (168) reinforces the potential benefit of
metformin in an antioxidative capacity to protect cells from diabetic oxidative stress and in the
regulation of DNA damages in the base excision repair system.
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

Epigenetic alterations. Epigenetic alterations include histone modifications, DNA methylation,


and chromatin remodeling, which affect all cells and tissues throughout life. Histone methyla-
tion is the hallmark of aging since histone demethylases modulate the life span by targeting key
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

longevity routes such as insulin and insulin-like growth factor-1 (IGF-1) signaling pathways (169).
AMPK influences histone methylation and activation of SIRT1 (170). Metformin may impact the
epigenome indirectly by modulation of metabolite levels, which is known to alter the activity of
histone and DNA-modulating enzymes. A recent study investigated 300,000 chemical compounds
and more than 9,000 protein binding cavities, yielding up to 41 putative metformin binding tar-
gets (171). Among these potential metformin targets, the H3K27me3 demethylase KDM6A/UTX
contains an experimentally validated unique metformin direct binding motif (171). SUV39H1, a
histone methyltransferase, has been reported to be downregulated by metformin (172).
Metformin has a direct effect on infiltrating immune cells—specifically, by increasing
CD8+ T cell recruitment, protecting them from apoptosis and exhaustion, increasing
CD8+ memory T cells, and regulating age-related inflammation (135, 173–178).

Loss of proteostasis. During aging, some biological processes such as the accumulation of
oxidative damage can lead to the mostly random accumulation of damage in cellular components
(e.g., proteins, DNA). When proteins are damaged as a consequence of various exogenous
and endogenous stress factors, it can lead to the accumulation of protein aggregates, causing
proteotoxic effects if not effectively cleared or recycled, contributing to aging and age-related
diseases. Studies have shown that, via AMPK/ERK1/2 signaling pathways, metformin can restore
antioxidant status and protein homeostasis within cells (179).

Deregulated nutrient sensing. Nutrient availability and sensing are major regulators of homeo-
stasis within cells. However, with aging, the cell begins to lose the ability to maintain homeo-
stasis. Thus, nutrient-sensing systems play a considerable role in aging, longevity, and nutrient
signaling.
The somatotropic axis in mammals comprises IGF-1 and insulin signaling (IIS) pathways, the
most conserved pathways responsible for controlling aging (180). Paradoxically, growth hormone
and IGF-1 levels decline during normal aging and in premature aging, assuming decreased IIS
is a common characteristic of physiological aging (181). Multiple genetic manipulations that
attenuate signaling intensity at different levels of IIS pathways extend the life span of worms and
mice (180). However, Garinis et al. (182) suggested that organisms with a constitutively decreased
IIS can survive longer due to the lower rate of cell growth and metabolism and, hence, lower
rates of cellular damage. Metformin has been shown to decrease the concentration of insulin and
IGF-1 in cancer cells (183, 184). mTOR, AMPK, and sirtuin signaling are the best-characterized

www.annualreviews.org • Gut Microbiome, Metformin, and Aging 93


nutrient-sensing systems, and several studies indicate that metformin is involved in regulating
these systems. Metformin is shown to directly inhibit mTORC1 via Rag-GTPase inhibition and
indirectly via REDD1 upregulation (185). Metformin was also found to be a direct activator of
sirtuins when in low nicotinamide adenine dinucleotide (NAD+) concentrations (186). Met-
formin may decrease perceived hunger and thereby induce dietary restriction (187), leading to
the regulated reduction of food, which can refine late-life health and increase life span in some
organisms (188). Metformin also induces a restriction diet–like state in Caenorhabditis elegans that
reduces brood size, delays reproductive timing, and increases life span independent of transcrip-
tion factor DAF-16/FOXO (189). Metformin’s beneficial effect on aging and energy metabolism
is likely a consequence of directly targeting key energy sensors such as mTOR and AMPK.

Mitochondrial dysfunction. The relationship between mitochondrial dysfunction and aging has
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

long been known. To date, two key molecular foci of metformin have been recognized, both of
which are contained in mitochondria. Having a positive charge, metformin is driven into the cell
and then into the mitochondria in concentrations up to 1,000-fold higher than are found in the
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

extracellular medium, due to differences in the membrane potentials across the mitochondrial
inner membrane and the plasma membrane. In the mitochondria, the most significant action of
metformin is inhibition of complex I of the respiratory chain. Two consequences associated with
suppression of the respiratory chain include inhibition of ATP production and changes in the
NAD+:NADH ratio, which contributes to the effect of metformin on gluconeogenesis. Repres-
sion of ATP production affects gluconeogenesis since ATP production is an energy-intensive pro-
cess, which increases cytoplasmic ADP:ATP and AMP:ATP ratios; these changes activate AMPK
(190, 191). After its activation, AMPK restores the cellular energy balance by activating catabolic
(i.e., ATP-generating) pathways and inhibiting anabolic (i.e., ATP-consuming) pathways. AMPK
also controls the whole-body energy equilibrium, mainly by stimulating food intake and energy
expenditure via the hypothalamus (192, 193).
Metformin also inhibits mitochondrial glycerophosphate dehydrogenase (mGPD), which is
a key component in the glycerophosphate shuttle that increases the lactate:pyruvate ratio and
thereby inhibits endogenous glucose production in the liver (194). AMP induces allosteric regu-
lation of key enzymes in gluconeogenesis by acting synergistically with fructose 2,6-bisphosphate
to inhibit fructose 1,6-bisphosphate and stimulate phosphofructokinase, which then inhibits glu-
coneogenesis. Our team has previously found that metformin can reduce elevated glutamate and
succinate levels to baseline levels in diabetic mice, suggesting that it has the ability to alter mito-
chondrial bioenergetic pathways (195). Metformin’s effects could be partially mediated through
the multifaceted actions of succinate (196).

Cellular senescence. Senescent cells are known to display dramatic alterations in their se-
cretome, proinflammatory cytokines, and matrix metalloproteinases, collectively known as
senescence-associated secretory phenotype (SASP), a phenotype that may contribute to ag-
ing (197). Chronic low-dose metformin administration delays senescence in human diploid
fibroblasts, irrespective of the fact that metformin does not exhibit senolytic properties (176).
Metformin prevents NF-κB translocation into the nucleus, thereby mediating anti-inflammatory
effects and downregulation of SASP (198). Noren Hooten et al. (199) suggested that in human
fibroblasts, metformin lowers the protein levels of p16 and p21 and the RNA levels of IL-6 and
IL-8, all of which increase significantly throughout aging. Recently, Kuang et al. (200) found
that metformin could alleviate oxidative stress–induced senescence by stimulating autophagy and
could partially recover the osteogenic potential of human periodontal ligament cells. Metformin

94 Induri et al.
was shown to reduce SASP in head and neck carcinoma (201), and Yi et al. (202) reported that
in hepatoma, a low concentration of metformin induces p53-dependent senescence. Therefore,
metformin’s anti- or pro-senescence effect varies in cases of aging and cancer tissue, respectively.

Stem cell exhaustion. Stem cell exhaustion holds significance in multiple types of aging-
associated injuries and likely comprises one of the ultimate causes of tissue and organismal aging.
Metformin targets stem cell exhaustion pathways, more specifically activation of glutathione per-
oxidase 7 via nuclear factor erythroid 2–related factor 2, inhibition of AKT/mTOR pathway, and
via the AMPK-activated atypical protein kinase C (aPKC)-CREB binding protein (CBP) pathway,
providing zero therapeutic effect and delaying stem cell aging (176, 203, 204). Metformin induced
rejuvenation and differentiation capacity in oligodendrocyte progenitor cells, thereby improving
remyelination and neurogenesis (205).
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

Altered intercellular communication. A prominent aging-associated alteration in intercellular


communication is inflammaging (164). Inflammaging (206) is a significant risk factor for both mor-
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

bidity and mortality in the elderly given that most—if not all—age-related diseases share inflam-
matory pathogenesis (207). Bauer et al. (208) showed that treatment with metformin can restore
intestinal sodium-glucose cotransporter 1 expression and glucose sensing while shifting the com-
position of the upper small intestinal microbiota, partly by increasing the abundance of Lactobacil-
lus. In another study, metformin acted in part through a Bacteroides fragilis–glycoursodeoxycholic
acid–intestinal farnesoid X receptor (FXR) axis to alter intercellular communication to improve
metabolic dysfunction, including hyperglycemia (209).

Telomere attrition. Telomeres, the terminal regions on the chromosome, are particularly suscep-
tible to age-related deterioration given that normal aging is accompanied by telomere attrition in
mammals. In diseases like pulmonary fibrosis, dyskeratosis congenita, and aplastic anemia, there
is a loss of regenerative capacity in different tissues due to telomerase deficiency and acceler-
ated aging due to telomere shortening (210). Moreover, genetically modified animal models with
shortened or lengthened telomeres exhibit decreased or increased life span, respectively (211).
Metformin’s action on mitochondrial function and cellular aging may give rise to protective feed-
back mechanisms on telomere preservation. Studies have reported that metformin is shown to
reduce telomere shortening in the diabetic patient (212, 213).

