ArticlePDF Available

Crosstalk between gut microbiota and RNA N6-methyladenosine modification in cancer

Authors:

Abstract and Figures

The gut microbiota plays a crucial role in regulating various host metabolic, immune, and neuroendocrine functions, and has a significant impact on human health. Several lines of evidence suggest that gut dysbiosis is associated with a variety of diseases, including cancer. The gut microbiota can impact the development and progression of cancer through a range of mechanisms, such as regulating cell proliferation and death, modulating the host immune response, and altering the host metabolic state. Gene regulatory programs are considered critical mediators between the gut microbiota and host phenotype, of which RNA N6-methyladenosine (m6A) modifications have attracted much attention recently. Aberrant m6A modifications have been shown to play a crucial role in cancer development. This review aims to provide an overview of the diverse roles of gut microbiota and RNA m6A modifications in cancer and highlight their potential interactions in cancer development.
Content may be subject to copyright.
FEMS Microbiology Reviews , 2023, 47 , 1–15
DOI: 10.1093/femsre/fuad036
Ad v ance access publication date: 5 July 2023
Re vie w Article
Crosstalk between gut microbiota and RNA
N6-methyladenosine modication in cancer
Hao Su
1 ,2
, Henley Cheung
1
, Harry Cheuk-Hay Lau
1
, Hongyan Chen
1 ,2
, Xiaoting Zhang
1 ,2
, Na Qin
1 ,2
, Yifei Wang
3
, Matthew Tak
Vai Chan
2
, William Ka Kei Wu
1 ,2
, Huarong Chen
1 ,2 ,*
1
Institute of Digestive Disease and Department of Medicine and Ther a peutics, State K ey labor atory of Digestiv e Disease , Li Ka Shing Institute of Health Sciences ,
CUHK Shenzhen Research Institute , T he Chinese University of Hong Kong , Hong Kon g 99077, China
2
Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute , T he Chinese University of Hong Kong, Hong Ko ng 99077, China
3
School of Biomedical Sciences , T he Chinese University of Hong Kong, Hong Kong 99077, China
Corresponding author. Department of Anaesthesia and Intensive Care , T he Chinese University of Hong Kong, Shatin, N.T., Hong Kong 99077, China. E-mail:
hchen2@cuhk.edu.hk
Editor: [Dennis Nielsen]
Abstract
The gut microbiota plays a crucial role in regulating various host metabolic, immune, and neuroendocrine functions, and has a signif-
icant impact on human health. Several lines of evidence suggest that gut dysbiosis is associated with a variety of diseases, including
cancer. The gut microbiota can impact the development and pr ogr ession of cancer through a range of mechanisms, such as reg-
ulating cell proliferation and death, modulating the host imm une r esponse, and altering the host metabolic state. Gene regulatory
pr ograms ar e consider ed critical mediators between the gut microbiota and host phenotype, of which RNA N6-methyladenosine (m6A)
modications have attracted much attention recently. Aberrant m6A modications have been shown to play a crucial role in cancer
dev elopment. This r eview aims to provide an overview of the diverse roles of gut microbiota and RNA m6A modications in cancer
and highlight their potential interactions in cancer development.
Ke yw ords: cancer, microbiota, N6-methyladenosine, crosstalk
Introduction
Ongoing efforts are being made to enhance our understanding of
the etiology of cancer. Cancer de v elopment is now regarded as a
m ultifaceted pr ocess inuenced by v arious risk factors, suc h as
genetic (e.g. inherited mutations) and environmental factors (e.g.
diet and infections), which cooperatively contribute to the inci-
dence and pr ogr ession of cancer. In the past two decades, gut mi-
cr obiota has dr awn incr easing attention and is now recognized
as an additional environmental risk factor for cancer. In a 70-
kg adult man, it is estimated that there are 38 trillion microbial
cells, comprising a ppr oximatel y 0.3% of the total body weight,
with the majority residing within the colon (Sender et al . 2016 ). Pre-
clinical studies have provided compelling evidence that gut dys-
biosis signicantly impacts cancer development and progression
thr ough m ultiple mec hanisms, suc h as disturbing the balance be-
tween cell pr olifer ation and death, modifying the immune system,
and manipulating host metabolism (Garrett 2015 ). Conversely, the
presence of tumors can also alter the microbiome composition,
which, in turn, potentially contributes to tumorigenesis and can-
cer pr ogr ession (Zitvogel et al. 2017 , Goodman and Gardner 2018 ).
Preclinical studies conducted in cell lines or mouse models have
identied probiotic bacteria, such as Lacticaseibacillus casei , Strep-
tococcus thermophilus , and Bidobacterium longum , as having the po-
tential to pr e v ent cancer or suppr ess tumor gr owth (Konishi et al.
2016 , Singh et al . 1997 , Li et al. 2021c ). Conv ersel y, pathogenic mi-
cr obes suc h as Helicobacter pylori , P eptostreptococcus anaerobius , and
Fusobacterium nucleatum have been found to facilitate cancer de-
velopment (Moss 2017 , Long et al. 2019 , Wu et al. 2019a ).
Recent evidence suggests that dysregulated epitranscriptomic
mechanisms within cancer cells contribute to cancer develop-
ment. RNA modication is a crucial regulatory mechanism that
controls gene expression, with over 160 types of RNA mod-
ications identied to date. Among these modications, N6-
methyladenosine (m6A) is the most pr e v alent mRNA modica-
tion in eukaryotic cells (Boccaletto et al. 2018 ). The m6A epitran-
scriptomic system is dynamically regulated by a group of proteins
known as writers , erasers , and readers . T he m6A writers , which
include methyltr ansfer ase-like 3 (METTL3), METTL14, METTL16,
WT1-associated protein (WTAP), and other cofactors, catalyze the
m6A modications. In contrast, the m6A erasers, such as fat
mass and obesity-associated protein (FTO) and AlkB Homolog 5
(ALKBH5), r emov e m6A marks fr om RNA. Furthermor e, the m6A
readers, including YTH domain-containing proteins (YTHDF1-3
and YTHDC1-2) and insulin-like growth factor 2 mRNA binding
proteins 1–3 (IGF2BP1-3), specically recognize and bind to m6A
marks on target genes to trigger subsequent RNA metabolism
(Sun et al . 2019 , Satterwhite and Manseld 2022 ).
The microbiome has been shown to have the capacity to alter
host epigenetic modications, such as DNA methylation, histone
modication, and non-coding RNA expression (Tahara et al. 2014 ,
Liang et al. 2015 , Koh et al. 2016 , Woo and Alenghat 2017 ). While its
contribution to RNA m6A modication is still being investigated,
studies have shown that compared to the germ-free (GF) mice,
specic pathogen-free (SPF) mice exhibit different transcriptome-
wide m6A methylome proles in the brain, intestine, and liver
(Wang et al . 2019 ), suggesting that that the presence of microbiome
Recei v ed 22 August 2022; revised 23 June 2023; accepted 28 June 2023
©The Author(s) 2023. Published by Oxford Uni v ersity Pr ess on behalf of FEMS. All rights r eserv ed. For permissions, please e-mail:
journals.permissions@oup.com
Downloaded from https://academic.oup.com/femsre/article/47/4/fuad036/7220014 by The Chinese University of Hong Kong user on 26 July 2023
2 | FEMS Microbiology Reviews , 2023, Vo l. 47, No. 4
inuences host RNA m6A pr oles. Furthermor e, c hanges in gut
micr obiota hav e been associated with alter ed RNA m6A modica-
tions in the cecum and liver tissues in mice carrying conventional,
modied, or no microbiota (Jabs et al. 2020 ). Commensal bacteria
such as Akkermansia muciniphila and Lactiplantibacillus plantarum
have also been shown to inuence m6A modications of genes
involved in cell growth and death (Jabs et al . 2020 ). These ndings
suggest that the gut microbiota could potentially alter host RNA
m6A modications , which ma y ha ve implications for cancer de-
v elopment. This r e vie w details the r ole of gut micr obiota and m6A
modication in cancer and summarizes k e y ndings establishing
their potential connections.
Gut microbiota and cancer
The gut microbiota can exert its effects on host cells through
v arious mec hanisms, including dir ectl y binding thr ough surface
receptors and secretion of metabolites such as short-chain fatty
acids (SCFAs) like butyrate , acetate , and propionate , bacterial vir-
ulence factors such as cytotoxin-associated gene A (CagA) pro-
tein and colibactin, and proteins such as beta-Galactosidase (Dal-
masso et al. 2014 , Li et al. 2021c ). These factors can cooper ativ el y
regulate tumor growth and metastasis. In the following section,
we will discuss the dual roles of the gut microbiota in cancer
(Table 1 ).
Gut microbiota as tumor promoters
P athogens ar e micr oor ganisms that can cause disease, and in
some cases , they ha v e been implicated as driv ers of carcinogen-
esis. Recent studies suggest that pathogenic bacteria may be re-
sponsible for de v eloping cancer in an estimated 15%–20% of hu-
man cases (Bhatt et al . 2017 ). This highlights the importance of
understanding the potential role that these microorganisms may
play in cancer pathogenesis. Helicobacter pylori is a well-studied
and widely known pathogen, classied as a class 1 carcinogen
by the Wor ld Health Organization (WHO). It is responsible for
a ppr oximatel y 90% of noncardiac gastric cancer (GC) cases in
humans (Moss 2017 ). The Ca gA pr otein expr essed by Helicobac-
ter pylori can interact with the apoptosis-stimulating of p53 pro-
tein 2 (ASPP2) in host cells, leading to enhanced degradation of
p53 protein and thereby abrogating its tumor-suppressive func-
tion, whic h may incr ease cancer susceptibility (Buti et al . 2011 ). In
addition, Helicobacter pylori secrete tumor necrosis factor- α(TNF-
α)-inducing proteins (TNFAIPs) to promote TNF- αexpression in
host cells, which is known to be involved gastric carcinogene-
sis in humans (Suganuma et al . 2021 ). Compared to healthy mu-
cosa, tumor biopsies taken from patients with colorectal cancer
(CRC) show a higher abundance of Esc heric hia coli , with the ma-
jority of these being polyketide synthase (pks) + Esc heric hia coli
(Dalmasso et al. 2014 ). The pks + Esc heric hia coli str ain r eleases
colibactin, a genotoxic compound that promotes tumor growth
in a xenograft mouse model (Dalmasso et al. 2014 ). Propionibac-
terium acnes has been identied in human prostate cancer tissues
using cultur e methods, uor escence micr oscopy methods, and
DNA analysis. Cutibacterium acnes can induce cyclooxygenase-2
signaling, which enhances cancer cell pr olifer ation (Goodman and
Gardner 2018 ). Fusobacterium nucleatum is an oral bacterium that
is enriched in CRC. Based on studies using CRC cell lines or mouse
models, it has been shown that Fusobacterium nucleatum promotes
color ectal carcinogenesis thr ough a unique adhesin called FadA,
which binds to E-cadherin and activates β-catenin signaling in
the host cells (Rubinstein et al . 2013 ). Furthermore, Fusobacterium
nucleatum -expr essing Fa p2 can dir ectl y inter act with TIGIT, a coin-
hibitory r eceptor expr essed on natur al killer (NK) cells, to inhibit
the cytotoxicity of primary human NK cells (Gur et al . 2015 ). En-
terotoxigenic Bacteroides fragilis ( ETBF ) also promotes β-catenin
signaling in CRC cells by secreting a metalloprotease toxin, which
facilitates CRC cell growth (Wu et al . 2007 ). Moreover, ETBF can
induce colon tumorigenesis in mice by activating Th17 cell re-
sponses (Wu et al. 2009 ). Toxigenic Clostridioides difcile is another
potential driver of CRC. A recent study sho w ed that gavage of a hu-
man colon cancer-derived Clostridioides difcile strain that secretes
the cytotoxin TcdB into germ-free APC
min / +
mice facilitated CRC
tumorigenesis (Dr e wes et al . 2022 ). Collectiv el y, pathogenic bacte-
ria can promote the de v elopment of cancer through both direct
and indirect interactions with the host.
Gut microbiota as tumor suppressors
Pr obiotics ar e liv e micr oor ganisms that pr ovide health benets to
the host through various mechanisms . T he Lactobacillaceae fam-
ily of lactic acid bacteria is generally considered non-pathogenic
(Zheng et al . 2020 ). Ho w e v er, some species within this famil y,
such as Lacticaseibacillus rhamnosus, may cause r ar e infections in
critically ill patients (Hazards et al. 2017 ). Lacticaseibacillus and
Lactobacillus ar e gener a of bacteria commonl y found in the hu-
man digestive and urinary tracts. Several species and strains
of these bacteria may benet human health, such as reducing
the risk of certain illnesses. For example, Lacticaseibacillus casei is
commonly used in probiotic supplements to impr ov e gastr oin-
testinal function. Recent r esearc h has also found that Lactica-
seibacillus casei may induce cancer cell death by secreting fer-
ric hr ome metabolite, whic h activ ates the JNK pathway in cell
lines and mouse models (Konishi et al. 2016 ). Similarl y, pr eclin-
ical studies using cell lines and mouse models have suggested
that supernatants of Lacticaseibacillus rhamnosus , Lactobacillus aci-
dophilus , and Lactobacillus gallinarum can inhibit the pr olifer ation
of CRC cells and induce apoptosis (Dehghani et al. 2021 , Sug-
im ur a et al . 2021 , Yue et al . 2021 ). Specically, Lactobacillus galli-
narum has been found to secrete indole-3-lactic, which inhibits
the growth of CRC cells and patient-derived CRC organoids, as
well as suppresses intestinal tumorigenesis in Apc
Min / +
mice (Sug-
im ur a et al . 2021 ). Aside from these, Lacticaseibacillus casei has been
shown to induce an anti-tumor immune response in mice, as ev-
idenced by the activated dendritic cells (DCs) and CD8
+ T cells
(Takagi et al. 2008 ).
Bidobacteria is another group of probiotics that has been stud-
ied for its potential health benets. In mice, the administration of
Bidobacterium bidum was found to suppress AOM/DSS-induced
colorectal tumorigenesis by modifying the gut microbial compo-
sition and metabolome (Wang et al. 2020 ). In addition, oral gav-
age of Bidobacterium breve in mice was shown to increase the ac-
cumulation of dendritic cells (DCs) in the intestinal villi, which
may enhance anti-tumor immunity (Li et al. 2021b ). Akkerman-
sia muciniphila , a gr am-negativ e bacterium natur all y found in the
human gut, has been the subject of recent research as a poten-
tial next-generation probiotic with anti-tumor effects. A preclini-
cal study in mice found that Akkermansia muciniphila suppressed
prostate cancer cell proliferation and invasion by modulating the
immune system (Luo et al. 2021 ). In addition, studies have demon-
strated that the probiotic bacteria Propionibacterium acidipropionici
and Propionibacterium freudenreichii generate SCFAs, namely ac-
etate and propionate, that are effective in inducing apoptosis in
CRC cells (Jan et al . 2002 ). Together, these ndings suggest that
probiotics may have anti-tumor properties . Nevertheless , it is
Downloaded from https://academic.oup.com/femsre/article/47/4/fuad036/7220014 by The Chinese University of Hong Kong user on 26 July 2023
Su et al. | 3
Tab l e 1. The role of gut microbiota in cancer
Bacteria Cancer type Abundance Mechanism Reference
Gut microbiota as tumor promoters
Peptostreptococcus anaerobius CRC enriched Inhibiting cancer growth (Long et al. 2019 : 2319–30)
Helicobacter pylori GC - Attaching to host cells and
deliv ering Ca gA into host
cells
(Buti et al. 2011 : 9238–43,
Suganuma et al. 2021 )
Esc heric hia coli CRC enriched Secretion of colibactin (Dalmasso et al. 2014 : 675–80)
Enterotoxigenic Bacteroides fragilis CRC - Secretion of BFT (Dejea et al. 2018: 592–7, Wu
et al. 2007 : 1944–52)
Clostridioides difcile CRC enriched Secretion of TcdB (Dr e wes et al. 2022 )
Fusobacterium nucleatum CRC enriched Suppressing anti-tumor
immunity
(Gao et al. 2021: 398, Gur
et al. 2015 : 344–55, Liu et al.
2022 : e2105222, Rubinstein et
al. 2013 : 195–206, Wu et al.
2022 : 1981–95)
Cutibacterium acnes PCa enriched unknown (Cohen et al. 2005: 1969–74)
Salmonella enterica GBC - Inducing MAPK/ERK,
PI3K/AKT/mTOR, CREB/SP-1,
BSG signaling
(Samonis et al. 2003: 5820–2)
P orph yromonas sp. CRC enriched Secretion of butyrate (Okum ur a et al. 2021: 5674)
Cutibacterium acnes EOC - Activating hedgehog pathway (Huang et al. 2022)
Gut microbiota as tumor suppressors
Streptococcus thermophilus CRC depleted Secretion of
beta-Galactosidase
(Li et al. 2021 : 1179–93 e14)
Lacticaseibacillus casei CRC depleted Secretion of ferrichrome
Regulating immune response
(Konishi et al. 2016 : 12 365,
Lee et al. 2004 : 41–8)
Bidobacterium longum CRC - Regulating immune response (Lee et al. 2004 : 41–8, Singh et
al. 1997 : 833–41)
Lactiplantibacillus plantarum CRC - Enhance butyrate uptake (Kim et al. 2022: 100–17)
Lactobacillus gallinarum CRC depleted Secretion of ILA (Sugim ur a et al. 2021 )
Bidobacterium bidum CRC - Regulating gut microbiota
and metabolic prole
(Wang et al. 2020 : 5915–28)
Bidobacterium breve CRC - Regulating immune response (Li et al. 2021 : 1 868 122)
Akkermansia muciniphila PCa - Regulating immune response (Luo et al. 2021 : 2949–63)
Propionibacterium acidipropionici and
Propionibacterium freudenreichii
CRC - Secretion of propionate and
acetate
(Jan et al. 2002 : 179–88)
Bidobacterium pseudolongum CRC - Secretion of inosine to
enhance anti-tumor
immunity
(Mager et al. 2020 : 1481–9)
Lacticaseibacillus rhamnosus CRC - Inducing cell apoptosis by
metabolites
Regulating immune response
(Dehghani et al. 2021 : 245–52,
Rahimpour et al. 2022 :
2622–31, Si et al. 2022 :
521–33)
Lactobacillus delbrueckii CRC - Inducing cell apoptosis (Wan et al. 2014: 1738–42)
Limosilactobacillus fermentum CRC - Inhibiting tumor metastasis (Liu et al. 2021 : 7281–93)
Lactobacillus acidophilus CRC - Inhibiting cell apoptosis
Regulating redox status
(Deepak et al. 2021: 225, Yu e
et al. 2021 : 788 040)
Lactobacillus brevis BRCA - Secretion of Lb-PPSPs (Pourbaferani et al. 2021:
982–92)
Bidobacterium breve HNC - Inducing cell cycle arrest and
cell apoptosis
(Wang et al. 2019 : 1044)
Clostridium sporogenes LC/CRC - Secretion of methionine
gamma-lyase
(Bhave et al. 2015: 15 681,
P okro vsky et al. 2019 : 201–9)
Faecalibaculum rodentium and
Holdemanella biformis
CRC depleted Secretion of SCFA (Zagato et al. 2020: 511–24)
Abbre via tion: BRC A, breast cancer; CRC, colorectal cancer; GBC, gallbladder cancer; GC, gastric cancer; HNC, head and neck cancer; LC, lung cancer; PCa, prostate
cancer; EOC, epithelial ova rian cancer.
Downloaded from https://academic.oup.com/femsre/article/47/4/fuad036/7220014 by The Chinese University of Hong Kong user on 26 July 2023
4 | FEMS Microbiology Reviews , 2023, Vo l. 47, No. 4
Figur e 1. T he function of m6A on RNA metabolism. RNA m6A modication is dynamically regulated by three types of proteins: writers , erasers , and
r eaders. Writers catal yze the addition of m6A marks, er asers r emov e m6A marks, and r eaders specicall y r ecognize and bind to m6A sites to impact
the RNA metabolism. m6A modications control RNA metabolism, including RNA splicing, nuclear export, translation, and RNA degradation.
Figur e 2.
T he role of m6A in cancers. m6A regulators play a critical role in regulating various aspects of cancer biology, including cancer stemness,
cancer gr owth, differ entiation, and metastasis, as well as tumor immunity and chemoresistance, by regulating the expression of cancer-related genes.
important to note that the benets and safety of probiotics in
humans are not yet fully understood, as many studies investigat-
ing their effects have been preclinical and conducted in animal
or cell culture models . T herefore , further research, particularly
well-designed clinical studies in humans, is necessary to deter-
mine the potential benets, optimal dosing, and safety of these
probiotics.
RNA m6A modication and cancer
The m6A modication is pr edominantl y found near stop codons
and in 3’-untranslated regions (3’-UTRs) of mRNA molecules, with
a consensus motif of RR(m6)ACH (R = G or A, H = A, C, or U) (Sun
et al. 2019 ). T he m6A modication pla ys a vital r ole in r egulat-
ing various aspects of RN A metabolism, including RN A splicing,
miRNA pr ocessing, nuclear export, tr anslation, and RNA decay
Downloaded from https://academic.oup.com/femsre/article/47/4/fuad036/7220014 by The Chinese University of Hong Kong user on 26 July 2023
Su et al. | 5
(Fig. 1 ) (Sun et al . 2019 ). Aberrant m6A modications have been
implicated in the regulation of multiple aspects of cancer biol-
ogy, including cancer stem cell pluripotency (Liu et al . 2021 ), cell
differentiation (Weng et al . 2018 ), cell pr olifer ation (H. Chen et al.
2021a ), cell metastasis (Zhou et al . 2021 ), anti-tumor immunity
(Bai et al . 2022 , Chen et al . 2022a ), and c hemor esistance (Jin et al.
2019 ). Her e we pr esent a thor ough ov ervie w of the m ultifaceted
roles that m6A modications play in different aspects of cancer
de v elopment (Fig. 2 and Table 2 ).
M6a writers
The m6A writer complex, composed of METTL3, an m6A methyl-
tr ansfer ase, and other ada ptor pr oteins, including METTL14,
WT AP, Vir -like m6A methyltr ansfer ase associated pr otein
(VIRMA), zinc nger CCCH-type containing 13 (ZC3H13), HAKAI,
RNA-binding motif protein 15 (RBM15), and RNA-binding motif
pr otein 15B (RBM15B), pr edominantl y catal yzes m6A methylation
on mRNA molecules (Zaccara et al . 2019 ). In addition to METTL3,
two new m6A methyltransferases have recently been identied,
namely METTL16 and Zinc Finger CCHC-Type Containing 4 (ZC-
CHC4). METTL16 methylates U6 spliceosomal small nuclear RNA,
MALAT long non-coding RNA, and the MAT2A mRN A (Bro wn
et al . 2016 , Pendleton et al. 2017 , Satterwhite and Manseld 2022 ),
while ZCCHC4 adds m6A to 28S rRNA (Ma et al . 2019 ). METTL3 is
fr equentl y ov er expr essed in v arious types of cancers, including
CRC , GC , hepatocellular carcinoma (HCC), lung cancer (LC), and
prostate cancer (PCa). Studies have shown that METTL3 upregu-
lates m6A le v els of GLUT1, miR-1246, and Sec62 in different CRC
cell lines and mouse models of CRC. This upregulation leads to
the activation of various oncogenic signaling pathwa ys , which
can further promote the pathogenesis of CRC (Chen et al . 2021a ,
Peng et al . 2019 , Liu et al . 2021 ). Furthermore, METTL3 facilitates
the formation of circHPS5 in an m6A-dependent manner, which
subsequently enhances HMGA2 expression and promotes HCC
pr ogr ession, as demonstr ated in both in vitro cell lines and in vivo
mouse models (Rong et al . 2021 ). According to Wang et al . ( 2022b ),
METTL14 is upregulated in primary PCa tissues and is associated
with poor prognosis of PCa patients. Functionally, METTL14
plays an oncogenic role by inhibiting THBS1 expression in an
m6A-dependent manner (Wang et al . 2022b ). Ho w e v er, in HCC
cells, METTL14 has been reported to have a tumor-suppressive
role (Du et al. 2021a ), suggesting that the role of METTL14 in
cancer may be context-dependent. Although it has no catalytic
activity, WTAP stabilizes the m6A methyltr ansfer ase complex
for m6A deposition. Similar to METTL3, WTAP is upregulated in
different types of human cancer, including HCC, bladder cancer
(BLCA), and breast cancer (BRCA). Studies have shown that WTAP
can promote cancer progression and metastasis by regulating
the expression of ENO1 in BRCA cells and TNFAIP3 in BLCA cells
(Ou et al. 2021 , Wei et al. 2021 ). Additionall y, studies hav e shown
that METTL16 dri ves leuk emogenesis in mice by regulating the
expression of BC AT1/BC AT2 (Han et al . 2023 ).
M6a erasers
FTO and ALKBH5 are tw o w ell-kno wn m6A erasers . FT O is the rst
m6A demethylase to be identied and is responsible for reducing
total RNA m6A le v els in cells (Jia et al . 2011 ). FTO has been found to
have both oncogenic and tumor-suppressive roles. In BLCA, CRC,
and GC cells, high FTO expression promotes cancer cell growth
and metastasis via promoting MALAT and MYC expression, and
inducing cav eolin-1 degr adation, r espectiv el y (Tao et al. 2021 , Yu e
et al . 2020 , Zhou et al . 2022 ). Mor eov er, a specic FTO inhibitor has
been shown to suppress the growth and lung colonization of BRCA
cells in mice (Xie et al . 2022 ). Ho w e v er, hypoxia-induced downr eg-
ulation of FTO has been reported to facilitate CRC growth and
metastatic potential (Ruan et al . 2021 ). In this study, protein ex-
pression of FTO, but not mRN A, w as reduced in CRC tissues, and
low FTO protein expression could predict a high recurrence rate
and poor prognosis in CRC patients (Ruan et al . 2021 ). Addition-
ally, SIRT1 has been reported to play an oncogenic role in HCC
by r epr essing FTO expr ession (Liu et al . 2020b ), suggesting that
the function of FTO in cancer is context-dependent. Similar to
FTO, abnormal expression of ALKBH5 has been observed in differ-
ent types of human cancer. ALKBH5 is ov er expr essed in multiple
myeloma (MM) and predicts a poor prognosis of MM patients (Qu
et al. 2022 ). Functionall y, ALKBH5 r educes the m6A le v el of TRAF1
mRNA to increase TRAF1 mRNA stability, leading to the activation
of NF- κB and MAPK pathways that contribute to MM tumorigen-
esis (Qu et al . 2022 ). Ho w e v er, low expr ession of ALKBH5 has been
identied in osteosarcoma, which is associated with poor survival
of osteosarcoma patients. ALKBH5-mediated m6A demethylation
inhibits the decay of SOCS3 mRNA, leading to the inactivation of
the ST A T3 pathway and consequent inhibition of cell pr olifer a-
tion, induction of cell apoptosis, and cell cycle arrest (Yang et al.
2022 ).