Metformin and Gut Microbiota


Metformin regulates the gut microbiome, and the identification of processes by which metformin
modulates gut dysbiosis to reduce age-related inflammation—and its interaction with mitochon-
drial dysfunction—is important for modulating inflammaging in the elderly. It is noteworthy that
metformin levels in the gut are 100- to 300-fold higher than those in serum, making the gut the
primary reservoir for metformin in humans and when administered orally. Indeed, the microbiome
appears to be a primary source for metabolic interactions (106, 214, 215). Further, not all individ-
uals who are prescribed metformin derive the same benefit, and some develop side effects (175,
216–218). These facts raise intriguing questions about the role played by the gut microbiome in
metformin tolerance or intolerance (215) and the molecular processes by which metformin regu-
lates the gut microbiome.
Changes in the gut microbiota may affect the gut metabolome, which, in turn, affects intestinal
output of butyrate and acetate (219). Bacteria metabolize unabsorbable carbohydrates into SCFA-
stimulating goblet cells, which leads to further mucin production and a subsequent increase in

www.annualreviews.org • Gut Microbiome, Metformin, and Aging 95


mucus layer thickness, a reduction in epithelial permeability, a decrease in inflammation, and
lowered glucose levels (214). Studies reported that participants treated with metformin exhibited
increased production of butyrate and propionate, while controls showed an increase in microbial
genes implicated in the degradation of glycine and tryptophan (219). SCFA-producing bacteria,
including Butyrivibrio (belonging to the class Clostridia) and Roseburia (belonging to the phylum
Firmicutes), produce SCFAs from the fermentation of indigestible food such as complex carbo-
hydrates (219). This is of particular interest given that glycine has been associated with insulin
sensitivity and that glycine supplementation has been reported to improve insulin sensitivity
(220). A meta-analysis of 199 individuals with type 2 diabetes and 544 healthy controls confirmed
that metformin significantly altered gut microbiota composition (132). Using a large diverse
cohort, Forslund et al. (221) suggested that metformin efficacy in the treatment of type 2 diabetes
is microbially mediated through the production of SCFAs, improving glucose control, but that
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

metformin can also produce adverse effects (e.g., greater relative abundance of Escherichia spp.
and associated virulence factors).
Though metformin was found to increase the relative abundance of A. muciniphila, the under-
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

lying mechanism is not fully understood (222). Metformin is also associated with an increase in
the density of mucin-producing cells (222). Interestingly, A. muciniphila is found predominantly
in the mucus layer of the colon, where it promotes mucus secretion and enhances intestinal in-
tegrity by reducing its epithelial permeability (223). Further supporting a microbiota-metformin
link to gut health, expression of the genes MUC2 and MUC5, two markers for analyzing mucin lev-
els, significantly increased in high-fat diet (HFD)-fed female mice treated with metformin (224).
However, Bauer et al. (208) did not find an increase of A. muciniphila with metformin. This is
probably due to the fact that the analysis was done in the upper small intestine and A. muciniphila
colonizes more in the cecum and colon. Montandon & Jornayvaz (225) summarize that metformin
enhances SCFA-producing bacterial taxa, including Allobaculum, Bacteroides, Blautia, Butyricicoccus,
Lactobacillus, Akkermansia, and Phascolarctobacterium in rats.
Bauer et al. (208) reported that a HFD reduces glucose sensing and SGLT-1 expression, both
of which are important for lowering glucose production in rodents. Metformin restores SGLT-1
expression and glucose sensing while partly modifying the composition of the upper small intesti-
nal microbiota by increasing the abundance of Lactobacillus (208). Zhou et al. (226) reported that
in mice fed a HFD, metformin restored the concentration of the tight junction protein occludin-1
in the gut, reversed the increased gut permeability, and increased the relative abundance of bene-
ficial bacteria Lactobacillus sp. and A. muciniphila. Using a metagenomic approach, Shin et al. (214)
described that metformin had a different effect on microbial composition in HFD mice and nor-
mal caloric diet mice, indicating that the effect is diet dependent. Metformin causes a decrease
in the proportions of Anaerotruncus, Lactococcus, Parabacteroides, Odoribacter, Lawsonia, Blautia, and
Lactonifactor and increases the proportions of Akkermansia and Alistipes.
Metformin reduces the concentration of B. fragilis; subsequently, the bile acid glycoursodeoxy-
cholic acid (GUDCA) is increased through a decrease in bacterial bile salt hydrolase activity.
GUDCA is known to exert anti-inflammatory effects by reducing the level of proinflammatory
cytokines and antagonizing the FXR (209, 227). The pathogenic capsular lipopolysaccharides of
B. fragilis are elevated in obese and diabetic patients and associated with dyslipidemia, increased
blood pressure, and insulin resistance (209).

Metformin, the Microbiome, and Aging


Studies have shown that the life span of C. elegans can only be extended with the use of metformin
if specific microbiota are present. Interestingly, this effect relies on the susceptibility of the

96 Induri et al.
nematode gut microbiota to metformin (when glucose is present) (228). When E. coli cells are
incapable of metabolizing metformin, nematode life span is decreased, suggesting a direct link
between the microbiota-specific metformin mode of action and host physiological benefits that
result in a prolonged life span (228).
Metformin directly affects methionine and folate metabolism in E. coli, resulting in methio-
nine restriction in the nematode host (228, 229). Disruption of microbial folate metabolism
increases host life span, although it does not affect host folate level, suggesting that other E. coli
folate-associated pathways may be involved. Inhibition of bacterial methionine synthase (MS)
irreversibly converts 5,10-methylene-THF to 5-methyl-THF and accumulates 5-methyl-THF
via the methyl trap mechanism. Metformin-induced impairment of the bacterial methionine
cycle caused an 86% increase in S-adenosylmethionine (SAMe) levels and a 33% increase in
S-adenosylhomocysteine (SAH) levels. SAMe inhibits the folate cycle, reduces methionine
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

production by blocking methylene THF reductase, and increases host life span.
The life-extending effect of metformin is mediated by metformin-sensitive E. coli, and in the
absence of E. coli, the life span of C. elegans is shortened due to drug toxicity (228). Conversely,
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

proliferating E. coli can block the alimentary canal in older worms; however, antibiotic treatment
can prevent this proliferation and increase worm life span (230). These differences suggest that
a harmony between host and microbiota must be established in order to maximize life-extending
benefits.
Another study that employed a four-way high-throughput screening method in conjunction
with in silico human microbiota metabolic modeling strongly suggested that microbially produced
agmatine is necessary for the beneficial effects of metformin on host lipid metabolism and aging
in the model organisms C. elegans and Drosophila melanogaster (231). These findings corroborate
an interplay between host nutrient sources (e.g., sugars), specific bacterial taxa (e.g., E. coli and
other Enterobacteriales), bacterial metabolism (e.g., production of agmatine), and host health, sug-
gesting that a holistic approach is needed for a comprehensive understanding of the roles played
by metformin in the gut microbiome and in aging (231).

CONCLUSION
Metformin has been one of the most common medications worldwide for six consecutive decades.
However, the mechanisms underlying its healthy aging–associated effects remain elusive. Preclin-
ical and observational data from humans suggest that metformin has antiaging and prolongevity
properties and thus has the potential to promote healthy aging (232, 233). Beyond glucose re-
duction, metformin appears to target a number of aging-related mechanisms (Figure 1). The
bioavailability of metformin in the gut is 300 times higher than in plasma (106, 214, 215), and
metformin is known to affect the gut microbiome (106, 214, 215). Further, not all individuals who
are prescribed metformin derive identical benefits, and some develop side effects or metformin
toxicity (175, 216–218). This raises intriguing questions about the role that the gut microbiome
plays in metformin tolerance or intolerance (215) and about how metformin regulates the gut mi-
crobiome. Studies using model organisms indicate that age-related gut dysbiosis may contribute
to unhealthy aging and reduced longevity. Gut dysbiosis can trigger the innate immune response
and chronic low-grade inflammation, leading to many age-related degenerative pathologies and
unhealthy aging (131–138). Metformin treatment can suppress the growth of pathogenic bacteria
in aging to inhibit inflammaging and immunosuppression promoted by various pathobionts. How-
ever, the effects of metformin on the microbiome in the context of aging and longevity require
further research.

www.annualreviews.org • Gut Microbiome, Metformin, and Aging 97


E AR LY LI FE
Microbiome
homeostasis

Gut-immunological
homeostasis

Dysbiosis

Leaky gut
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

Metabolic
disorder
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

Immune
dysfunction

Inflammation

A G I NG
Figure 1
The gut microbiome maintains gut homeostasis during early life; however, dysbiosis may occur later in life
and contribute to immune/metabolic disorders. Metformin has the potential to attenuate this dysfunction at
multiple stages. Figure adapted from an image created with BioRender.com.

DISCLOSURE STATEMENT
The authors are not aware of any affiliations, memberships, funding, or financial holdings that
might be perceived as affecting the objectivity of this review.