M6a readers
T he m6A readers , such as YTHDF1-3, YTHDC1-2, and IGF2BP1-3,
specicall y r ecognize and bind to m6A-modied RNAs and regu-
late their biological functions. Each m6A reader plays a distinct
role in RNA metabolism, contributing to the intricate nature of
the m6A epitranscriptome. YTHDF1 has been shown to function
as an oncogene in various cancers by accelerating the transla-
tion of m6A-modied mRNAs (Chen et al. 2021b ; Liu et al. 2020a ,
Wan g et al. 2015 , 2021 a). In contrast, YTHDF2 reduces the stability
of m6A-modied targeted mRNAs, making it the primary decay-
inducing m6A r eader. Additionall y, YTHDF3 pr omotes YTHDF2-
mediated mRNA decay by increasing the accessibility of RNA to
YTHDF2 (Shi et al . 2017 ). YTHDF2 has been found to play a role in
tumorigenesis and cancer pr ogr ession. In liv er cancer cells, Zhang
et al. reported that YTHDF2 promotes cancer stemness and
metastasis by increasing OCT4 expression in an m6A-dependent
manner (Zhang et al. 2020a ). Additionally, Li et al . sho w ed that
YTHDF2 promotes CRC cell proliferation by targeting m6A-
modied GSK3- βmRNA for degr adation, whic h leads to the ac-
tivation of Wnt/beta-catenin/Cyclin D1 signaling pathway (Li et
al . 2021a ). In contrast, Zhong et al. demonstrated that YTHDF2
suppressed HCC cell growth by destabilizing m6A-modied EGFR
mRNA (Zhong et al. 2019 ). The discrepancies in these ndings
could be due to the differing m6A-modied RNAs targeted by
YTHDF2 across various cancer types and cell lines. YTHDC1
is known to regulate the alternative splicing of m6A-modied
RNAs, while YTHDC2 inuences the translation efciency of tar-
get genes in an m6A-dependent manner (Yang and Chen 2021 ).
Both YTHDC1 and YTHDC2 play a pivotal role in various cancer
types by targeting different m6A-modied mRNAs (Hou et al. 2021 ,
Tanabe et al. 2016 , Ma et al . 2021 , Sheng et al. 2021 , Wu et al. 2021 ).
The family of IGF2BP proteins has recently been recognized as
m6A readers. Unlike YTHDF2-induced RNA decay, IGF2BP1-3 can
stabilize m6A-containing RN A b y binding to the m6A sites (Huang
et al. 2018 ). Furthermor e, the tr anscriptome-wide binding sites of
YTHDF2 and IGF2BP1-3 are distinct (Huang et al. 2018 ). The role of
IGF2BP proteins in tumorigenesis and cancer pr ogr ession has been
documented in different cancer types, including gastrointestinal
Downloaded from https://academic.oup.com/femsre/article/47/4/fuad036/7220014 by The Chinese University of Hong Kong user on 26 July 2023
6 | FEMS Microbiology Reviews , 2023, Vo l. 47, No. 4
Tab l e 2. The role of m6A regulators in cancer.
Regulator Cancer type Role Targets Function Reference
METTL3 CRC oncogene SOX2 , GLUT1 ,
YPEL5 , miR-1246 ,
CRB3 , Sec62
Tumorigenesis and
metastasis
(Chen et al. 2021 : 1284–300
e16, Liu et al. 2021 : 132, Pan
et al. 2022 : 19, Peng et al.
2019 : 393, Zhou et al. 2021 :
2172–84)
METTL3 HCC oncogene SOCS2 , LINC00958 ,
Has_circ_0 058 493 ,
CircHPS5, HMGA2 ,
RDM1
Tumor pr ogr ession (Chen et al. 2018 : 2254–70,
Chen et al. 2020 : 373–86,
Rong et al. 2021 : 637–48, Wu
et al. 2021 : 762588, Zuo et al.
2020 : 5)
METTL3 GC oncogene HDGF , ZMYM1 ,
SPHK2 , THAP7-AS1 ,
PBX1/GCH1
Tumor angiogenesis,
tumor gr owth, and liv er
metastasis
(Huo et al. 2021 : 2968–81, Liu
et al. 2022 : 627–41, Wang et
al. 2020 , Liu et al. 2022 :
1193–205, Yue et al. 2019 : 142)
METTL3 PCa oncogene USP4/ELAVL1 , PCAT6 Migration and
invasion
(Chen et al. 2021: 7640–57,
Lang et al. 2021 : e426)
METTL3 LC oncogene ABHD11-AS1 , ZBTB4 ,
YAP
Pr olifer ation,
metastasis, and drug
resistance
(Cheng et al. 2021: 487–500,
Jin et al. 2019 : 135, Xue et al.
2021 : 2649–58)
METTL14 PC oncogene THBS1 Pr olifer ation (Wang et al. 2022 : 143)
METTL14 CRC TSG MiR-375 Tumorigenicity and
metastasis
(Chen et al. 2020: 599–612)
METTL14 HCC TSG USP48 , HNF3 γ Gl ycol ysis, malignancy,
and anti-cancer
c hemother a py
(Du et al. 2021 : 3822–34, Zhou
et al. 2020 : 296)
METTL16 AML oncogene BC AT1/ B C AT2 Leukemogenesis (Han et al. 2023 : 52–68 e13)
WTAP HCC oncogene ETS1 Tu m o r pr ogr ession (Chen et al. 2019: 127)
WTAP OS oncogene HMBOX1 Growth and
metastasis
(Chen et al. 2020: 659)
WTAP BRCA oncogene ENO1 Gl ycol ysis and growth (Ou et al. 2021 : 737)
WTAP DLBCL oncogene HK2 Tu m o r pr ogr ession (Han et al. 2021: 1603–14)
FTO BLCA oncogene MALAT /miR-
384/MAL2
Tumorigenesis (Tao et al. 2021 : e310)
FTO CRC oncogene MYC Tu m o r pr ogr ession (Yue et al. 2020 : 240)
FTO GC oncogene Caveolin-1 Growth and
metastasis
(Zhou et al. 2022 : 72)
FTO CRC TSG MTA1 Metastasis (Ruan et al. 2021 : 5168–81)
FTO HCC TSG GNAO1 Tu m o r pr ogr ession (Liu et al. 2020 : 2029–50)
FTO HCC oncogene SOX2, KLF4, NANOG Cancer stemness (Bian et al. 2021: e352)
ALKBH5 MM oncogene TRAF1 Tumorigenesis (Qu et al. 2022 : 400–13)
ALKBH5 OS TSG SOCS3 Pr olifer ation and
tumorigenicity
(Yang et al. 2022 : 104 019)
YTHDF1 GC oncogene USP14 Tumorigenesis and
metastasis
(Bai et al. 2022 , Chen et al.
2021: 647702)
YTHDF1 CRC oncogene ARHGEF2 Tu m o r growth and
metastasis
(Wang et al. 2021 )
YTHDF1 HCC oncogene A TG2A , A TG14 ,
FZD5 , EGFR
Autophagy and
autopha gy-r elated
malignancy
(Liu et al. 2020 : 750–65,
Ouyang et al. 2021 : 1217, Su
et al. 2021 : 1339–56)
YTHDF2 HCC oncogene OCT4 Cancer stemness and
metastasis
(Zhang et al. 2020 : 4507–18)
YTHDF2 CRC oncogene GSK3 β Pr olifer ation (Li et al. 2021 : e602)
YTHDF2 HCC TSG IL11 , SERPINE2 ,
EGFR
Inammation, vascular
reconstruction, and
metastasis
(Hou et al. 2019: 163, Zhong
et al. 2019 : 252–61)
YTHDF3 CRC oncogene LncRNA GAS5 Tum o r pr ogr ession (Ni et al. 2019: 143)
YTHDC1 HCC oncogene Has_circ_0 058 493 Growth and
metastasis
(Wu et al. 2021 : 762588)
YTHDC1 AML oncogene MCM4 Tumor de v elopment (Sheng et al. 2021 : 2838–52)
YTHDC1 PC TSG MiR-30d Tumorigenesis (Hou et al. 2021 : 3105–24)
Downloaded from https://academic.oup.com/femsre/article/47/4/fuad036/7220014 by The Chinese University of Hong Kong user on 26 July 2023
Su et al. | 7
Tab l e 2. Continued
Regulator Cancer type Role Targets Function Reference
YTHDC2 CRC oncogene HIF-1 α Tu m o r metastasis (Tanabe et al. 2016 : 34–42)
YTHDC2 LC TSG SLC7A11 Tumorigenesis (Ma et al. 2021 : 101801)
IGF2BP1 HCC oncogene c-MYC , MKI67 Tumorigenesis (Gutschner et al. 2014 :
1900–11)
IGF2BP1 CRC oncogene LDHA Anticancer ther a py (Zhang et al. 2021: 835–46)
IGF2BP1 LC oncogene Kras Growth and migration (Wallis et al. 2022: 26–43)
Abbre via tion: AML, acute myeloid leukemia; BLCA, bladder carcinoma; BRCA, breast cancer; CRC, colorectal cancer; GC, gastric cancer; HCC, hepatocellular carci-
noma; LC, lung cancer; MM, multiple myeloma; OS, osteosarcoma; PC, pancreatic cancer; PCa, prostate cancer; TSG, tumor suppressor gene.
cancer , breast cancer , and lung cancer (Du et al. 2021b ). For ex-
ample, IGF2BP1 has been found to promote HCC cell pr olifer ation
while inhibiting apoptosis by stabilizing c-MYC and MKI67 mRNAs
(Gutschner et al. 2014 ). Overall, the functions of m6A regulators in
cancer are context dependent.
Crosstalk between gut microbiota and m6A
modication
Both gut microbiota and m6A modication have been shown to
play a signicant role in cancer development. Understanding their
individual and combined effects could pro vide no vel insights into
cancer biology and lead to the de v elopment of ne w ther a peutic
strategies for cancer patients.
The inuence of gut microbiota on host RNA m6a
modication
Studies have demonstrated that gut microbiota composition al-
ter ations can signicantl y c hange host epitr anscriptomic pr o-
les (Fig. 3 ). In a recent publication, signicant differences were
found in the m6A proles of the cecum and liver tissues between
conv entional and germ-fr ee mice, whic h could be attributed
to altered expression of m6A methyltransferase METTL16 (Jabs
et al. 2020 ). These differential m6A peaks were enriched in the
metabolic , inammatory response , and antimicr obial r ecognition
and signaling pathways (Jabs et al. 2020 ). In support of this, an-
other study using m6A-MeRIP sequencing of various tissues from
germ-free and SPF mice revealed higher m6A content of the brain,
intestine, and kidney tissues of germ-free mice compared to SPF
mice (Wang et al. 2019 ). Intriguingly, this study found that m6A
writers, METTL3 and METTL14, as well as m6A erasers, ALKBH5
and FTO, were upregulated in brain tissues of GF mice compared
to SPF mice. At the same time, higher expression of METTL14 was
observed in the intestine of SPF mice compared to GF mice (Wang
et al . 2019 ). Additionally, a study sho w ed that a high-fat diet de-
cr eased global m6A le v els in pig jejunal, whic h could be r estor ed
by fecal microbiota transplant (FMT) of wild boar fecal suspen-
sion (Zhu et al. 2022 ). These ndings suggest that gut microbiota
can signicantly impact the host RNA m6A prole.
We next ask whether a specic micr oor ganism can impact the
m6A patterns of its host. Gr owing e vidence suggests that certain
bacteria have the ability to increase RNA m6A modications in
the host cells. Enterotoxigenic Escherichia coli has been shown to
promote global RNA m6A level mediated by METTL3 in intesti-
nal epithelial cells, leading to increased expression of β-defensin
(Zong et al . 2021 ). Additionally, heat-killed Salmonella typhimurium
infection has been shown to increase WTAP-mediated global RNA
m6A le v els in THP-1 cells (Wu et al . 2020a ). Lactobacilli and Bi-
dobacterium species have also been found to incr ease m6A le v els
in the total RNA of gut tissues, which favors intestinal de v elop-
ment (Wu et al. 2020b ). On the other hand, certain bacterial in-
fections have been shown to suppress m6A enrichment in host
cells . For example , a r ecent publication demonstr ated that Fu-
sobacterium nucleatum could decrease the RNA m6A le v el in CRC
cells and patient-deriv ed xenogr aft tissues by activating YA P sig-
naling and inhibiting FOXD3 expr ession, whic h leads to reduced
METTL3 transcription and decreased m6A levels of kinesin fam-
ily member 268 (KIF268) (Chen et al . 2022 c). T his , in turn, abro-
gates YTHDF2-dependent degradation of KIF268 mRNA, result-
ing in upregulated KIF268 expression that contributes to Fusobac-
terium nucleatum -induced CRC metastasis (Chen et al . 2022 c). More-
over, ETBF has been found to inhibit METTL14 expression and dis-
rupts the splicing process of pri-miR-149 in an m6A-dependent
manner, leading to reduced miR-149–3p expression and increased
CRC cell pr olifer ation (Cao et al . 2021 ). All these ndings suggest
that certain bacteria have the potential to alter the RNA m6A pro-
le of their host, which may, in turn, inuence the de v elopment
of cancer. Ne v ertheless, it is essential to note that onl y a limited
number of studies have investigated the potential role of host RNA
m6A modication as an intermediary between microbial dysbio-
sis and cancer de v elopment. Ther efor e, further r esearc h is needed
to examine the potential interplays between gut microbiota and
host RNA m6A modication to better understand their contribu-
tions to cancer de v elopment.
What mechanisms underlie the regulatory role of microbiota
on the host m6A pr ogr am? Lactiplantibacillus plantarum and Lati-
lactobacillus sakei have been found to upregulate the AMPK/SIRT1
pathway in mice (Jang et al . 2019 , Wang et al . 2021 b). Importantly,
SIRT1 has been demonstrated to play a crucial role in various
types of human cancers, including leukemia, BRCA, and CRC (Her-
ranz et al. 2013 , Chen et al. 2014 , Yuan et al. 2012 , Jin et al . 2018 ).