ACKNOWLEDGMENTS
This research project was supported by the National Institutes of Health grant AG068857 (D.S.,
X.L.) and the NYU Mega-Grant Initiative (D.S., X.L.).

LITERATURE CITED
1. Gevers D, Knight R, Petrosino JF, Huang K, McGuire AL, et al. 2012. The Human Microbiome Project:
a community resource for the healthy human microbiome. PLOS Biol. 10:e1001377
2. Ursell LK, Clemente JC, Rideout JR, Gevers D, Caporaso JG, Knight R. 2012. The interpersonal
and intrapersonal diversity of human-associated microbiota in key body sites. J. Allergy Clin. Immunol.
129:1204–8
3. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. 2007. The human mi-
crobiome project. Nature 449:804–10
4. Marchesi JR, Ravel J. 2015. The vocabulary of microbiome research: a proposal. Microbiome 3:31
5. Lederberg J, McCray AT. 2001. ‘Ome sweet ‘omics—a genealogical treasury of words. Scientist 15:8

98 Induri et al.
6. Berg G, Rybakova D, Fischer D, Cernava T, Verges MC, et al. 2020. Microbiome definition re-visited:
old concepts and new challenges. Microbiome 8:103
7. Scepanovic P, Hodel F, Mondot S, Partula V, Byrd A, et al. 2019. A comprehensive assessment of de-
mographic, environmental, and host genetic associations with gut microbiome diversity in healthy indi-
viduals. Microbiome 7:130
8. Flores GE, Caporaso JG, Henley JB, Rideout JR, Domogala D, et al. 2014. Temporal variability is a
personalized feature of the human microbiome. Genome Biol. 15:531
9. Wilmanski T, Diener C, Rappaport N, Patwardhan S, Wiedrick J, et al. 2021. Gut microbiome pattern
reflects healthy ageing and predicts survival in humans. Nat. Metab. 3:274–86
10. Gilbert JA. 2015. Our unique microbial identity. Genome Biol. 16:97
11. Honda K, Littman DR. 2012. The microbiome in infectious disease and inflammation. Annu. Rev.
Immunol. 30:759–95
12. Baquero F, Nombela C. 2012. The microbiome as a human organ. Clin. Microbiol. Infect. 18(Suppl. 4):2–4
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

13. Niemeyer-van der Kolk T, van der Wall HEC, Balmforth C, Van Doorn MBA, Rissmann R. 2018.
A systematic literature review of the human skin microbiome as biomarker for dermatological drug
development. Br. J. Clin. Pharmacol. 84:2178–93
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

14. Gevers D, Knight R, Petrosino JF, Huang K, McGuire AL, et al. 2012. The human microbiome project:
a community resource for the healthy human microbiome. PLOS Biol. 10:e1001377
15. Hum. Microbiome Proj. Consort. 2012. A framework for human microbiome research. Nature 486:215–
21
16. Gill SR, Pop M, Deboy RT, Eckburg PB, Turnbaugh PJ, et al. 2006. Metagenomic analysis of the human
distal gut microbiome. Science 312:1355–59
17. Neish AS. 2009. Microbes in gastrointestinal health and disease. Gastroenterology 136:65–80
18. Verstraelen H. 2008. Cutting edge: the vaginal microflora and bacterial vaginosis. Verh. K. Acad. Geneeskd.
Belg. 70:147–74
19. Faner R, Sibila O, Agusti A, Bernasconi E, Chalmers JD, et al. 2017. The microbiome in respiratory
medicine: current challenges and future perspectives. Eur. Respir. J. 49:1602086
20. Frank DN, St. Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR. 2007. Molecular-
phylogenetic characterization of microbial community imbalances in human inflammatory bowel dis-
eases. PNAS 104:13780–85
21. Nayfach S, Roux S, Seshadri R, Udwary D, Varghese N, et al. 2020. A genomic catalog of Earth’s mi-
crobiomes. Nat. Biotechnol. 39:499–509
22. Almeida A, Nayfach S, Boland M, Strozzi F, Beracochea M, et al. 2021. A unified catalog of 204,938
reference genomes from the human gut microbiome. Nat. Biotechnol. 39:105–14
23. Gebbers JO, Laissue JA. 1989. Immunologic structures and functions of the gut. Schweiz. Arch. Tierheilkd
131:221–38
24. Ley RE, Peterson DA, Gordon JI. 2006. Ecological and evolutionary forces shaping microbial diversity
in the human intestine. Cell 124:837–48
25. Ursell LK, Clemente JC, Rideout JR, Gevers D, Caporaso JG, Knight R. 2012. The interpersonal
and intrapersonal diversity of human-associated microbiota in key body sites. J. Allergy Clin. Immunol.
129:1204–8
26. Whitman WB, Coleman DC, Wiebe WJ. 1998. Prokaryotes: the unseen majority. PNAS 95:6578–83
27. Berg RD. 1996. The indigenous gastrointestinal microflora. Trends Microbiol. 4:430–35
28. Dickson RP, Erb-Downward JR, Huffnagle GB. 2015. Homeostasis and its disruption in the lung mi-
crobiome. Am. J. Physiol. Lung Cell Mol. Physiol. 309:L1047–55
29. Manfredo Vieira S, Hiltensperger M, Kumar V, Zegarra-Ruiz D, Dehner C, et al. 2018. Translocation
of a gut pathobiont drives autoimmunity in mice and humans. Science 359:1156–61
30. Savage D. 1977. Microbial ecology of the gastrointestinal tract. Annu. Rev. Microbiol. 31:107–33
31. Tannock GW. 1999. Analysis of the intestinal microflora: a renaissance. Antonie Van Leeuwenhoek 76:265–
78
32. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, et al. 2005. Diversity of the human
intestinal microbial flora. Science 308:1635–38

www.annualreviews.org • Gut Microbiome, Metformin, and Aging 99


33. Backhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. 2005. Host-bacterial mutualism in the
human intestine. Science 307:1915–20
34. Muller CA, Autenrieth IB, Peschel A. 2005. Innate defenses of the intestinal epithelial barrier. Cell Mol.
Life Sci. 62:1297–307
35. Nava GM, Stappenbeck TS. 2011. Diversity of the autochthonous colonic microbiota. Gut Microbes
2:99–104
36. Peterson LW, Artis D. 2014. Intestinal epithelial cells: regulators of barrier function and immune ho-
meostasis. Nat. Rev. Immunol. 14:141–53
37. Abrams GD, Bauer H, Sprinz H. 1963. Influence of the normal flora on mucosal morphology and cellular
renewal in the ileum. A comparison of germ-free and conventional mice. Lab. Investig. 12:355–64
38. Bry L, Falk PG, Midtvedt T, Gordon JI. 1996. A model of host-microbial interactions in an open mam-
malian ecosystem. Science 273:1380–83
39. Alam M, Midtvedt T, Uribe A. 1994. Differential cell kinetics in the ileum and colon of germfree rats.
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

Scand. J. Gastroenterol. 29:445–51


40. Sommer F, Bäckhed F. 2013. The gut microbiota—masters of host development and physiology. Nat.
Rev. Microbiol. 11:227–38
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

41. Gordon HA, Bruckner-Kardoss E. 1961. Effect of normal microbial flora on intestinal surface area. Am.
J. Physiol. 201:175–78
42. Husebye E, Hellstrom PM, Midtvedt T. 1994. Intestinal microflora stimulates myoelectric activity of rat
small intestine by promoting cyclic initiation and aboral propagation of migrating myoelectric complex.
Dig. Dis. Sci. 39:946–56
43. Xu J, Bjursell MK, Himrod J, Deng S, Carmichael LK, et al. 2003. A genomic view of the human-
Bacteroides thetaiotaomicron symbiosis. Science 299:2074–76
44. Macfarlane S, Macfarlane GT. 2003. Regulation of short-chain fatty acid production. Proc. Nutr. Soc.
62:67–72
45. Yoshii K, Hosomi K, Sawane K, Kunisawa J. 2019. Metabolism of dietary and microbial vitamin B family
in the regulation of host immunity. Front. Nutr. 6:48
46. Marin L, Miguelez EM, Villar CJ, Lombo F. 2015. Bioavailability of dietary polyphenols and gut micro-
biota metabolism: antimicrobial properties. Biomed. Res. Int. 2015:905215
47. Zheng D, Liwinski T, Elinav E. 2020. Interaction between microbiota and immunity in health and
disease. Cell Res. 30:492–506
48. Macpherson AJ, Harris NL. 2004. Interactions between commensal intestinal bacteria and the immune
system. Nat. Rev. Immunol. 4:478–85
49. Round JL, Mazmanian SK. 2009. The gut microbiota shapes intestinal immune responses during health
and disease. Nat. Rev. Immunol. 9:313–23
50. Bouskra D, Brezillon C, Berard M, Werts C, Varona R, et al. 2008. Lymphoid tissue genesis induced by
commensals through NOD1 regulates intestinal homeostasis. Nature 456:507–10
51. Suzuki K, Maruya M, Kawamoto S, Sitnik K, Kitamura H, et al. 2010. The sensing of environmental
stimuli by follicular dendritic cells promotes immunoglobulin A generation in the gut. Immunity 33:71–
83
52. Cario E, Gerken G, Podolsky DK. 2007. Toll-like receptor 2 controls mucosal inflammation by regu-
lating epithelial barrier function. Gastroenterology 132:1359–74
53. Cools N, Petrizzo A, Smits E, Buonaguro FM, Tornesello ML, et al. 2011. Dendritic cells in the patho-
genesis and treatment of human diseases: a Janus Bifrons? Immunotherapy 3:1203–22
54. Shen C, He Y, Cheng K, Zhang D, Miao S, et al. 2011. Killer artificial antigen-presenting cells deplete
alloantigen-specific T cells in a murine model of alloskin transplantation. Immunol. Lett. 138:144–55
55. Kelsall BL, Leon F. 2005. Involvement of intestinal dendritic cells in oral tolerance, immunity to
pathogens, and inflammatory bowel disease. Immunol. Rev. 206:132–48
56. Iwasaki A, Kelsall BL. 1999. Freshly isolated Peyer’s patch, but not spleen, dendritic cells produce inter-
leukin 10 and induce the differentiation of T helper type 2 cells. J. Exp. Med. 190:229–39
57. Pickard JM, Zeng MY, Caruso R, Núñez G. 2017. Gut microbiota: role in pathogen colonization, im-
mune responses, and inflammatory disease. Immunol. Rev. 279:70–89