SIRT1 can promote the degradation of FTO through RANBP2-
mediated SUMOylation of FTO, which results in the alteration of
the m6A methylome and the tuning of downstr eam tar get expr es-
sion (Liu et al . 2020b ). Ther efor e, the AMPK/SIR T1 pathw ay r epr e-
sents a possible mediator between bacterial infections and alter-
ations in host m6A le v els . T he IL-6/ST A T3 pathway is another pos-
sible intermediate link. IL-6 is an inammatory factor that plays
a vital role in various biological processes (Heinrich et al . 2003 ,
Waldner et al . 2012 ), and the IL-6/ST A T3 pathway is crucial for
cancer growth and development. Bacteria, such as Escherichia coli ,
Staph ylococcus hominis , and Staph ylococcus lentus , can promote the
production of IL-6 (Larsson et al. 1999 ). Additionally, the admin-
istr ation of lipopol ysacc haride (LPS), an essential component of
Gr am-negativ e bacteria, consistentl y activ ates IL-6 and ST A T3 sig-
naling in the mouse br ain (Beur el and J ope 2009 ). Furthermore ,
bacteria-deriv ed extr acellular v esicles hav e been demonstr ated
to r elease LPS, whic h can stim ulate the secr etion of IL-6 in pe-
ripheral blood mononuclear cells (Tulkens et al. 2020 ). T hus , the
Downloaded from https://academic.oup.com/femsre/article/47/4/fuad036/7220014 by The Chinese University of Hong Kong user on 26 July 2023
8 | FEMS Microbiology Reviews , 2023, Vo l. 47, No. 4
Figur e 3.
T he inuences of gut microbiota on host m6A prole. Different bacterial species may modulate m6A modication through various
mechanisms . For example , Lactiplantibacillus plantarum and Latilactobacillus sakei upregulate AMPK/SIRT1 signaling to promote FTO degradation, while
Esc heric hia coli , Staphylococcus hominis , and Staphylococcus lentus activate IL-6/ST A T3 signaling to enhance the expression of m6A methyltransferase
complex (METTL3/METTL14/WTAP) and ele v ate RNA m6A le v els. Akkermansia muciniphila and Lactiplantibacillus plantarum have also been shown to alter
host m6A le v els. Additionall y, enter otoxigenic Esc heric hia coli pr omotes global RNA m6A le v els mediated by METTL3, while Salmonella typhimurium
induces global RNA m6A le v els thr ough WTAP. Fusobacterium nucleatum , on the other hand, decr eases RNA m6A le v els by r epr essing METTL3
expr ession, and enter otoxigenic Bacteroides fragilis inhibits METTL14 to suppr ess miR-149–3p expr ession.
gut microbiota can manipulate the IL-6/ST A T3 pathway . Notably ,
the activation of IL-6/ST A T3 has been reported to increase RNA
m6A le v el by enhancing the expression of the m6A methyltrans-
fer ase complex (METTL3/METTL14/WTAP), r esulting in an alter ed
m6A methylome that promotes cancer stemness (Ye et al . 2021 ).
Together, the microbiome can potentially trigger intracellular cas-
cades within host cells that inuence RNA m6A pr ole, ultimatel y
impacting cancer de v elopment.
The inuence of host RNA m6a modication on
the microbiome
The gut microbiota is not only affected by external factors but
also by host gene expr ession pr ogr ams . T hr ough the anal y-
sis of metagenomic data from the Human Microbiome Project,
Blekhman et al. discov er ed signicant corr elations between se v-
eral host signaling pathwa ys , such as Leptin signaling, JAK/ST A T
signaling, and IL12-mediated signaling, and gut microbiota com-
position (Blekhman et al . 2015 ). Furthermore, in a study that
analyzed the 16S ribosomal RNA gene sequences of intestinal
biopsies and genotypes of 474 individuals, Knights et al. found a
signicant correlation between the host NOD2 risk allele dosage
and increased Enterobacteriaceae abundance (Knights et al . 2014 ).
T hese ndings , in conjunction with those of Blekhman et al., sug-
gest a connection between host gene expression and gut micro-
biota composition. Furthermore, Nod2 -decient mice exhibit gut
dysbiosis, leading to more severe colitis and a higher incidence of
colitis-associated carcinogenesis (Couturier-Maillard et al . 2013 ).
ALKBH5 has been shown to control bacterial infections in trans-
genic mouse models (Liu et al . 2022 ). In a mouse model of sep-
sis, mice with Alkbh5 knoc k out exhibit a higher bacterial burden
in the peritoneal cavity and blood, leading to increased sepsis-
related mortality (Liu et al . 2022 ). Collectively, alterations in host
gene expr ession thr ough genetic , epigenetic , or epitranscriptomic
r epr ogr amming can potentially modify the gut microbiota.
The host RNA m6A pr ogr am may impact micr obiome compo-
sition by regulating the expression of genes closely related to the
gut micr obiota, suc h as the famil y of suppr essors of cytokine sig-
naling (SOCS), vitamin D receptor (VDR), and mucin. The family
of SOCS pr oteins serv es as inhibitors of cytokine signaling, in-
cluding JAK/ST A T (Croker et al . 2008 ). Importantl y, SOCS famil y
genes, including SOCS1, SOCS2, SOCS3, and CISH, exhibit enriched
m6A peaks and are subject to m6A-dependent mRNA degradation
(Chen et al . 2018 , Li et al . 2017 ). In naive T cells decient in METTL3,
the loss of m6A marks on SOCS1, SOCS3, and CISH mRN A slo ws
do wn their mRN A decay, r esulting in incr eased mRNA and pr otein
expr ession le v els (Li et al . 2017 ). As a consequence, the inactiva-
tion of IL-7-mediated ST A T5 signaling occurs upon the silencing
of METTL3 (Li et al . 2017 ). This nding is consistent with the ob-
servation that the knockdown of METTL3 reduces the m6A level
Downloaded from https://academic.oup.com/femsre/article/47/4/fuad036/7220014 by The Chinese University of Hong Kong user on 26 July 2023
Su et al. | 9
of SOCS2 mRNA, leading to increased SOCS2 expression in HCC
cells (Chen et al . 2018 ). The JAK/ST A T pathway is critical for main-
taining intestinal and microbial homeostasis. Upon nematode in-
fection, activation of ST A T6 in mouse epithelium inhibits ion ab-
sorption while increasing mucous secretion and release of antimi-
crobial compounds to facilitate infection clearance (Spencer et al.
2001 ). Similarl y, activ ated ST A T3 in the epithelium promotes the
r elease of antimicr obial compounds, suc h as β-defensins (Wolk
et al . 2004 ). Ther efor e, the host m6A pr ogr am has the potential to
inuence gut microbiota composition by regulating the JAK/ST A T
pathway.
Deletion of VDR in the mouse intestine has been demonstrated
to induce gut dysbiosis through the modulation of the JAK/ST A T
pathway (Zhang et al . 2020b ). VDR is expressed at high le v els in
the intestine and is recognized to have a protective role in can-
cer (Carlberg and Munoz 2020 ). A genome-wide association study
comprising 1812 individuals r e v eals a signicant association
between VDR genotypes and ov er all gut micr obial v ariation (Wang
et al . 2016 ), suggesting that host VDR activation may alter the gut
microbiota composition. Consistent with this, constitutive knock-
out of Vdr in mice has been shown to result in a signicant shift
in gut microbiota composition (Wang et al . 2016 ). Specically, a
higher abundance of Parabacteroides is observed in mice when Vdr
is deleted (Wang et al. 2016 ). VDR not only affects bacteria but
also regulates the gut virome. In an intestinal Vdr knockout mouse
model, silencing of Vdr has been found to increase the abundance
of Vibrio phage JSF5 and bovine viral diarrhea virus-1 in the stool
(Zhang et al . 2021b ). In contrast, Lactobacillus prophage Lj771 , Lac-
tobacillus prophage phiadh , Lactobacillus prophage KC5a , Macacine al-
phaherpesvirus 1 , and Catovirus CTV1 are depleted in female mice
with Vdr deletion (Zhang et al . 2021b ). Mec hanisticall y, VDR r egu-
lates the expression and signaling of target genes that contribute
to gut dysbiosis. When Vdr is knocked out in the intestinal, it leads
to impair ed P aneth cell function and r educed pr oduction of an-
timicrobial peptides (Wu et al . 2015 ). Collectiv el y, VDR plays a cru-
cial role in maintaining the balance of the intestinal microbiome.
Inter estingl y, emer ging e vidence has shown that the expr ession
of VDR is regulated by various m6A regulators. For instance, the
knockdown of YTHDF1 has been found to increase the translation
efcienc y of VDR b y anal yzing ribosome pr oling data (Wang et al .
2015 ). Ad ditionally, de pletion of METTL14 has been shown to re-
duce VDR mRNA expression by analyzing RNA-seq data in both
HepG2 and NB4 cells (Weng et al . 2018 , Huang et al . 2019a ). How-
e v er, none of these studies have conrmed the presence of m6A
marks on VDR mRNA. Ther efor e, whether VDR is a direct down-
str eam tar get of the m6A pr ogr am r emains to be determined.
Like VDR, mucin also plays a protective role in humans and has
a close relationship with the gut microbiota (Bergstrom et al. 2020 ,
Hansson 2020 , Paone and Cani 2020 ). Dysregulated mucin expres-
sion is str ongl y linked to various diseases, including CRC (Pothu-
raju et al . 2020 ). There are two types of mucins: transmembrane
mucins (MUC1, MUC3, MUC4, MU12, MUC13, MUC15, MUC17,
MUC20, MUC21, and MUC22) and secreted mucins (MUC2, MUC5B,
and MUC6) (Paone and Cani 2020 ). Tr ansmembr ane m ucins ar e
present on the apical surfaces of epithelial cells and are involved
in host-microbe interactions. MUC1 has been found to prevent
the binding of Helicobacter pylori to enterocytes in mice (McGuckin
et al. 2007 ). Furthermore , o verexpression of MUC17 can protect ep-
ithelial cells by blocking the attachment of enteropathogenic Es-
c heric hia coli (Schneider et al. 2019 ). In contrast, secreted mucins,
mainl y pr oduced by goblet cells, pr ovide nutrients and attach-
ment sites for gut microbiota (Hansson 2020 ). MUC2 is the pri-
maril y secr eted m ucin in the intestine, forming the m ucus skele-
ton (Paone and Cani 2020 ). MUC2 maintains a healthy gut mi-
cr obiota, and dysr egulated MUC2 expr ession can lead to micro-
biome dysbiosis (Ber gstr om et al . 2020 ). Supporting this observ a-
tion, distinct microbiome compositions have been identied be-
tween Muc2
/ mice and their control littermates (Leon-Coria et
al . 2021 ). As a result, Muc2
/ mice are susceptible to chemical,
bacterial, and parasite-induced colitis (Leon-Coria et al . 2021 ). Sev-
eral members of the MUC family genes are m6A-modied, in-
cluding MUC3A (Zhao and Xie 2021 ), MUC5B (Ruan et al . 2022 ),
and MUC15 (Gan et al. 2021 ). Their expressions can be modulated
by differ ent m6A r egulators. In addition, se v er al m6A-modied
genes, such as Galectin-3 and KLF4, have been shown to regu-
late m ucin expr ession. Galectin-3 has been identied as a tar-
get of the m6A eraser ALKBH5 (Zhang et al. 2022 ). In CRC cells,
Galectin-3 has been shown to regulate MUC2 transcription (Song
et al . 2005 ). In addition, KLF4, a zinc-nger transcription factor, is
suppressed by METTL3 (Chien et al. 2021 , Wu et al. 2019b ). Deletion
of KLF4 has been found to increase MUC2 expression in GC cells
(Yu et al . 2016 ).
All of these studies suggest that disruptions to the host RNA
m6A pr ogr am hav e the potential to alter microbiome composition
by altering host gene expression.
Use of probiotics and small molecules
targeting m6A regulators for cancer
treatment
Pr obiotics ar e belie v ed to offer potential health benets, including
reducing certain side effects of cancer treatment. Some preclinical
studies have even suggested that probiotics may help prevent cer-
tain types of cancer (Luo et al . 2021 , An and Ha 2022 ). Meanwhile,
dysregulation of m6A modication has been linked to the de v el-
opment and pr ogr ession of v arious cancers, making it a pr omising
target for cancer treatment. By targeting m6A regulators, it may
modulate the le v els of m6A modication in cancer cells and al-
ter their behavior (Chen et al. 2012 , Dolbois et al. 2021 , Su et al.
2018 , 2020 , Huang et al. 2019b , Moroz-Omori et al . 2021 , Yank ov a
et al . 2021 , Zheng et al. 2014 ). Although these studies are still pre-
clinical, probiotics or targeting m6A regulators may be considered
potential adjuvants to cancer therapy.
Probiotics for cancer treatment
Pr obiotics hav e gained signicant attention in recent years due to
their potential health benets . T her e ar e ov er 1 900 clinical studies
r egister ed at ClinicalTrials .go v ( https://clinicaltrials.go v/) in vesti-
gating the ther a peutic potential of probiotics, and an increasing
number of these studies are specically focusing on using probi-
otics as food supplements to support cancer patients during an-
ticancer tr eatments. Se v er al published clinical trials hav e shown
pr omising r esults for using pr obiotics in cancer patients. For ex-
ample, Enterococcus faecium M-74 has been administered to prevent
febrile neutropenia, a severe side effect of anti-cancer chemother-
ap y, and w as found to be effective for cancer patients (Mego et al.
2005 ). In another study, supplementation of Lacticaseibacillus rham-
nosus GG alle viated c hemother a py-induced diarrhea in cancer pa-
tients r eceiving 5-Fluor our acil (5-FU) tr eatment (Osterlund et al .
2007 ). Additionally, Lacticaseibacillus rhamnosus GKLC1 has been re-
ported to reduce cisplatin-induced chronic ne phroto xicity in pa-
tients (Tsai et al. 2022 ). These encour a ging r esults fr om the clini-
cal trials suggest that probiotics may have potential benets as a
supportiv e ther a py for cancer patients.
Downloaded from https://academic.oup.com/femsre/article/47/4/fuad036/7220014 by The Chinese University of Hong Kong user on 26 July 2023
10 | FEMS Microbiology Reviews , 2023, Vo l. 47, No. 4
Preclinical studies conducted in cell lines or mouse models also
suggest that probiotics may enhance tumor response to treat-
ment. For instance, Lactiplantibacillus plantarum -derived extracel-
lular vesicles have been shown to restore the chemosensitivity of
5-FU-resistant CRC cells (An and Ha 2022 ). Additionally, Akkerman-
sia muciniphila has shown promising results in enhancing the anti-
tumor efcacy of c hemother a py and imm unother a py in immune-
competent mice and is considered a next-generation probiotic
(Luo et al . 2021 ). T hus , probiotics ma y serve as adjuv ant ther a py
for cancer by reducing intestinal toxicity and improving the host’s
anti-tumor r esponses. Notabl y, Nazli et al. r ecentl y conducted
an open-label and single-center pr ospectiv e study (NCT03829111)
demonstrating the potential benets of supplementing the diet of
adv anced r enal cell carcinoma patients receiving dual immune
c hec kpoint bloc kade (Nivolumab plus ipilim umab) with liv e bac-
terial product CBM588 containing Clostridium butyricum (Dizman et
al. 2022 ). Despite the small sample size of this study, patients with
metastatic renal cell carcinoma who received CBM588 achieved
higher objective response rates and prolonged progression-free
survival (Dizman et al . 2022 ). While these ndings are promising, it
is crucial to consider the potential health risks of using probiotics
in imm unocompr omised cancer patients individuall y. Lar ge-scale
clinical trials are needed to evaluate the safety and efcacy of ad-
ministering probiotics during anticancer therapy.
Inhibition of m6A regulators for cancer treatment
There is growing research focused on developing specic in-
hibitors of m6A regulators. For instance, se v er al small molecules
have been identied as potent inhibitors of METTL3, including
UZH1a (Moroz-Omori et al. 2021 ), STM2457 (Yankova et al . 2021 ),
UZH2 (Dolbois et al . 2021 ). Among these, STM2457 has been
shown to inhibit the methyltr ansfer ase activity of METTL3 specif-
icall y and effectiv el y. In pr eclinical studies, the administration of
STM2457 signicantl y impair ed the engr aftment of acute myeloid
leukemia (AML) in mice and prolonged their survival (Yankova et
al . 2021 ). Consistentl y, our r esearc h team has found that treat-
ment with STM2457 reduces the growth of mouse CRC allograft
and enhances the efcacy of anti-PD1 treatment by modulating
anti-tumor imm une r esponse (Chen et al . 2022b ). FTO is also a
potential target for cancer ther a py. Se v er al specic inhibitors of
FTO, suc h as R-2-Hydr oxyglutar ate (R-2HG) (Su et al. 2018 ), Rhein
(Chen et al . 2012 ), FB23-2 (Huang et al . 2019b ), CS1 (Bisantrene) and
CS2 (Brequinar) (Su et al. 2020 ), and MO-I-500 (Zheng et al . 2014 ),
have been identied. Preclinical studies conducted in cell lines
or mouse models have shown that R-2HG, FB23-2, CS1, and CS2
display strong anti-proliferative effects against leukemia (Su et al .
2018 , 2020 , Huang et al . 2019b ). Furthermor e, Rhein has been r e-
ported to suppress the growth of pancreatic cancer and CRC cells
markedly (Yang et al . 2019 , Zhang et al. 2021a ). While these small
molecules targeting the aberrant m6A epitranscriptome in can-
cer hav e demonstr ated potential in cancer treatment, these nd-
ings are based solely on preclinical studies . T herefore , future clin-
ical trials are necessary to assess the safety and efcacy of these
molecules for cancer patients.
T her apeutic potential of combing probiotics and
small molecules targeting m6A regulators for
cancer treatment
Imm unother a py, particularl y imm une c hec kpoint bloc kade ther-
apy, is one of the most promising strategies for cancer treat-
ment. Tw o rst-in-human clinical trials conducted by Baruch et al.
and Davar et al. indicate that the gut microbiota composition
can impact the effectiveness of cancer imm unother a pies (Baruc h
et al . 2021 , Davar et al . 2021 ). In these clinical trials, FMT was
used in metastatic melanoma patients undergoing anti-PD-1 im-
m unother a py. The r esults showed that FMT led to an increase
in benecial bacteria, which was associated with an augmented
anti-tumor imm une r esponse when combined with anti-PD-1
treatment. T hus , it is possible that taking probiotic supplements
could impr ov e the body’s r eaction to cancer imm unother a p y b y
activ ating ada ptiv e imm une r esponses. Lacticaseibacillus casei Shi-
rota and Bidobacterium breve have been shown to promote the
maturation of bone marrow cell-derived dendritic cells (BMDCs)
and recruit mature DCs to the tumor site in mice, where DCs can
secret IL12 to induce T cells accumulation and enhance antitu-
mor immunity (Takagi et al . 2008 , Li et al. 2021b ). In addition, sup-
plementation of Lactiplantibacillus plantarum to mice can increase
CD8 + T cell inltration by secreting acetate (Wang et al. 2022a ).
It is noteworthy that targeting m6A regulators also have the po-
tential to impr ov e the host’s r esponse to cancer imm unother a py
but through distinct mechanisms. Our research team has shown
that targeting METTL3 with STM2457 compromises the ability of
CRC cells to drive the accumulation and suppressive potency of
m yeloid-deri v ed suppr essor cells (MDSCs), leading to sustained
activation and expansion of T cells in mice (Chen et al . 2022b ). Sim-
ilarly, administering an ALKBH5-specic inhibitor in tumor cells
would enhance the efcacy of imm unother a py by inhibiting the
accumulation of suppressive Tre g and MDSCs in mice (Li et al .
2020 ). Intriguingly, depleting ALKBH5 in intrahepatic cholangio-
carcinoma cells has been found to promote PD-L1 mRNA degra-
dation, resulting in enhanced antitumor T- c e l l immunity in mice
(Qiu et al . 2021 ). Building on these ndings, combining the target-
ing of m6A regulators in cancer cells and probiotic supplementa-
tion may lead to a syner gistic effect. Ta r geting m6A r egulators can
enhance the immune response against cancer cells by blocking
imm une-suppr essiv e activity, while pr obiotics may have the po-
tential to prime and activate anti-tumor CD8 + T cells. Nonethe-
less, further studies are needed to determine the optimal combi-
nation dosing and scheduling and the safety and efcacy of this
a ppr oac h in cancer patients undergoing anticancer therapy.
Conclusion and future direction
Recent advances in shotgun metagenomic sequencing and m6A
sequencing have enabled comprehensive proling of the gut mi-
crobiota and host m6A e pitranscriptome, respecti vely. Vari ou s ex-
perimental models, both in vitro and in vivo , have been developed
to study the connection between cancer and the gut microbiota or
m6A, alongside the underlying mechanisms that drive this corre-
lation. Ne v ertheless, the inter actions between the gut microbiota
and host m6A pr ogr am r emain lar gel y unclear and r equir e ex-
tensiv e inv estigations. On the one hand, changes in the gut mi-
crobiota may reshape the host m6A prole and alter the expres-
sion of m6A-modied target genes. On the other hand, these m6A-
modied genes may regulate host defense and inuence gut mi-
crobiota composition. The interplay between the gut microbiota
and RNA m6A modication could potentially inuence cancer de-
velopment by manipulating the cancer cell life cycle and host
anti-tumor immunity. Understanding their interactions will pro-
vide new insights into the mechanistic basis of cancer develop-
ment.
The use of probiotics is potentially benecial to human health.
In preclinical cancer models, probiotics have demonstrated ef-
fectiveness in inducing tumor r egr ession and impr oving surviv al,
both as a standalone ther a py and combined with conventional
Downloaded from https://academic.oup.com/femsre/article/47/4/fuad036/7220014 by The Chinese University of Hong Kong user on 26 July 2023
Su et al. | 11
tr eatments suc h as c hemother a py or imm unother a py. In mouse
models of CRC, Bidobacterium pseudolongum has been found to
enhance the efcacy of ICB (Mager et al. 2020 ). It is worth men-
tioning that a recent study investigated the use of the live bac-
terial product CBM588 as a dietary supplement for patients with
metastatic renal cell carcinoma who wer e r eceiving dual imm une
c hec kpoint bloc kade (Dizman et al . 2022 ). Although the study
had a small sample size, the results sho w ed higher objective re-
sponse rates and prolonged progression-free survival among the
treated patients (Dizman et al . 2022 ). Ther efor e, pr obiotics may
hold promise as a complementary approach to improve host anti-
tumor responses. In addition to probiotics, small-molecule in-
hibitors of m6A regulators have also been developed for cancer
tr eatment. Tar geting m6A r egulators can suppress cancer stem
cell self-r ene wal and cancer cell pr olifer ation while also elicit-
ing robust anti-tumor immune responses in various mouse can-
cer models (Su et al. 2020 , Yank ov a et al. 2021 , Chen et al . 2022b ).
Despite these promising results, it remains unclear how targeting
the m6A writer (METTL3) and erasers (FTO and ALKBH5) could
ac hie v e similar anti-cancer effects . T his ma y be partially due to
their differ ent downstr eam tar gets, and further r esearc h is needed
to elucidate the mechanisms underlying their anti-cancer effects.
Modulating either tumor-intrinsic factors (such as the m6A pro-
gram) or extrinsic factors (such as the microbiota) has been shown
to impr ov e the efcacy of cancer tr eatment and boost anti-tumor
defense mechanisms in preclinical studies. Considering the po-
tential for crosstalk between these factors , it ma y be worthwhile
to explore the synergistic effects of combining probiotics with spe-
cic inhibitors of m6A regulators for cancer tr eatment. Suc h a
combination ther a py could offer a pr omising a ppr oac h to enhanc-
ing the immune response against cancer cells, potentially leading
to impr ov ed outcomes for cancer patients.
Author contributions
HS wrote the manuscript. HC , HCHL, HYC , XTZ, NQ, YFW, MTVC,
and WKKW r e vised the manuscript, and HRC supervised and r e-
vised the paper.
Conict of interest. The authors declared no conict of interest.
Funding
This project was supported by the National Natural Science Foun-
dation of China (NSFC; 82103245, 82272989, 81972576); RGC-GRF
Hong Kong (14111621, 14107321, 14101922); Heath and Medical
Research Fund (HMRF) (18190951, 22210032); and CUHK Direct
Grant for Research (2022.003).
References
An J , Ha EM. Extr acellular v esicles deriv ed fr om Lactobacillus plan-
tarum r estor e c hemosensitivity thr ough the PDK2-mediated glu-
cose metabolic pathway in 5-FU-resistant colorectal cancer cells.
J Microbiol 2022; 60 :735–45.
Bai X , Wong CC, Pan Y et al. Loss of YTHDF1 in gastric tumors r estor es
sensitivity to antitumor immunity by recruiting mature dendritic
cells. J Immunother Cancer 2022; 10 : e003663.
Baruch EN , Youngster I, Ben-Betzalel G et al. Fecal microbiota
tr ansplant pr omotes r esponse in imm unother a py-r efr actory
melanoma patients. Science 2021; 371 :602–9.
Ber gstr om K , Shan X, Casero D et al. Proximal colon-derived O-
gl ycosylated m ucus enca psulates and modulates the micr obiota.
Science 2020; 370 :467–72.
Beurel E , Jope RS. Lipopol ysacc haride-induced interleukin-6 pr oduc-
tion is controlled by glycogen synthase kinase-3 and ST A T3 in the
brain. J Neuroinammation 2009; 6 :9.
Bhatt AP , Redinbo MR, Bultman SJ. The role of the microbiome in
cancer de v elopment and ther a py. CA Cancer J Clin 2017; 67 :326–
44.
Blekhman R , Goodrich JK, Huang K et al. Host genetic variation im-
pacts microbiome composition across human body sites. Genome
Biol 2015; 16 :191.
Boccaletto P , Mac hnic ka MA, Purta E et al. MODOMICS: a database
of RN A modication pathw a ys . 2017 update . Nucleic Acids Res
2018; 46 :D303–D7.
Brown JA , Kinzig CG, DeGregorio SJ et al. Methyltransferase-
like protein 16 binds the 3’-terminal triple helix of MALAT1
long noncoding RNA. Proc Natl Acad Sci USA 2016; 113 :
14013–8.
Buti L , Spooner E, Va n der Veen AG et al. Helicobacter pylori cytotoxin-
associated gene A (Ca gA) sub v erts the a poptosis-stim ulating pr o-
tein of p53 (ASPP2) tumor suppressor pathway of the host. Proc
Natl Acad Sci USA 2011; 108 :9238–43.
Cao Y , Wang Z, Yan Y et al. Enterotoxigenic bacteroidesfragilis pro-
motes intestinal inammation and malignancy by inhibit-
ing exosome-pac ka ged miR-149-3p. Gastroenterolog y 2021; 161 :
1552–66 e12.
Carlberg C , Munoz A. An update on vitamin D signaling and cancer.
Semin Cancer Biol 2020. https:// doi.org/ 10.1016/ j.semcancer.2020.0
5.018
Chen B , Ye F, Yu L et al. De v elopment of cell-activ e N6-
meth yladenosine RNA demeth ylase FTO inhibitor. J Am Chem Soc
2012; 134 :17963–71.
Chen H , Gao S, Liu W et al. RNA N(6)-meth yladenosine meth yl-
tr ansfer ase METTL3 facilitates colorectal cancer by activating the
m(6)A-GLUT1-mTORC1 axis and is a ther a peutic tar get. Gastroen-
terology 2021; 160 :1284–300 e16.
Chen H , Pan Y, Zhou Q et al. METTL3 inhibits anti-tumor immunity
by targeting m(6)A-BHLHE41-CXCL1/CXCR2 axis to promote col-
or ectal cancer. Gastroenterolog y 2022a. https:// doi.org/ 10.1053/ j.ga
stro.2022.06.024
Chen H , Pan Y, Zhou Q et al. METTL3 Inhibits antitumor immunity
by targeting m(6)A-BHLHE41-CXCL1/CXCR2 axis to promote col-
or ectal cancer. Gastroenterolog y 2022b. https:// doi.org/ 10.1053/ j.
gastro.2022.06.024
Chen M , Wei L, Law CT et al. RNA N6-methyladenosine
methyltr ansfer ase-like 3 pr omotes liv er cancer pr ogr ession
through YTHDF2-dependent posttranscriptional silencing of
SOCS2. Hepatology 2018; 67 :2254–70.