100 Induri et al.


58. Kho ZY, Lal SK. 2018. The human gut microbiome—a potential controller of wellness and disease.
Front. Microbiol. 9:1835
59. Kamada N, Chen GY, Inohara N, Nunez G. 2013. Control of pathogens and pathobionts by the gut
microbiota. Nat. Immunol. 14:685–90
60. Schamberger GP, Diez-Gonzalez F. 2002. Selection of recently isolated colicinogenic Escherichia coli
strains inhibitory to Escherichia coli O157:H7. J. Food Prot. 65:1381–87
61. Hammami R, Fernandez B, Lacroix C, Fliss I. 2013. Anti-infective properties of bacteriocins: an update.
Cell Mol. Life Sci. 70:2947–67
62. Momose Y, Hirayama K, Itoh K. 2008. Competition for proline between indigenous Escherichia coli and
E. coli O157:H7 in gnotobiotic mice associated with infant intestinal microbiota and its contribution to
the colonization resistance against E. coli O157:H7. Antonie Van Leeuwenhoek 94:165–71
63. Gantois I, Ducatelle R, Pasmans F, Haesebrouck F, Hautefort I, et al. 2006. Butyrate specifically down-
regulates Salmonella pathogenicity island 1 gene expression. Appl. Environ. Microbiol. 72:946–49
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

64. Bartlett JG. 2002. Antibiotic-associated diarrhea. N. Engl. J. Med. 346:334–39


65. Sorg JA, Sonenshein AL. 2008. Bile salts and glycine as cogerminants for Clostridium difficile spores.
J. Bacteriol. 190:2505–12
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

66. Van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal EG, et al. 2013. Duodenal infusion of donor
feces for recurrent Clostridium difficile. N. Engl. J. Med. 368:407–15
67. Konturek PC, Koziel J, Dieterich W, Haziri D, Wirtz S, et al. 2016. Successful therapy of Clostridium
difficile infection with fecal microbiota transplantation. J. Physiol. Pharmacol. 67:859–66
68. Fabbrizzi A, Amedei A, Lavorini F, Renda T, Fontana G. 2019. The lung microbiome: clinical and
therapeutic implications. Intern. Emerg. Med. 14:1241–50
69. Motta J-P, Wallace JL, Buret AG, Deraison C, Vergnolle N. 2021. Gastrointestinal biofilms in health
and disease. Nat. Rev. Gastroenterol. Hepatol. 18:314–34
70. Belizário JE, Faintuch J. 2018. Microbiome and gut dysbiosis. In Metabolic Interaction in Infection, ed.
R Silvestre, E Torrado, pp. 459–76. Cham, Switz.: Springer
71. De Palma G, Nadal I, Medina M, Donat E, Ribes-Koninckx C, et al. 2010. Intestinal dysbiosis and
reduced immunoglobulin-coated bacteria associated with coeliac disease in children. BMC Microbiol.
10:63
72. Shen XJ, Rawls JF, Randall T, Burcal L, Mpande CN, et al. 2010. Molecular characterization of mucosal
adherent bacteria and associations with colorectal adenomas. Gut Microbes 1:138–47
73. Maruvada P, Leone V, Kaplan LM, Chang EB. 2017. The human microbiome and obesity: moving
beyond associations. Cell Host Microbe 22:589–99
74. Wei B, Wang Y, Xiang S, Jiang Y, Chen R, Hu N. 2020. Alterations of gut microbiome in patients
with type 2 diabetes mellitus who had undergone cholecystectomy. Am. J. Physiol. Endocrinol. Metab.
320:E113–21
75. Sedighi M, Razavi S, Navab-Moghadam F, Khamseh ME, Alaei-Shahmiri F, et al. 2017. Comparison of
gut microbiota in adult patients with type 2 diabetes and healthy individuals. Microbial. Pathog. 111:362–
69
76. Tilg H, Moschen AR. 2014. Microbiota and diabetes: an evolving relationship. Gut 63:1513–21
77. Bryrup T, Thomsen CW, Kern T, Allin KH, Brandslund I, et al. 2019. Metformin-induced changes
of the gut microbiota in healthy young men: results of a non-blinded, one-armed intervention study.
Diabetologia 62:1024–35
78. Foster JA, McVey Neufeld KA. 2013. Gut-brain axis: how the microbiome influences anxiety and de-
pression. Trends Neurosci. 36:305–12
79. Mussell M, Kroenke K, Spitzer RL, Williams JB, Herzog W, Lowe B. 2008. Gastrointestinal symptoms
in primary care: prevalence and association with depression and anxiety. J. Psychosom. Res. 64:605–12
80. De Angelis M, Piccolo M, Vannini L, Siragusa S, De Giacomo A, et al. 2013. Fecal microbiota and
metabolome of children with autism and pervasive developmental disorder not otherwise specified.
PLOS ONE 8:e76993
81. Castro-Nallar E, Bendall ML, Perez-Losada M, Sabuncyan S, Severance EG, et al. 2015. Composition,
taxonomy and functional diversity of the oropharynx microbiome in individuals with schizophrenia and
controls. PeerJ 3:e1140

www.annualreviews.org • Gut Microbiome, Metformin, and Aging 101


82. De Luca F, Shoenfeld Y. 2019. The microbiome in autoimmune diseases. Clin. Exp. Immunol. 195:74–
85
83. Şafak B, Altunan B, Topçu B, Eren Topkaya A. 2020. The gut microbiome in epilepsy. Microbial. Pathog.
139:103853
84. Haran JP, McCormick BA. 2021. Aging, frailty, and the microbiome—how dysbiosis influences human
aging and disease. Gastroenterology 160:507–23
85. Ragonnaud E, Biragyn A. 2021. Gut microbiota as the key controllers of “healthy” aging of elderly
people. Immunity Ageing 18:2
86. Zimmermann M, Zimmermann-Kogadeeva M, Wegmann R, Goodman AL. 2019. Mapping human mi-
crobiome drug metabolism by gut bacteria and their genes. Nature 570:462–67
87. Wilson ID, Nicholson JK. 2017. Gut microbiome interactions with drug metabolism, efficacy, and tox-
icity. Transl. Res. 179:204–22
88. Freedberg DE, Toussaint NC, Chen SP, Ratner AJ, Whittier S, et al. 2015. Proton pump inhibitors
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

alter specific taxa in the human gastrointestinal microbiome: a crossover trial. Gastroenterology 149:883–
85.e9
89. Sun C, Chen L, Shen Z. 2019. Mechanisms of gastrointestinal microflora on drug metabolism in clinical
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

practice. Saudi Pharm. J. 27:1146–56


90. Clayton TA, Baker D, Lindon JC, Everett JR, Nicholson JK. 2009. Pharmacometabonomic identifica-
tion of a significant host-microbiome metabolic interaction affecting human drug metabolism. PNAS
106:14728–33
91. Wallace BD, Wang H, Lane KT, Scott JE, Orans J, et al. 2010. Alleviating cancer drug toxicity by
inhibiting a bacterial enzyme. Science 330:831–35
92. Uejima M, Kinouchi T, Kataoka K, Hiraoka I, Ohnishi Y. 1996. Role of intestinal bacteria in ileal ulcer
formation in rats treated with a nonsteroidal antiinflammatory drug. Microbiol. Immunol. 40:553–60
93. Robert A, Asano T. 1977. Resistance of germfree rats to indomethacin-induced intestinal lesions.
Prostaglandins 14:333–41
94. Kaddurah-Daouk R, Baillie RA, Zhu H, Zeng ZB, Wiest MM, et al. 2011. Enteric microbiome metabo-
lites correlate with response to simvastatin treatment. PLOS ONE 6:e25482
95. Jackson MA, Verdi S, Maxan ME, Shin CM, Zierer J, et al. 2018. Gut microbiota associations with
common diseases and prescription medications in a population-based cohort. Nat. Commun. 9:2655
96. Maurice CF, Haiser HJ, Turnbaugh PJ. 2013. Xenobiotics shape the physiology and gene expression of
the active human gut microbiome. Cell 152:39–50
97. Vich Vila A, Collij V, Sanna S, Sinha T, Imhann F, et al. 2020. Impact of commonly used drugs on the
composition and metabolic function of the gut microbiota. Nat. Commun. 11:362
98. Hill DA, Hoffmann C, Abt MC, Du Y, Kobuley D, et al. 2010. Metagenomic analyses reveal antibiotic-
induced temporal and spatial changes in intestinal microbiota with associated alterations in immune cell
homeostasis. Mucosal Immunol. 3:148–58
99. Ianiro G, Tilg H, Gasbarrini A. 2016. Antibiotics as deep modulators of gut microbiota: between good
and evil. Gut 65:1906–15
100. Jernberg C, Lofmark S, Edlund C, Jansson JK. 2007. Long-term ecological impacts of antibiotic admin-
istration on the human intestinal microbiota. ISME J. 1:56–66
101. Hansen CH, Krych L, Nielsen DS, Vogensen FK, Hansen LH, et al. 2012. Early life treatment with
vancomycin propagates Akkermansia muciniphila and reduces diabetes incidence in the NOD mouse.
Diabetologia 55:2285–94
102. Jakobsson HE, Jernberg C, Andersson AF, Sjolund-Karlsson M, Jansson JK, Engstrand L. 2010. Short-
term antibiotic treatment has differing long-term impacts on the human throat and gut microbiome.
PLOS ONE 5:e9836
103. Imhann F, Bonder MJ, Vich Vila A, Fu J, Mujagic Z, et al. 2016. Proton pump inhibitors affect the gut
microbiome. Gut 65:740–48
104. Maier L, Pruteanu M, Kuhn M, Zeller G, Telzerow A, et al. 2018. Extensive impact of non-antibiotic
drugs on human gut bacteria. Nature 555:623–28

102 Induri et al.


105. Janarthanan S, Ditah I, Adler DG, Ehrinpreis MN. 2012. Clostridium difficile-associated diarrhea and
proton pump inhibitor therapy: a meta-analysis. Am. J. Gastroenterol. 107:1001–10
106. Wu H, Esteve E, Tremaroli V, Khan MT, Caesar R, et al. 2017. Metformin alters the gut microbiome
of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug.
Nat. Med. 23:850–58
107. Forslund K, Hildebrand F, Nielsen T, Falony G, Le Chatelier E, et al. 2015. Disentangling type 2
diabetes and metformin treatment signatures in the human gut microbiota. Nature 528:262–66
108. Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, et al. 2010. Delivery mode
shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns.
PNAS 107:11971–75
109. Huang S, Haiminen N, Carrieri A-P, Hu R, Jiang L, et al. 2020. Human skin, oral, and gut microbiomes
predict chronological age. mSystems 5:e00630-19
110. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, et al. 2012. Human gut micro-
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

biome viewed across age and geography. Nature 486:222–27


111. Maynard C, Weinkove D. 2018. The gut microbiota and ageing. In Biochemistry and Cell Biology of Ageing:
Part I Biomedical Science, ed. JR Harris, VI Korolchuk, pp. 351–71. Singapore: Springer
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

112. Kennedy BK, Berger SL, Brunet A, Campisi J, Cuervo AM, et al. 2014. Geroscience: linking aging to
chronic disease. Cell 159:709–13
113. Elyahu Y, Hekselman I, Eizenberg-Magar I, Berner O, Strominger I, et al. 2019. Aging promotes re-
organization of the CD4 T cell landscape toward extreme regulatory and effector phenotypes. Sci. Adv.
5:eaaw8330
114. Fessler J, Ficjan A, Duftner C, Dejaco C. 2013. The impact of aging on regulatory T-cells. Front. Immunol.
4:231
115. Goronzy JJ, Fang F, Cavanagh MM, Qi Q, Weyand CM. 2015. Naive T cell maintenance and function
in human aging. J. Immunol. 194:4073–80
116. Haynes L, Maue AC. 2009. Effects of aging on T cell function. Curr. Opin. Immunol. 21:414–17
117. Lee N, Kim W-U. 2017. Microbiota in T-cell homeostasis and inflammatory diseases. Exp. Mol. Med.
49:e340
118. Toor D, Wasson MK, Kumar P, Karthikeyan G, Kaushik NK, et al. 2019. Dysbiosis disrupts gut immune
homeostasis and promotes gastric diseases. Int. J. Mol. Sci. 20:2432
119. Megias J, Yanez A, Moriano S, O’Connor JE, Gozalbo D, Gil ML. 2012. Direct Toll-like receptor-
mediated stimulation of hematopoietic stem and progenitor cells occurs in vivo and promotes differen-
tiation toward macrophages. Stem Cells 30:1486–95
120. Mitroulis I, Kalafati L, Hajishengallis G, Chavakis T. 2018. Myelopoiesis in the context of innate im-
munity. J. Innate Immun. 10:365–72
121. Yanez A, Goodridge HS, Gozalbo D, Gil ML. 2013. TLRs control hematopoiesis during infection. Eur.
J. Immunol. 43:2526–33
122. Shintouo CM, Mets T, Beckwee D, Bautmans I, Ghogomu SM, et al. 2020. Is inflammageing influenced
by the microbiota in the aged gut? A systematic review. Exp. Gerontol. 141:111079
123. Claesson MJ, Jeffery IB, Conde S, Power SE, O’Connor EM, et al. 2012. Gut microbiota composition
correlates with diet and health in the elderly. Nature 488:178–84
124. Kumar A, Wu H, Collier-Hyams LS, Hansen JM, Li T, et al. 2007. Commensal bacteria modulate
cullin-dependent signaling via generation of reactive oxygen species. EMBO J. 26:4457–66
125. Pamer EG. 2007. Immune responses to commensal and environmental microbes. Nat. Immunol. 8:1173–
78
126. Kelly CP, Pothoulakis C, Lamont JT. 1994. Clostridium difficile colitis. N. Engl. J. Med. 330:257–62
127. Toward R, Montandon S, Walton G, Gibson GR. 2012. Effect of prebiotics on the human gut microbiota
of elderly persons. Gut Microbes 3:57–60
128. Weaver LK, Minichino D, Biswas C, Chu N, Lee J-J, et al. 2019. Microbiota-dependent signals are
required to sustain TLR-mediated immune responses. JCI Insight 4:e124370
129. Gorjifard S, Goldszmid RS. 2016. Microbiota–myeloid cell crosstalk beyond the gut. J. Leukoc. Biol.
100:865–79

www.annualreviews.org • Gut Microbiome, Metformin, and Aging 103


130. Kovtonyuk LV, Fritsch K, Feng X, Manz MG, Takizawa H. 2016. Inflamm-aging of hematopoiesis,
hematopoietic stem cells, and the bone marrow microenvironment. Front. Immunol. 7:502
131. Clements SJ, Carding SR. 2018. Diet, the intestinal microbiota, and immune health in aging. Crit. Rev.
Food Sci. Nutr. 58:651–61
132. Durand A, Audemard-Verger A, Guichard V, Mattiuz R, Delpoux A, et al. 2018. Profiling the lymphoid-
resident T cell pool reveals modulation by age and microbiota. Nat. Commun. 9:68
133. Gadecka A, Bielak-Zmijewska A. 2019. Slowing down ageing: the role of nutrients and microbiota in
modulation of the epigenome. Nutrients 11:1251
134. Kim S, Jazwinski SM. 2018. The gut microbiota and healthy aging: a mini-review. Gerontology 64:513–
20
135. Macaulay R, Akbar AN, Henson SM. 2013. The role of the T cell in age-related inflammation. Age
35:563–72
136. Peniche AG, Spinler JK, Boonma P, Savidge TC, Dann SM. 2018. Aging impairs protective host defenses
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

against Clostridioides (Clostridium) difficile infection in mice by suppressing neutrophil and IL-22 mediated
immunity. Anaerobe 54:83–91
137. Smith CK, Trinchieri G. 2018. The interplay between neutrophils and microbiota in cancer. J. Leukoc.
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