Chen S , Zhang L, Li M et al. Fusobacterium nucleatum re-
duces METTL3-mediated m(6)A modication and con-
tributes to colorectal cancer metastasis. Nat Commun 2022; 13 :
1248.
Chen SL , Liu LL, Wang CH et al. Loss of RDM1 enhances hepatocel-
lular carcinoma pr ogr ession via p53 and Ras/Raf/ERK pathwa ys .
Mol Oncol 2020; 14 :373–86.
Chen X , Sun K, Jiao S et al. High le v els of SIRT1 expression enhance
tumorigenesis and associate with a poor prognosis of colorectal
carcinoma patients. Sci Rep 2014; 4 :7481.
Chen XY , Liang R, Yi YC et al. The m(6)A reader YTHDF1 facilitates
the tumorigenesis and metastasis of gastric cancer via USP14
translation in an m(6)A-dependent manner. Front Cell Dev Biol
2021; 9 :647702.
Downloaded from https://academic.oup.com/femsre/article/47/4/fuad036/7220014 by The Chinese University of Hong Kong user on 26 July 2023
12 | FEMS Microbiology Reviews , 2023, Vo l. 47, No. 4
Chien CS , Li JY, Chien Y et al. METTL3-dependent N(6)-
methyladenosine RNA modication mediates the atherogenic
inammatory cascades in vascular endothelium. Proc Natl Acad
Sci USA 2021; 118 :e2025070118.
Couturier-Maillard A , Secher T, Rehman A et al. NOD2-mediated dys-
biosis predisposes mice to transmissible colitis and colorectal
cancer. J Clin Invest 2013; 123 :700–11.
Croker BA , Kiu H, Nicholson SE. SOCS regulation of the JAK/ST A T sig-
nalling pathway. Semin Cell Dev Biol 2008; 19 :414–22.
Dalmasso G , Cougnoux A, Delmas J et al. The bacterial genotoxin col-
ibactin promotes colon tumor gro wth b y modifying the tumor mi-
cr oenvir onment. Gut Microbes 2014; 5 :675–80.
Dav ar D , Dzutse v AK, McCulloc h JA et al. Fecal micr obiota tr ansplant
ov ercomes r esistance to anti-PD-1 ther a py in melanoma patients.
Science 2021; 371 :595–602.
Dehghani N , Tafvizi F, Jafari P. Cell cycle arrest and anti-cancer po-
tential of probiotic Lactobacillus rhamnosus against HT-29 cancer
cells. Bioimpacts 2021; 11 :245–52.
Dizman N , Meza L, Ber ger ot P et al. Nivolumab plus ipilimumab
with or without live bacterial supplementation in metastatic re-
nal cell carcinoma: a randomized phase 1 trial. Nat Med 2022; 28 :
704–12.
Dolbois A , Bedi RK, Boc henk ov a E et al. 1,4,9-Triazaspir o[5.5]undecan-
2-one deri vati ves as potent and selective METTL3 inhibitors. J Med
Chem 2021; 64 :12738–60.
Dr e wes JL , Chen J, Markham NO et al. Human colon cancer-derived
Clostridioides difcile strains drive colonic tumorigenesis in
mice. Cancer Discov 2022. https:// doi.org/ 10.1158/ 2159- 8290.CD- 2
1-1273
Du L , Li Y, Kang M et al. USP48 Is upregulated by Mettl14 to attenu-
ate hepatocellular carcinoma via regulating SIRT6 stabilization.
Cancer Res 2021; 81 :3822–34.
Du QY , Zhu ZM, Pei DS. The biological function of IGF2BPs and their
role in tumorigenesis. Invest New Drugs 2021; 39 :1682–93.
Gan Y , Li A, Liu J et al. m(6)A-mRNA methylation regulates gene
expression and programmable m(6)A modication of cellular
rnas with CRISPR-Cas13b in renal cell carcinoma. Front Genet
2021; 12 :795611.
Garrett WS . Cancer and the microbiota. Science 2015; 348 :80–6.
Goodman B , Gardner H. The microbiome and cancer. J Pathol
2018; 244 :667–76.
Gur C , Ibrahim Y, Isaacson B et al. Binding of the Fap2 protein of fu-
sobacterium nucleatum to human inhibitory r eceptor TIGIT pr o-
tects tumors from immune cell attack. Immunity 2015; 42 :344–55.
Gutschner T , Hammerle M, Pazaitis N et al. Insulin-like growth
factor 2 mRNA-binding protein 1 (IGF2BP1) is an important
protumorigenic factor in hepatocellular carcinoma. Hepatology
2014; 59 :1900–11.
Han L , Dong L, Leung K et al. METTL16 dri ves leuk emogenesis
and leukemia stem cell self-r ene w al b y r epr ogr amming BCAA
metabolism. Cell Stem Cell 2023; 30 :52–68 e13.
Hansson GC . Mucins and the microbiome. Annu Rev Biochem
2020; 89 :769–93.
Hazards EPB , Ricci A, Allende A et al. Scientic Opinion on the update
of the list of QPS-recommended biological agents intentionally
added to food or feed as notied to EFSA. EFSA J 2017; 15 :e04664.
Heinrich PC , Behrmann I, Haan S et al. Principles of interleukin (IL)-6-
type cytokine signalling and its r egulation. Bioc hem J 2003; 374 :1–
20.
Herr anz D , Mar av er A, Canamer o M et al. SIRT1 pr omotes thyr oid car-
cinogenesis driven by PTEN deciency. Oncogene 2013; 32 :4052–6.
Hou Y , Zhang Q, Pang W et al. YTHDC1-mediated augmentation of
miR-30d in r epr essing pancr eatic tumorigenesis via attenuation
of RUNX1-induced transcriptional activation of Warburg effect.
Cell Death Differ 2021; 28 :3105–24.
Huang H , Weng H, Sun W et al. Recognition of RNA N(6)-
methyladenosine by IGF2BP proteins enhances mRNA stability
and translation. Nat Cell Biol 2018; 20 :285–95.
Huang H , Wen g H, Zhou K et al. Histone H3 trimethylation at lysine
36 guides m(6)A RNA modication co-tr anscriptionall y. Nature
2019; 567 :414–9.
Huang Y , Su R, Sheng Y et al. Small-molecule targeting of onco-
genic FTO demethylase in acute myeloid leukemia. Cancer Cell
2019; 35 :677–91 e10.
Huo FC , Zhu ZM, Zhu WT et al. METTL3-mediated m(6)A methylation
of SPHK2 promotes gastric cancer progression by targeting KLF2.
Oncogene 2021; 40 :2968–81.
Jabs S , Biton A, Becavin C et al. Impact of the gut microbiota on the
m(6)A epitranscriptome of mouse cecum and liver. Nat Commun
2020; 11 :1344.
Jan G , Belzacq AS, Haouzi D et al. Pr opionibacteria induce a poptosis
of colorectal carcinoma cells via short-chain fatty acids acting on
mitochondria. Cell Death Differ 2002; 9 :179–88.
Jang HM , Han SK, Kim JK et al. Lactobacillus sakei alleviates
high-fat-diet-induced obesity and anxiety in mice by induc-
ing AMPK activation and SIRT1 expression and inhibiting gut
micr obiota-mediated NF-ka ppaB activ ation. Mol Nutr Food Res
2019; 63 :e1800978.
Jia G , Fu Y, Zhao X et al. N6-methyladenosine in nuclear RNA is
a major substrate of the obesity-associated FTO. Nat Chem Biol
2011; 7 :885–7.
Jin D , Guo J, Wu Y et al. m(6)A mRNA methylation initiated by METTL3
dir ectl y pr omotes YAP tr anslation and incr eases YAP activity by
regulating the MALA T1-miR-1914-3p-Y AP axis to induce NSCLC
drug resistance and metastasis. J Hematol Oncol 2019; 12 :135.
Jin X , Wei Y, Xu F et al. SIRT1 promotes formation of breast cancer
through modulating Akt activity. J Cancer 2018; 9 :2012–23.
Knights D , Silv erber g MS, Weersma RK et al. Complex host genetics
inuence the microbiome in inammatory bo w el disease. Genome
Med 2014; 6 :107.
Koh A , De Va dde r F, K o v atc he v a-Datc hary P et al. From dietary
Fiber to host physiology: short-chain fatty acids as k e y bacterial
metabolites. Cell 2016; 165 :1332–45.
Konishi H , Fujiya M, Tanaka H et al. Pr obiotic-deriv ed ferric hr ome in-
hibits colon cancer pr ogr ession via JNK-mediated apoptosis. Nat
Commun 2016; 7 :12365.
Lang C , Yin C, Lin K et al. m(6) A modication of lncRNA PCAT6
promotes bone metastasis in prostate cancer through IGF2BP2-
mediated IGF1R mRNA stabilization. Clin Tra n s l Med 2021; 11 :e426.
Larsson BM , Larsson K, Malmberg P et al. Gram positive bacteria in-
duce IL-6 and IL-8 production in human alveolar macrophages
and epithelial cells. Inammation 1999; 23 :217–30.
Lee JW , Shin JG, Kim EH et al. Immunomodulatory and antitumor
effects in vivo by the cytoplasmic fraction of Lactobacillus casei
and Bidobacterium longum. J Vet Sci 2004; 5 :41–8.
Leon-Coria A , Kumar M, Workentine M et al. Muc2 Mucin and non-
m ucin micr obiota confer distinct innate host defense in dis-
ease susceptibility and colonic injury. Cell Mol Gastroenterol Hepatol
2021; 11 :77–98.
Li H , Zhang N, Jiao X et al. Downregulation of microRNA-6125
pr omotes color ectal cancer gr owth thr ough YTHDF2-dependent
recognition of N6-methyladenosine-modied GSK3beta. Clin
Tra n s l Med 2021; 11 :e602.
Li HB , Ton g J, Zhu S et al. m(6)A mRNA methylation controls T cell
homeostasis by targeting the IL-7/ST A T5/SOCS pathways. Nature
2017; 548 :338–42.
Downloaded from https://academic.oup.com/femsre/article/47/4/fuad036/7220014 by The Chinese University of Hong Kong user on 26 July 2023
Su et al. | 13
Li N , Kang Y, Wan g L et al. ALKBH5 regulates anti-PD-1 therapy re-
sponse by modulating lactate and suppr essiv e imm une cell ac-
cumulation in tumor microenvironment. Proc Natl Acad Sci USA
2020; 117 :20159–70.
Li Q , Hu W, Liu WX et al. Streptococcus thermophilus inhibits colorec-
tal tumorigenesis through secreting beta-galactosidase. Gastroen-
terology 2021; 160 :1179–93 e14.
Li Q , Li Y, Wang Y et al. Or al administr ation of Bidobacterium
br e v e pr omotes antitumor efcacy via dendritic cells-deriv ed in-
terleukin 12. Oncoimmunology 2021; 10 :1868122.
Liang L , Ai L, Qian J et al. Long noncoding RNA expression proles in
gut tissues constitute molecular signatures that reect the types
of microbes. Sci Rep 2015; 5 :11763.
Liu X , Liu J, Xiao W et al. SIRT1 Regulates N(6) -methyladenosine
RN A modication in hepatocar cinogenesis b y inducing RANBP2-
dependent FTO sumoylation. Hepatology 2020; 72 :2029–50.
Liu X , Qin J, Gao T et al. YTHDF1 Facilitates the pr ogr ession of hepa-
tocellular car cinoma b y promoting FZD5 mRN A translation in an
m6A-dependent manner. Mol Ther Nucleic Acids 2020; 22 :750–65.
Liu X , Su K, Sun X et al. Sec62 pr omotes stemness and c hemor esis-
tance of human colorectal cancer through activating wnt/beta-
catenin pathway. J Exp Clin Cancer Res 2021; 40 :132.
Liu Y , Song R, Zhao L et al. m(6)A demethylase ALKBH5 is r equir ed for
antibacterial innate defense by intrinsic motivation of neutrophil
migration. Signal Transduct Targ e t Ther 2022; 7 :194.
Long X , Wo ng CC, To n g L et al. Peptostreptococcus anaerobius promotes
colorectal carcinogenesis and modulates tumour immunity. Nat
Microbiol 2019; 4 :2319–30.
Luo ZW , Xia K, Liu YW et al. Extracellular vesicles from Akkermansia
muciniphila elicit antitumor immunity against prostate cancer via
modulation of CD8( + ) T cells and macr opha ges. Int J Nanomedicine
2021; 16 :2949–63.
Ma H , Wang X, Cai J et al. N(6-)methyladenosine methyltr ansfer ase
ZCCHC4 mediates ribosomal RNA methylation. Nat Chem Biol
2019; 15 :88–94.
Ma L , Chen T, Zhang X et al. The m(6)A reader YTHDC2
inhibits lung adenocarcinoma tumorigenesis by suppressing
SLC7A11-de pendent antio xidant function. Redox Biol 2021; 38 :
101801.
Mager LF , Burkhard R, Pett N et al. Micr obiome-deriv ed inosine mod-
ulates response to checkpoint inhibitor immunotherapy. Science
2020; 369 :1481–9.
McGuc kin MA , Ev ery AL, Skene CD et al. Muc1 mucin limits both He-
licobacter pylori colonization of the murine gastric mucosa and
associated gastritis. Gastroenterology 2007; 133 :1210–8.
Mego M , Ebringer L, Drgona L et al. Pr e v ention of febrile neutropenia
in cancer patients by probiotic strain Enterococcus faecium M-74.
Pilot study phase I. Neoplasma 2005; 52 :159–64.
Moroz-Omori EV , Huang D, Kumar Bedi R et al. METTL3 Inhibitors
for epitranscriptomic modulation of cellular processes. ChemMed-
Chem 2021; 16 :3035–43.