Biol. 104:701–15
138. Stavropoulou E, Bezirtzoglou E. 2019. Human microbiota in aging and infection: a review. Crit. Rev.
Food Sci. Nutr. 59:537–45
139. Bailey CJ. 2017. Metformin: historical overview. Diabetologia 60:1566–76
140. Bailey C, Day C. 2004. Metformin: its botanical background. Pract. Diabetes Int. 21:115–17
141. Wang YW, He SJ, Feng X, Cheng J, Luo YT, et al. 2017. Metformin: a review of its potential indications.
Drug Des. Devel. Ther. 11:2421–29
142. Moon MK, Hur KY, Ko SH, Park SO, Lee BW, et al. 2017. Combination therapy of oral hypoglycemic
agents in patients with type 2 diabetes mellitus. Diabetes Metab. J. 41:357–66
143. Wagstaff AJ, Figgitt DP. 2004. Extended-release metformin hydrochloride: single-composition osmotic
tablet formulation. Treat. Endocrinol. 3:327–32
144. Wang GS, Hoyte C. 2019. Review of biguanide (metformin) toxicity. J. Intensive Care Med. 34:863–
76
145. Johnson HK, Waterhouse C. 1968. Lactic acidosis and phenformin: report of two successfully treated
patients. Arch. Intern. Med. 122:367–70
146. Ball S, Woods HF, Alberti KGMM. 1974. Lacticacidosis, ketoacidosis, and hyperalaninaemia in a
phenformin-treated diabetic patient. Br. Med. J. 4:699–700
147. Kruse JA. 2001. Metformin-associated lactic acidosis. J. Emerg. Med. 20:267–72
148. Vecchio S, Giampreti A, Petrolini VM, Lonati D, Protti A, et al. 2014. Metformin accumulation: lactic
acidosis and high plasmatic metformin levels in a retrospective case series of 66 patients on chronic
therapy. Clin. Toxicol. 52:129–35
149. Viollet B, Guigas B, Sanz Garcia N, Leclerc J, Foretz M, Andreelli F. 2012. Cellular and molecular
mechanisms of metformin: an overview. Clin. Sci. 122:253–70
150. Duong JK, Kumar SS, Kirkpatrick CM, Greenup LC, Arora M, et al. 2013. Population pharmacokinetics
of metformin in healthy subjects and patients with type 2 diabetes mellitus: simulation of doses according
to renal function. Clin. Pharmacokinet. 52:373–84
151. Kajbaf F, Lalau JD. 2013. The prognostic value of blood pH and lactate and metformin concentrations
in severe metformin-associated lactic acidosis. BMC Pharmacol. Toxicol. 14:22
152. Pasquel FJ, Klein R, Adigweme A, Hinedi Z, Coralli R, et al. 2015. Metformin-associated lactic acidosis.
Am. J. Med. Sci. 349:263–67
153. Bell S, Farran B, McGurnaghan S, McCrimmon RJ, Leese GP, et al. 2017. Risk of acute kidney injury
and survival in patients treated with metformin: an observational cohort study. BMC Nephrol. 18:163
154. Corchia A, Wynckel A, Journet J, Moussi Frances J, Skandrani N, et al. 2020. Metformin-related lac-
tic acidosis with acute kidney injury: results of a French observational multicenter study. Clin. Toxicol.
58:375–82

104 Induri et al.


155. Dunn CJ, Peters DH. 1995. Metformin. A review of its pharmacological properties and therapeutic use
in non-insulin-dependent diabetes mellitus. Drugs 49:721–49
156. Svare A. 2009. A patient presenting with symptomatic hypomagnesemia caused by metformin-induced
diarrhoea: a case report. Cases J. 2:156
157. Babich MM, Pike I, Shiffman ML. 1998. Metformin-induced acute hepatitis. Am. J. Med. 104:490–92
158. Nammour FE, Fayad NF, Peikin SR. 2003. Metformin-induced cholestatic hepatitis. Endocr. Pract.
9:307–9
159. Cone CJ, Bachyrycz AM, Murata GH. 2010. Hepatotoxicity associated with metformin therapy in treat-
ment of type 2 diabetes mellitus with nonalcoholic fatty liver disease. Ann. Pharmacother. 44:1655–59
160. Miralles-Linares F, Puerta-Fernandez S, Bernal-Lopez MR, Tinahones FJ, Andrade RJ, Gomez-
Huelgas R. 2012. Metformin-induced hepatotoxicity. Diabetes Care 35:e21
161. Callaghan TS, Hadden DR, Tomkin GH. 1980. Megaloblastic anaemia due to vitamin B12 malabsorp-
tion associated with long-term metformin treatment. Br. Med. J. 280:1214–15
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

162. Bauman WA, Shaw S, Jayatilleke E, Spungen AM, Herbert V. 2000. Increased intake of calcium reverses
vitamin B12 malabsorption induced by metformin. Diabetes Care 23:1227–31
163. Mazokopakis EE, Starakis IK. 2012. Recommendations for diagnosis and management of metformin-
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

induced vitamin B12 (Cbl) deficiency. Diabetes Res. Clin. Pract. 97:359–67
164. Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. 2013. The hallmarks of aging. Cell
153:1194–217
165. Moskalev AA, Shaposhnikov MV, Plyusnina EN, Zhavoronkov A, Budovsky A, et al. 2013. The role of
DNA damage and repair in aging through the prism of Koch-like criteria. Ageing Res. Rev. 12:661–84
166. Dogan Turacli I, Candar T, Yuksel BE, Demirtas S. 2018. Role of metformin on base excision repair
pathway in p53 wild-type H2009 and HepG2 cancer cells. Hum. Exp. Toxicol. 37:909–19
167. Szewczuk M, Boguszewska K, Kazmierczak-Baranska J, Karwowski BT. 2020. The role of AMPK in
metabolism and its influence on DNA damage repair. Mol. Biol. Rep. 47:9075–86
168. Dogan Turacli I, Candar T, Yuksel EB, Kalay S, Oguz AK, Demirtas S. 2018. Potential effects of met-
formin in DNA BER system based on oxidative status in type 2 diabetes. Biochimie 154:62–68
169. Jin C, Li J, Green CD, Yu X, Tang X, et al. 2011. Histone demethylase UTX-1 regulates C. elegans life
span by targeting the insulin/IGF-1 signaling pathway. Cell Metab. 14:161–72
170. Bridgeman SC, Ellison GC, Melton PE, Newsholme P, Mamotte CDS. 2018. Epigenetic effects of
metformin: from molecular mechanisms to clinical implications. Diabetes Obes. Metab. 20:1553–62
171. Cuyàs E, Verdura S, Llorach-Pares L, Fernández-Arroyo S, Luciano-Mateo F, et al. 2018. Metformin
directly targets the H3K27me3 demethylase KDM6A/UTX. Aging Cell 17:e12772
172. Yu T, Wang C, Yang J, Guo Y, Wu Y, Li X. 2017. Metformin inhibits SUV39H1-mediated migration
of prostate cancer cells. Oncogenesis 6:e324
173. Chen J, Ou Y, Li Y, Hu S, Shao LW, Liu Y. 2017. Metformin extends C. elegans lifespan through lyso-
somal pathway. eLife 6:e31268
174. Crawley D, Chandra A, Loda M, Gillett C, Cathcart P, et al. 2017. Metformin and longevity (METAL):
a window of opportunity study investigating the biological effects of metformin in localised prostate
cancer. BMC Cancer 17:494
175. Csaba G. 2019. Immunity and longevity. Acta Microbiol. Immunol. Hung. 66:1–17
176. Fang J, Yang J, Wu X, Zhang G, Li T, et al. 2018. Metformin alleviates human cellular aging by upreg-
ulating the endoplasmic reticulum glutathione peroxidase 7. Aging Cell 17:e12765
177. Garg G, Singh S, Singh AK, Rizvi SI. 2017. Metformin alleviates altered erythrocyte redox status during
aging in rats. Rejuvenation Res. 20:15–24
178. Sica A, Guarneri V, Gennari A. 2019. Myelopoiesis, metabolism and therapy: a crucial crossroads in
cancer progression. Cell Stress 3:284–94
179. Cai H, Han B, Hu Y, Zhao X, He Z, et al. 2020. Metformin attenuates the D–galactose–induced aging
process via the UPR through the AMPK/ERK1/2 signaling pathways. Int. J. Mol. Med. 45:715–30
180. Fontana L, Partridge L, Longo VD. 2010. Extending healthy life span—from yeast to humans. Science
328:321–26
181. Schumacher B, van der Pluijm I, Moorhouse MJ, Kosteas T, Robinson AR, et al. 2008. Delayed and
accelerated aging share common longevity assurance mechanisms. PLOS Genet. 4:e1000161

www.annualreviews.org • Gut Microbiome, Metformin, and Aging 105


182. Garinis GA, van der Horst GT, Vijg J, Hoeijmakers JH. 2008. DNA damage and ageing: new-age ideas
for an age-old problem. Nat. Cell Biol. 10:1241–47
183. Xie Y, Wang JL, Ji M, Yuan ZF, Peng Z, et al. 2014. Regulation of insulin-like growth factor signaling
by metformin in endometrial cancer cells. Oncol. Lett. 8:1993–99
184. Klement RJ, Fink MK. 2016. Dietary and pharmacological modification of the insulin/IGF-1 system:
exploiting the full repertoire against cancer. Oncogenesis 5:e193
185. Amin S, Lux A, O’Callaghan F. 2019. The journey of metformin from glycaemic control to mTOR
inhibition and the suppression of tumour growth. Br. J. Clin. Pharmacol. 85:37–46
186. Cuyas E, Verdura S, Llorach L, Fernández Arroyo S, Joven J, et al. 2018. Metformin is a direct SIRT1-
activating compound: computational modeling and experimental validation. Front. Endocrinol. 9:657
187. Adeyemo MA, McDuffie JR, Kozlosky M, Krakoff J, Calis KA, et al. 2015. Effects of metformin on
energy intake and satiety in obese children. Diabetes Obes. Metab. 17:363–70
188. Mair W, Dillin A. 2008. Aging and survival: the genetics of life span extension by dietary restriction.
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

Annu. Rev. Biochem. 77:727–54


189. Onken B, Driscoll M. 2010. Metformin induces a dietary restriction-like state and the oxidative stress
response to extend C. elegans healthspan via AMPK, LKB1, and SKN-1. PLOS ONE 5:e8758
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

190. Bridges HR, Jones AJ, Pollak MN, Hirst J. 2014. Effects of metformin and other biguanides on oxidative
phosphorylation in mitochondria. Biochem. J. 462:475–87
191. El-Mir MY, Nogueira V, Fontaine E, Avéret N, Rigoulet M, Leverve X. 2000. Dimethylbiguanide in-
hibits cell respiration via an indirect effect targeted on the respiratory chain complex I. J. Biol. Chem.
275:223–28
192. Hardie DG, Ross FA, Hawley SA. 2012. AMPK: a nutrient and energy sensor that maintains energy
homeostasis. Nat. Rev. Mol. Cell Biol. 13:251–62
193. Zhou G, Myers R, Li Y, Chen Y, Shen X, et al. 2001. Role of AMP-activated protein kinase in mechanism
of metformin action. J. Clin. Investig. 108:1167–74
194. Madiraju AK, Erion DM, Rahimi Y, Zhang XM, Braddock DT, et al. 2014. Metformin suppresses glu-
coneogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase. Nature 510:542–46
195. Li X, Guo Y, Yan W, Snyder MP, Li X. 2015. Metformin improves diabetic bone health by re-balancing
catabolism and nitrogen disposal. PLOS ONE 10:e0146152
196. Guo Y, Cho SW, Saxena D, Li X. 2020. Multifaceted actions of succinate as a signaling transmitter vary
with its cellular locations. Endocrinol. Metab. 35:36–43
197. Kuilman T, Michaloglou C, Mooi WJ, Peeper DS. 2010. The essence of senescence. Genes Dev. 24:2463–
79
198. Moiseeva O, Deschênes-Simard X, St-Germain E, Igelmann S, Huot G, et al. 2013. Metformin inhibits
the senescence-associated secretory phenotype by interfering with IKK/NF-κB activation. Aging Cell
12:489–98
199. Noren Hooten N, Martin-Montalvo A, Dluzen DF, Zhang Y, Bernier M, et al. 2016. Metformin-
mediated increase in DICER1 regulates microRNA expression and cellular senescence. Aging Cell
15:572–81
200. Kuang Y, Hu B, Feng G, Xiang M, Deng Y, et al. 2020. Metformin prevents against oxidative stress-
induced senescence in human periodontal ligament cells. Biogerontology 21:13–27
201. Hu Q, Peng J, Jiang L, Li W, Su Q, et al. 2020. Metformin as a senostatic drug enhances the anticancer
efficacy of CDK4/6 inhibitor in head and neck squamous cell carcinoma. Cell Death Dis. 11:925
202. Yi G, He Z, Zhou X, Xian L, Yuan T, et al. 2013. Low concentration of metformin induces a p53-
dependent senescence in hepatoma cells via activation of the AMPK pathway. Int. J. Oncol. 43:1503–10
203. Na HJ, Pyo JH, Jeon HJ, Park JS, Chung HY, Yoo MA. 2018. Deficiency of Atg6 impairs beneficial
effect of metformin on intestinal stem cell aging in Drosophila. Biochem. Biophys. Res. Commun. 498:18–24
204. Fatt M, Hsu K, He L, Wondisford F, Miller FD, et al. 2015. Metformin acts on two different molecular
pathways to enhance adult neural precursor proliferation/self-renewal and differentiation. Stem Cell Rep.
5:988–95
205. Neumann B, Baror R, Zhao C, Segel M, Dietmann S, et al. 2019. Metformin restores CNS remyelination
capacity by rejuvenating aged stem cells. Cell Stem Cell 25:473–85.e8

106 Induri et al.


206. Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca M, et al. 2000. Inflamm-aging: an evolutionary
perspective on immunosenescence. Ann. N. Y. Acad. Sci. 908:244–54
207. Franceschi C, Campisi J. 2014. Chronic inflammation (inflammaging) and its potential contribution to
age-associated diseases. J. Gerontol. A Biol. Sci. Med. Sci. 69(Suppl. 1):S4–9
208. Bauer PV, Duca FA, Waise TMZ, Rasmussen BA, Abraham MA, et al. 2018. Metformin alters upper
small intestinal microbiota that impact a glucose-SGLT1-sensing glucoregulatory pathway. Cell Metab.
27:101–17.e5
209. Sun L, Xie C, Wang G, Wu Y, Wu Q, et al. 2018. Gut microbiota and intestinal FXR mediate the clinical
benefits of metformin. Nat. Med. 24:1919–29
210. Armanios M, Blackburn EH. 2012. The telomere syndromes. Nat. Rev. Genet. 13:693–704
211. Tomás-Loba A, Flores I, Fernández-Marcos PJ, Cayuela ML, Maraver A, et al. 2008. Telomerase reverse
transcriptase delays aging in cancer-resistant mice. Cell 135:609–22
212. Kulkarni AS, Gubbi S, Barzilai N. 2020. Benefits of metformin in attenuating the hallmarks of aging.
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

Cell Metab. 32:15–30


213. Liu J, Ge Y, Wu S, Ma D, Xu W, et al. 2019. Association between antidiabetic agents use and leukocyte
telomere shortening rates in patients with type 2 diabetes. Aging 11:741–55
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

214. Shin NR, Lee JC, Lee HY, Kim MS, Whon TW, et al. 2014. An increase in the Akkermansia spp. popu-
lation induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut
63:727–35
215. Vallianou NG, Stratigou T, Tsagarakis S. 2019. Metformin and gut microbiota: their interactions and
their impact on diabetes. Hormones 18:141–44
216. Check Hayden E. 2015. Anti-ageing pill pushed as bona fide drug. Nature 522:265–66
217. Newman JC, Milman S, Hashmi SK, Austad SN, Kirkland JL, et al. 2016. Strategies and challenges in
clinical trials targeting human aging. J. Gerontol. A Biol. Sci. Med. Sci. 71:1424–34
218. Soukas AA, Hao H, Wu L. 2019. Metformin as anti-aging therapy: Is it for everyone? Trends Endocrinol.
Metab. 30:745–55
219. Pedersen HK, Gudmundsdottir V, Nielsen HB, Hyotylainen T, Nielsen T, et al. 2016. Human gut
microbes impact host serum metabolome and insulin sensitivity. Nature 535:376–81
220. Perry RJ, Peng L, Barry NA, Cline GW, Zhang D, et al. 2016. Acetate mediates a microbiome-brain-
β-cell axis to promote metabolic syndrome. Nature 534:213–17
221. Forslund K, Hildebrand F, Nielsen T, Falony G, Le Chatelier E, et al. 2015. Disentangling type 2
diabetes and metformin treatment signatures in the human gut microbiota. Nature 528:262–66
222. de la Cuesta-Zuluaga J, Mueller NT, Corrales-Agudelo V, Velasquez-Mejia EP, Carmona JA, et al. 2017.
Metformin is associated with higher relative abundance of mucin-degrading Akkermansia muciniphila and
several short-chain fatty acid-producing microbiota in the gut. Diabetes Care 40:54–62
223. Geerlings SY, Kostopoulos I, de Vos WM, Belzer C. 2018. Akkermansia muciniphila in the human gas-
trointestinal tract: when, where, and how? Microorganisms 6:75
224. Lee H, Ko G. 2014. Effect of metformin on metabolic improvement and gut microbiota. Appl. Environ.
Microbiol. 80:5935–43
225. Montandon SA, Jornayvaz FR. 2017. Effects of antidiabetic drugs on gut microbiota composition. Genes
8:250
226. Zhou ZY, Ren LW, Zhan P, Yang HY, Chai DD, Yu ZW. 2016. Metformin exerts glucose-lowering
action in high-fat fed mice via attenuating endotoxemia and enhancing insulin signaling. Acta Pharmacol.
Sin. 37:1063–75
227. Fernandes A, Vaz AR, Falcao AS, Silva RF, Brito MA, Brites D. 2007. Glycoursodeoxycholic acid and
interleukin-10 modulate the reactivity of rat cortical astrocytes to unconjugated bilirubin. J. Neuropathol.
Exp. Neurol. 66:789–98
228. Cabreiro F, Au C, Leung KY, Vergara-Irigaray N, Cochemé HM, et al. 2013. Metformin retards aging
in C. elegans by altering microbial folate and methionine metabolism. Cell 153:228–39
229. Nijhout HF, Reed MC, Budu P, Ulrich CM. 2004. A mathematical model of the folate cycle: new insights
into folate homeostasis. J. Biol. Chem. 279:55008–16
230. Garigan D, Hsu AL, Fraser AG, Kamath RS, Ahringer J, Kenyon C. 2002. Genetic analysis of tissue aging
in Caenorhabditis elegans: a role for heat-shock factor and bacterial proliferation. Genetics 161:1101–12

www.annualreviews.org • Gut Microbiome, Metformin, and Aging 107


231. Pryor R, Norvaisas P, Marinos G, Best L, Thingholm LB, et al. 2019. Host-microbe-drug-nutrient
screen identifies bacterial effectors of metformin therapy. Cell 178:1299–312.e29
232. Barzilai N, Crandall JP, Kritchevsky SB, Espeland MA. 2016. Metformin as a tool to target aging. Cell
Metab. 23:1060–65
233. Barzilai N, Huffman DM, Muzumdar RH, Bartke A. 2012. The critical role of metabolic pathways in
aging. Diabetes 61:1315–22
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