Moss SF . The clinical evidence linking Helicobacter pylori to gastric
cancer. Cell Mol Gastroenterol Hepatol 2017; 3 :183–91.
Osterlund P , Ruotsalainen T, Korpela R et al. Lactobacillus supple-
mentation for diarrhoea related to chemotherapy of colorectal
cancer: a randomised study. Br J Cancer 2007; 97 :1028–34.
Ou B , Liu Y, Ya n g X et al. C5aR1-positive neutrophils promote breast
cancer gl ycol ysis thr ough WTAP-dependent m6A methylation of
ENO1. Cell Death Dis 2021; 12 :737.
Pan J , Liu F, Xiao X et al. METTL3 pr omotes color ectal carcinoma pr o-
gression by regulating the m6A-CRB3-Hippo axis. J Exp Clin Cancer
Res 2022; 41 :19.
Paone P , Cani PD. Mucus barrier, mucins and gut microbiota: the ex-
pected slimy partners? Gut 2020; 69 :2232–43.
Pendleton KE , Chen B, Liu K et al. The U6 snRNA m(6)A methyltrans-
ferase METTL16 regulates SAM synthetase intron retention. Cell
2017; 169 :824–35 e14.
Peng W , Li J, Chen R et al. Upregulated METTL3 promotes metastasis
of colorectal cancer via miR-1246/SPRED2/MAPK signaling path-
way. J Exp Clin Cancer Res 2019; 38 :393.
P okro vsky VS , Anisimo va N Yu , Da vydo v D Zh et al. Methionine
gamma l yase fr om Clostridium spor ogenes incr eases the anti-
cancer effect of doxorubicin in A549 cells and human cancer
xenografts. Invest New Drugs 2019; 37 :201–9.
Pothuraju R , Krishn SR, Gautam SK et al. Mechanistic and functional
shades of mucins and associated glycans in colon cancer. Cancers
(Basel) 2020; 12 :649.
Qiu X , Yang S, Wang S et al. M(6)A demethylase ALKBH5 regulates
PD-L1 expression and tumor immunoenvironment in intrahep-
atic cholangiocarcinoma. Cancer Res 2021; 81 :4778–93.
Qu J , Hou Y, Chen Q et al. RNA demethylase ALKBH5 promotes
tumorigenesis in multiple myeloma via TRAF1-mediated acti-
vation of NF-kappaB and MAPK signaling pathwa ys . Oncogene
2022; 41 :400–13.
Rahimpour M , Ashabi G, Rahimi AM et al. Lactobacillus rham-
nosus R0011 Treatment Enhanced Efcacy of Capecitabine
against Colon Cancer in Male Balb/c Mice. Nutr Cancer 2022; 74 :
2622–31.
Rong D , Wu F, Lu C et al. m6A modication of circHPS5 and hepato-
cellular carcinoma pr ogr ession thr ough HMGA2 expression. Mol
Ther Nucleic Acids 2021; 26 :637–48.
Ruan DY , Li T, Wan g YN et al. FTO downregulation mediated
by hypoxia facilitates colorectal cancer metastasis. Oncogene
2021; 40 :5168–81.
Ruan F , Liu C, Wan g Y et al. Role of RNA m(6)A modication in tita-
nium dioxide nanoparticle-induced acute pulmonary injury: an
in vitro and in vivo study. Environ Pollut 2022; 311 :119986.
Rubinstein MR , Wang X, Liu W et al. Fusobacterium nuclea-
tum pr omotes color ectal car cinogenesis b y modulating E-
cadherin/beta-catenin signaling via its FadA adhesin. Cell Host Mi-
crobe 2013; 14 :195–206.
Satterwhite ER , Manseld KD. RNA methyltr ansfer ase METTL16: tar-
gets and function. Wiley Interdiscip Rev RNA 2022; 13 :e1681.
Sc hneider H , Ber ger E, Dolan B et al. The human tr ansmembr ane
m ucin MUC17 r esponds to TNFalpha by incr eased pr esentation
at the plasma membr ane. Bioc hem J 2019; 476 :2281–95.
Sender R , Fuchs S, Milo R. Revised estimates for the number of Hu-
man and bacteria cells in the body. PLoS Biol 2016; 14 :e1002533.
Sheng Y , Wei J, Yu F et al. A critical role of nuclear m6A
reader YTHDC1 in leukemogenesis by regulating MCM complex-
mediated DNA replication. Blood 2021; 138 :2838–52.
Shi H , Wang X, Lu Z et al. YTHDF3 facilitates translation and de-
cay of N(6)-methyladenosine-modied RNA. Cell Res 2017; 27 :
315–28.
Si W , Liang H, Bugno J et al. Lactobacillus rhamnosus GG induces
cGAS/STING- dependent type I interferon and improves response
to immune checkpoint blockade. Gut 2022; 71 :521–33.
Singh J , Rivenson A, Tomita M et al. Bidobacterium longum, a lac-
tic acid-producing intestinal bacterium inhibits colon cancer and
modulates the intermediate biomarkers of colon carcinogenesis.
Carcinogenesis 1997; 18 :833–41.
Song S , Byrd JC, Mazurek N et al. Galectin-3 modulates MUC2 mucin
expression in human colon cancer cells at the level of transcrip-
tion via AP-1 activ ation. Gastroenterolog y 2005; 129 :1581–91.
Downloaded from https://academic.oup.com/femsre/article/47/4/fuad036/7220014 by The Chinese University of Hong Kong user on 26 July 2023
14 | FEMS Microbiology Reviews , 2023, Vo l. 47, No. 4
Spencer L , Shultz L, Rajan TV. Interleukin-4 receptor-Stat6 signaling
in murine infections with a tissue-dwelling nematode parasite.
Infect Immun 2001; 69 :7743–52.
Su R , Dong L, Li C et al. R-2HG exhibits anti-tumor activity by target-
ing FTO/m(6)A/MYC/CEBPA signaling. Cell 2018; 172 :90–105 e23.
Su R , Dong L, Li Y et al. Targeting FTO suppresses cancer stem cell
maintenance and immune evasion. Cancer Cell 2020; 38 :79–96 e11.
Su T , Huang M, Liao J et al. Insufcient Radiofrequency Ablation
Promotes Hepatocellular Carcinoma Metastasis Through N6-
Meth yladenosine mRNA Meth ylation-Dependent Mechanism.
Hepatology 2021; 74 :1339–56.
Suganuma M , Watanabe T, Sueoka E et al. Role of TNF-alpha-
inducing pr otein secr eted b y Helicobacter p ylori as a tumor pro-
moter in gastric cancer and emerging preventive strategies. Tox ins
(Basel) 2021; 13 :181.
Sugim ur a N , Li Q, Chu ESH et al. Lactobacillus gallinarum mod-
ulates the gut microbiota and produces anti-cancer metabo-
lites to protect against colorectal tumourigenesis. Gut 2021; 71 :
2011–21.
Sun T , Wu R, Ming L. The role of m6A RNA methylation in cancer.
Biomed Pharmacother 2019; 112 :108613.
Tahar a T , Yamamoto E, Suzuki H et al. Fusobacterium in colonic
ora and molecular features of colorectal carcinoma. Cancer Res
2014; 74 :1311–8.
Taka gi A , Ikem ur a H, Matsuzaki T et al. Relationship between the in
vitr o r esponse of dendritic cells to Lactobacillus and pr e v ention
of tumorigenesis in the mouse. J Gastroenterol 2008; 43 :661–9.
T anabe A , T anikaw a K, Tsunetomi M et al. RN A helicase YTHDC2 pro-
motes cancer metastasis via the enhancement of the efciency
by which HIF-1alpha mRNA is translated. Cancer Lett 2016; 376 :34–
42.
Tao L , Mu X, Chen H et al. FTO modies the m6A le v el of
MALAT and promotes bladder cancer progression. Clin Tr a n s l Med
2021; 11 :e310.
Tsai YS , Chen YP, Lin SW et al. Lactobacillus rhamnosus GKLC1
ameliorates cisplatin-induced chronic ne phroto xicity by in-
hibiting cell inammation and apoptosis. Biomed Pharmacother
2022; 147 :112701.
Tul k e n s J , Ver ga uwen G, Van Deun J et al. Incr eased le v els of systemic
LPS-positive bacterial extracellular vesicles in patients with in-
testinal barrier dysfunction. Gut 2020; 69 :191–3.
Waldner MJ , Foersch S, Neurath MF. Interleukin-6–a k e y regulator of
colorectal cancer development. Int J Biol Sci 2012; 8 :1248–53.
Wan g J , Thingholm LB, Skieceviciene J et al. Genome-wide association
anal ysis identies v ariation in vitamin D r eceptor and other host
factors inuencing the gut microbiota. Nat Genet 2016; 48 :1396–
406.
Wan g KK , He KY, Ya n g JY et al. Lactobacillus suppresses tumorigen-
esis of oropharyngeal cancer via enhancing anti-tumor immune
response . F ront Cell Dev Biol 2022; 10 :842153.
W ang Q , W ang K, W u W et al. Administration of bidobacterium bi-
dum CGMCC 15068 modulates gut microbiota and metabolome
in azoxymethane (AOM)/dextran sulphate sodium (DSS)-induced
colitis-associated colon cancer (CAC) in mice. Appl Microbiol
Biotechnol 2020; 104 :5915–28.
Wan g S , Gao S, Zeng Y et al. N6-Methyladenosine reader YTHDF1
promotes ARHGEF2 translation and RhoA signaling in colorectal
cancer. Gastroenterology 2021. https:// doi.org/ 10.1053/ j.gastro.202
1.12.269
Wan g W , Liu F, Xu C et al. Lactobacillus plantarum 69-2 combined
with galacto-oligosaccharides alleviates d-galactose-induced ag-
ing by regulating the AMPK/SIRT1 signaling pathway and gut mi-
crobiota in mice. J Agric Food Chem 2021; 69 :2745–57.
Wan g X , Li Y, Chen W et al. Tr anscriptome-wide r epr ogr amming of
N(6)-methyladenosine modication by the mouse microbiome.
Cell Res 2019; 29 :167–70.
Wan g X , Zhao BS, Roundtree IA et al. N(6)-methyladenosine modu-
lates messenger RNA translation efciency. Cell 2015; 161 :1388–
99.
Wan g Y , Chen J, Gao WQ et al. METTL14 pr omotes pr ostate tumorige-
nesis by inhibiting THBS1 via an m6A-YTHDF2-dependent mech-
anism. Cell Death Discov 2022; 8 :143.
Wei W , Sun J, Zhang H et al. Circ0008399 Interaction with WTAP
pr omotes assembl y and activity of the m(6)A methyltr ansfer ase
complex and promotes cisplatin resistance in bladder cancer.
Cancer Res 2021; 81 :6142–56.
Wen g H , Huang H, Wu H et al. METTL14 Inhibits hematopoi-
etic stem/pr ogenitor differ entiation and pr omotes leukemoge-
nesis via mRNA m(6)A modication. Cell Stem Cell 2018; 22 :
191–205 e9.
Wolk K , Kunz S, Witte E et al. IL-22 increases the innate immunity of
tissues. Immunity 2004; 21 :241–54.
Woo V , Alenghat T. Host-micr obiota inter actions: epigenomic r egu-
lation. Curr Opin Immunol 2017; 44 :52–60.
Wu A , Hu Y, Xu Y et al. Methyltr ansfer ase-like 3-mediated m6A
methylation of Hsa_circ_0058493 accelerates hepatocellular car-
cinoma pr ogr ession by binding to YTH domain-containing pro-
tein 1. Front Cell Dev Biol 2021; 9 :762588.
Wu C , Chen W, He J et al. Interplay of m(6)A and H3K27 trimethy-
lation r estr ains inammation during bacterial infection. Sci Adv
2020; 6 :eaba0647.
Wu J , Li Q, Fu XS. Fusobacterium nucleatum contributes to the car-
cinogenesis of colorectal cancer by inducing inammation and
suppressing host immunity. Tr a n s l Oncol 2019; 12 :846–51.
Wu J , Zhao Y, Wa ng X et al. Dietary nutrients shape gut microbes and
intestinal mucosa via epigenetic modications. Crit Rev Food Sci
Nutr 2020; https://doi.org/10.1080/10408398.2020.1828813: 1-15.
Wu N , Feng YQ, Lyu N et al. Fusobacterium nucleatum promotes
colon cancer pr ogr ession by c hanging the m ucosal micr obiota
and colon transcriptome in a mouse model. Wor ld J Gastroenterol
2022; 28 :1981–95.
Wu R , Liu Y, Zhao Y et al. m(6)A methylation controls pluripo-
tenc y of por cine induced pluripotent stem cells by targeting
SOCS3/JAK2/ST A T3 pathway in a YTHDF1/YTHDF2-orc hestr ated
manner. Cell Death Dis 2019; 10 :171.
Wu S , Rhee KJ, Albesiano E et al. A human colonic commensal pro-
motes colon tumorigenesis via activation of T helper type 17 T
cell responses. Nat Med 2009; 15 :1016–22.
Wu S , Rhee KJ, Zhang M et al. Bacteroides fragilis toxin stimu-
lates intestinal epithelial cell shedding and gamma-secretase-
dependent E-cadherin cleav a ge. J Cell Sci 2007; 120 :1944–52.
Wu S , Zhang YG, Lu R et al. Intestinal epithelial vitamin D receptor
deletion leads to defectiv e autopha gy in colitis. Gut 2015; 64 :1082–
94.
Xie G , Wu XN, Ling Y et al. A novel inhibitor of N (6)-methyladenosine
demethylase FTO induces mRNA methylation and shows anti-
cancer activities. Acta Pharm Sin B 2022; 12 :853–66.
Xue L , Li J, Lin Y et al. m(6) A tr ansfer ase METTL3-induced lncRNA
ABHD11-AS1 promotes the Warburg effect of non-small-cell lung
cancer. J Cell Physiol 2021; 236 :2649–58.
Yan g B , Chen Q. Cross-talk between o xidati ve stress and m(6)A
RNA methylation in cancer. Oxid Med Cell Longev 2021; 2021 :
6545728.