108 Induri et al.


PA62_Front_Matter ARjats.cls November 11, 2021 17:25

Annual Review of
Pharmacology and
Toxicology
Contents Volume 62, 2022

Pushing Forward the Future Tense: Perspectives of a Scientist


Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

Lee E. Limbird p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 1
Introduction to the Theme “New Insights, Strategies, and
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

Therapeutics for Common Diseases”


Paul A. Insel, Terrence F. Blaschke, Susan G. Amara, and Urs A. Meyer p p p p p p p p p p p p p p p p p p19
Experimental Models of SARS-CoV-2 Infection: Possible Platforms to
Study COVID-19 Pathogenesis and Potential Treatments
Sareh Pandamooz, Benjamin Jurek, Carl-Philipp Meinung, Zahra Baharvand,
Alireza Sahebi Shahem-abadi, Silke Haerteis, Jaleel A. Miyan, James Downing,
Mehdi Dianatpour, Afshin Borhani-Haghighi, and Mohammad Saied Salehi p p p p p p p p p25
Central Nervous System Control of Glucose Homeostasis:
A Therapeutic Target for Type 2 Diabetes?
Zaman Mirzadeh, Chelsea L. Faber, and Michael W. Schwartz p p p p p p p p p p p p p p p p p p p p p p p p p p p p55
The Gut Microbiome, Metformin, and Aging
Sri Nitya Reddy Induri, Payalben Kansara, Scott C. Thomas, Fangxi Xu,
Deepak Saxena, and Xin Li p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p85
Sodium-Glucose Cotransporter 2 Inhibitors in Heart Failure
Kevin S. Shah and James C. Fang p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 109
Repurposing Colchicine for Heart Disease
Nadia Bouabdallaoui and Jean-Claude Tardif p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 121
A New Old Target: Androgen Receptor Signaling and Advanced
Prostate Cancer
Daniel Westaby, Maria de los Dolores Fenor de La Maza, Alec Paschalis,
Juan M. Jimenez-Vacas, Jon Welti, Johann de Bono, and Adam Sharp p p p p p p p p p p p p p p 131
Synthetic Retinoids Beyond Cancer Therapy
Lorraine J. Gudas p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 155
Thioredoxin Reductase Inhibition for Cancer Therapy
Radosveta Gencheva and Elias S.J. Arnér p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 177

v
PA62_Front_Matter ARjats.cls November 11, 2021 17:25

Emerging Therapeutics, Technologies, and Drug Development


Strategies to Address Patient Nonadherence and Improve
Tuberculosis Treatment
Maria Garcia-Cremades, Belen P. Solans, Natasha Strydom, Bernard Vrijens,
Goonaseelan Colin Pillai, Craig Shaffer, Bruce Thomas, and Rada M. Savic p p p p p p p p p 197
Prenatal and Postnatal Pharmacotherapy in Down Syndrome: The
Search to Prevent or Ameliorate Neurodevelopmental and
Neurodegenerative Disorders
Renata Bartesaghi, Stefano Vicari, and William C. Mobley p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 211
Noncanonical Metabotropic Glutamate Receptor 5 Signaling in
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

Alzheimer’s Disease
Khaled S. Abd-Elrahman and Stephen S.G. Ferguson p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 235
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

Brain-Protective Mechanisms of Transcription Factor NRF2: Toward a


Common Strategy for Neurodegenerative Diseases
Antonio Cuadrado p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 255
Targeting NRF2 and Its Downstream Processes: Opportunities and
Challenges
Laura Torrente and Gina M. DeNicola p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 279
E-Cigarette Toxicology
Terry Gordon, Emma Karey, Meghan E. Rebuli, Yael-Natalie H. Escobar,
Ilona Jaspers, and Lung Chi Chen p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 301
Thirty Years of Neuroscientific Investigation of Placebo and Nocebo:
The Interesting, the Good, and the Bad
Fabrizio Benedetti, Elisa Frisaldi, and Aziz Shaibani p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 323
Patient Centricity Driving Formulation Innovation: Improvements in
Patient Care Facilitated by Novel Therapeutics and Drug Delivery
Technologies
Susanne Page, Tarik Khan, Peter Kühl, Gregoire Schwach, Kirsten Storch,
and Hitesh Chokshi p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 341
Fragile X Syndrome: Lessons Learned and What New Treatment
Avenues Are on the Horizon
Randi J. Hagerman and Paul J. Hagerman p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 365
Aryl Hydrocarbon Receptor and Its Diverse Ligands and Functions:
An Exposome Receptor
Lucie Larigot, Louise Benoit, Meriem Koual, Céline Tomkiewicz, Robert Barouki,
and Xavier Coumoul p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 383
Non-P450 Drug-Metabolizing Enzymes: Contribution to Drug
Disposition, Toxicity, and Development
Tatsuki Fukami, Tsuyoshi Yokoi, and Miki Nakajima p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 405

vi Contents
PA62_Front_Matter ARjats.cls November 11, 2021 17:25

Pharmacology of TRPC Channels and Its Potential in Cardiovascular


and Metabolic Medicine
Robin S. Bon, David J. Wright, David J. Beech, and Piruthivi Sukumar p p p p p p p p p p p p p p p 427
KCNQ Potassium Channels as Targets of Botanical Folk Medicines
Kaitlyn E. Redford and Geoffrey W. Abbott p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 447
Drug Target Identification in Tissues by Thermal Proteome Profiling
André Mateus, Nils Kurzawa, Jessica Perrin, Giovanna Bergamini,
and Mikhail M. Savitski p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 465
Endocannabinoid-Based Therapies
Daniele Piomelli and Alex Mabou Tagne p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 483
Annu. Rev. Pharmacol. Toxicol. 2022.62:85-108. Downloaded from www.annualreviews.org

HLA Allele–Restricted Immune-Mediated Adverse Drug Reactions:


Framework for Genetic Prediction
Access provided by Afghanistan - R4L on 04/01/22. For personal use only.

Kanoot Jaruthamsophon, Paul J. Thomson, Chonlaphat Sukasem, Dean J. Naisbitt,


and Munir Pirmohamed p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 509
Measuring Pharmacogene Variant Function at Scale Using
Multiplexed Assays
Renee C. Geck, Gabriel Boyle, Clara J. Amorosi, Douglas M. Fowler,
and Maitreya J. Dunham p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 531
Chemogenetic Approaches to Probe Redox Pathways: Implications for
Cardiovascular Pharmacology and Toxicology
Benjamin Steinhorn, Emrah Eroglu, and Thomas Michel p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 551
Endocrine-Disrupting Chemicals and Child Health
Akhgar Ghassabian, Laura Vandenberg, Kurunthachalam Kannan,
and Leonardo Trasande p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 573
Systems Biology of the Vasopressin V2 Receptor: New Tools for
Discovery of Molecular Actions of a GPCR
Lihe Chen, Hyun Jun Jung, Arnab Datta, Euijung Park, Brian G. Poll,
Hiroaki Kikuchi, Kirby T. Leo, Yash Mehta, Spencer Lewis, Syed J. Khundmiri,
Shaza Khan, Chung-Lin Chou, Viswanathan Raghuram, Chin-Rang Yang,
and Mark A. Knepper p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 595
Oxidative Stress and Metabolism: A Mechanistic Insight for
Glyphosate Toxicology
Xiaojing Wang, Qirong Lu, Jingchao Guo, Irma Ares, Marta Martínez,
María-Rosa Martínez-Larrañaga, Xu Wang, Arturo Anadón,
and María-Aránzazu Martínez p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 617
Precision Medicine Approaches for Infantile-Onset Developmental
and Epileptic Encephalopathies
Kenneth A. Myers and Ingrid E. Scheffer p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 641

Contents vii

You might also like