Yan g L , Lin S, Kang Y et al. Rhein sensitizes human pancreatic cancer
cells to EGFR inhibitors by inhibiting ST A T3 pathway. J Exp Clin
Cancer Res 2019; 38 :31.
Downloaded from https://academic.oup.com/femsre/article/47/4/fuad036/7220014 by The Chinese University of Hong Kong user on 26 July 2023
Su et al. | 15
Yan g Z , Cai Z, Ya n g C et al. ALKBH5 regulates STAT 3 activity
to affect the pr olifer ation and tumorigenicity of osteosarcoma
via an m6A-YTHDF2-dependent manner. EBioMedicine 2022; 80 :
104019.
Yan k ov a E , Blac kaby W, Albertella M et al. Small-molecule inhibi-
tion of METTL3 as a strategy against myeloid leukaemia. Nature
2021; 593 :597–601.
Ye H , Chen T, Zeng Z et al. The m6A writers regulated by the IL-
6/ST A T3 inammatory pathway facilitate cancer cell stemness
in cholangiocarcinoma. Cancer Biol Med 2021. https:// doi.org/ 10.2
0892/j.issn.2095-3941.2020.0661
Yu T , Chen X, Lin T et al. KLF4 deletion alters gastric cell lineage and
induces MUC2 expression. Cell Death Dis 2016; 7 :e2255.
Yua n H , Wa ng Z, Li L et al. Activ ation of str ess r esponse gene
SIR T1 b y BCR-ABL promotes leukemogenesis. Blood 2012; 119 :
1904–14.
Yue B , Song C, Ya ng L et al. METTL3-mediated N6-methyladenosine
modication is critical for epithelial-mesenchymal transition
and metastasis of gastric cancer. Mol Cancer 2019; 18 :142.
Yue C , Chen J, Li Z et al. microRNA-96 promotes occurrence and pro-
gression of colorectal cancer via regulation of the AMPKalpha2-
FTO-m6A/MYC axis. J Exp Clin Cancer Res 2020; 39 :240.
Yue Y , Wan g S, Shi J et al. Effects of Lactobacillus acidophilus
KLDS1.0901 on pr olifer ation and apoptosis of colon cancer cells.
Front Microbiol 2021; 12 :788040.
Zaccara S , Ries RJ, Jaffrey SR Reading, writing and erasing mRNA
methylation. Nat Rev Mol Cell Biol 2019; 20 :608–24.
Zhang C , Huang S, Zhuang H et al. YTHDF2 promotes the liver cancer
stem cell phenotype and cancer metastasis by regulating OCT4
expression via m6A RNA methylation. Oncogene 2020; 39 :4507–18.
Zhang H , Yi JK, Huang H et al. Rhein suppr esses color ectal cancer
cell gro wth b y inhibiting the mTOR pathw ay In vitro and In vivo.
Cancers (Basel) 2021; 13 :2176.
Zhang J , Zhang Y, Xia Y et al. Imbalance of the intestinal virome and
alter ed vir al-bacterial inter actions caused by a conditional dele-
tion of the vitamin D receptor. Gut Microbes 2021; 13 :1957408.
Zhang M , Chen Y, Chen H et al. Tanshinone IIA alleviates cardiac hy-
pertr ophy thr ough m6A modication of galectin-3. Bioengineered
2022; 13 :4260–70.
Zhang YG , Lu R, Wu S et al. Vitamin D receptor protects against dys-
biosis and tumorigenesis via the JA K/ST A T pathwa y in Intestine .
Cell Mol Gastroenterol Hepatol 2020; 10 :729–46.
Zhao W , Xie Y. KIAA1429 pr omotes the pr ogr ession of lung adeno-
car cinoma b y regulating the m6A le v el of MUC3A. Pathol Res Pract
2021; 217 :153284.
Zheng G , Cox T, Tribbey L et al. Synthesis of a FTO inhibitor with
anticonvulsant activity. ACS Chem Neurosci 2014; 5 :658–65.
Zheng J , Wittouck S, Salvetti E et al. A taxonomic note on the
genus Lactobacillus: description of 23 novel genera, emended de-
scription of the genus Lactobacillus Beijerinck 1901, and union
of Lactobacillaceae and Leuconostocaceae . Int J Syst Evol Microbiol
2020; 70 :2782–858.
Zhong L , Liao D, Zhang M et al. YTHDF2 suppresses cell proliferation
and growth via destabilizing the EGFR mRNA in hepatocellular
carcinoma. Cancer Lett 2019; 442 :252–61.
Zhou D , Ta n g W, Xu Y et al. METTL3/YTHDF2 m6A axis acceler-
ates colorectal carcinogenesis through epigenetically suppress-
ing YPEL5. Mol Oncol 2021; 15 :2172–84.
Zhou T , Li S, Xiang D et al. m6A RNA methylation-mediated
HNF3gamma reduction renders hepatocellular carcinoma dedif-
fer entiation and sor afenib r esistance. Signal T ransduct T arget Ther
2020; 5 :296.
Zhou Y , Wan g Q, Deng H et al. N6-methyladenosine demethylase
FTO pr omotes gr owth and metastasis of gastric cancer via m(6)A
modication of caveolin-1 and metabolic regulation of mitochon-
drial dynamics. Cell Death Dis 2022; 13 :72.
Zhu L , Fu J, Xiao X et al. Faecal microbiota transplantation-mediated
jejunal microbiota changes halt high-fat diet-induced obesity
in mice via retarding intestinal fat absorption. Microb Biotechnol
2022; 15 :337–52.
Zitvogel L , Daillere R, Roberti MP et al. Anticancer effects of the mi-
crobiome and its products. Nat Rev Microbiol 2017; 15 :465–78.
Zong X , Wan g H, Xiao X et al. Enter otoxigenic Esc heric hia coli infection
promotes enteric defensin expression via FOXO6-METTL3-m(6)A-
GPR161 signalling axis. RNA Biol 2021; 18 :576–86.
Zuo X , Chen Z, Gao W et al. M6A-mediated upregulation of LINC00958
increases lipogenesis and acts as a nanother a peutic tar get in
hepatocellular carcinoma. J Hematol Oncol 2020; 13 :5.
Recei v ed 22 August 2022; revised 23 June 2023; accepted 28 June 2023
©The Author(s) 2023. Published by Oxford Uni v ersity Pr ess on behalf of FEMS. All rights r eserv ed. For permissions, please e-mail: journals.permissions@oup.com
Downloaded from https://academic.oup.com/femsre/article/47/4/fuad036/7220014 by The Chinese University of Hong Kong user on 26 July 2023
... The human intestinal tract hosts trillions of crucial microorganisms that contribute to nutrition, infection risk reduction, and autoimmune disorder prevention [68,69]. Research has highlighted the significant influence of both gut microbiota and m6A modification on the development of cancer, hinting at the possible interplay between these two factors within the disease's progression [70]. Notably, the presence of m6A methylation on bacterial genomes has been identified, which suggests a potential link between m6A modification and the integrity of the gut microbiota barrier [71][72][73]. ...
Article
Full-text available
The intestinal lumen acts as a critical interface connecting the external environment with the body’s internal state. It’s essential to prevent the passage of harmful antigens and bacteria while facilitating nutrient and water absorption. The intestinal barriers encompass microbial, mechanical, immunological, and chemical elements, working together to maintain intestinal balance. Numerous studies have associated m6A modification with intestinal homeostasis. This review comprehensively outlines potential mechanisms through which m6A modification could initiate, exacerbate, or sustain barrier damage from an intestinal perspective. The pivotal role of m6A modification in preserving intestinal equilibrium provides new insights, guiding the exploration of m6A modification as a target for optimizing preventive and therapeutic strategies for intestinal homeostasis.
Chapter
RNA epitranscriptomic markers, such as N6-methyladenosine (m6A) and N1-methyladenosine (m1A), are essential for controlling RNA metabolism including splicing, transport, decay, and translation. Mounting evidence suggests that dysregulation of RNA modifications plays a critical part in governing the behavior of cancer cells, encompassing maintenance of stemness, hyperproliferation, invasion, metastasis, metabolic reprogramming, and immune evasion. This chapter explores the emerging understanding of how RNA modifications contribute to these critical aspects of cancer cell behavior, with a particular focus on gastrointestinal (GI) cancers that represent over a quarter of all cancers. It also highlights the potential of targeting RNA modifications as therapeutic interventions in different GI cancer types based on the published studies. Understanding the pivotal role of RNA modifications in the pathogenesis of GI cancers offers promising avenues for developing novel and personalized cancer therapies.
Article
Full-text available
Neutrophil migration into the site of infection is necessary for antibacterial innate defense, whereas impaired neutrophil migration may result in excessive inflammation and even sepsis. The neutrophil migration directed by extracellular signals such as chemokines has been extensively studied, yet the intrinsic mechanism for determining neutrophil ability to migrate needs further investigation. N6-methyladenosine (m6A) RNA modification is important in immunity and inflammation, and our preliminary data indicate downregulation of RNA m6A demethylase alkB homolog 5 (ALKBH5) in neutrophils during bacterial infection. Whether m6A modification and ALKBH5 might intrinsically modulate neutrophil innate response remain unknown. Here we report that ALKBH5 is required for antibacterial innate defense by enhancing intrinsic ability of neutrophil migration. We found that deficiency of ALKBH5 increased mortality of mice with polymicrobial sepsis induced by cecal ligation and puncture (CLP), and Alkbh5-deficient CLP mice exhibited higher bacterial burden and massive proinflammatory cytokine production in the peritoneal cavity and blood because of less neutrophil migration. Alkbh5-deficient neutrophils had lower CXCR2 expression, thus exhibiting impaired migration toward chemokine CXCL2. Mechanistically, ALKBH5-mediated m6A demethylation empowered neutrophils with high migration capability through altering the RNA decay, consequently regulating protein expression of its targets, neutrophil migration-related molecules, including increased expression of neutrophil migration-promoting CXCR2 and NLRP12, but decreased expression of neutrophil migration-suppressive PTGER4, TNC, and WNK1. Our findings reveal a previously unknown role of ALKBH5 in imprinting migration-promoting transcriptome signatures in neutrophils and intrinsically promoting neutrophil migration for antibacterial defense, highlighting the potential application of targeting neutrophil m6A modification in controlling bacterial infections.
Article
Full-text available
Background & Aims N⁶-Methyladenosine (m⁶A) is the most prevalent RNA modification and recognized as an important epitranscriptomic mechanism in colorectal cancer (CRC). We aim to exploit whether and how tumor-intrinsic m⁶A modification drove by methyltransferase like 3 (METTL3) can dictate the immune landscape of CRC. Methods Mettl3 knockout mice, CD34⁺ humanized mice and different syngeneic mice models were employed. Immune cells composition and cytokines level were analyzed by flow cytometry and Cytokine 23-Plex immunoassay, respectively. M⁶A-seq and RNA-seq were performed to identify downstream targets and pathways of METTL3. Human CRC specimens (n=176) were used to evaluate correlation between METTL3 expression and myeloid-derived suppressor cells (MDSCs) infiltration. Results We demonstrated that silencing of METTL3 in CRC cells reduced MDSCs accumulation to sustain activation and proliferation of CD4⁺ and CD8⁺ T cell, and eventually suppressed CRC in ApcMin/+Mettl3+/- mice, CD34⁺ humanized mice and syngeneic mice models. Mechanistically, METTL3 activated m⁶A-BHLHE41-CXCL1 axis by analysis of m⁶A-seq, RNA-seq and cytokines arrays. METTL3 promoted BHLHE41 expression in m⁶A-dependent manner, which subsequently induced CXCL1 transcription to enhance MDSC migration in vitro. However, the effect was negligible upon BHLHE41 depletion, CXCL1 protein or CXCR2 inhibitor SB265610 administration, inferring that METTL3 promotes MDSC migration via BHLHE41-CXCL1/CXCR2. Consistently, depletion of MDSCs by anti-Gr1 antibody or SB265610 blocked tumor-promoting effect of METTL3 in vivo. Importantly, targeting METTL3 by METTL3-sgRNA or specific inhibitor potentiated the effect of anti-PD1 treatment. Conclusions Our study identifies METTL3 as a potential therapeutic target for CRC immunotherapy whose inhibition reverses immune suppression through m⁶A-BHLHE41-CXCL1 axis. METTL3 inhibition plus anti-PD-1 treatment show promising antitumor efficacy against CRC.
Article
Full-text available
Defining the complex role of the microbiome in colorectal cancer and the discovery of novel, protumorigenic microbes are areas of active investigation. In the present study, culturing and reassociation experiments revealed that toxigenic strains of Clostridioides difficile drove the tumorigenic phenotype of a subset of colorectal cancer patient–derived mucosal slurries in germ-free ApcMin/+ mice. Tumorigenesis was dependent on the C. difficile toxin TcdB and was associated with induction of Wnt signaling, reactive oxygen species, and protumorigenic mucosal immune responses marked by the infiltration of activated myeloid cells and IL17-producing lymphoid and innate lymphoid cell subsets. These findings suggest that chronic colonization with toxigenic C. difficile is a potential driver of colorectal cancer in patients. Significance Colorectal cancer is a leading cause of cancer and cancer-related deaths worldwide, with a multifactorial etiology that likely includes procarcinogenic bacteria. Using human colon cancer specimens, culturing, and murine models, we demonstrate that chronic infection with the enteric pathogen C. difficile is a previously unrecognized contributor to colonic tumorigenesis.
Article
Full-text available
Background: Fusobacterium nucleatum (F. nucleatum) has long been known to cause opportunistic infections and has recently been implicated in colorectal cancer (CRC), which has attracted broad attention. However, the mechanism by which it is involved in CRC development is not fully understood. Aim: To explore its potential causative role in CRC development, we evaluated the colon pathology, mucosa barrier, colon microbiota and host transcriptome profile after F. nucleatum infection in an azoxymethane/dextran sulfate sodium salt (AOM/DSS) mouse model. Methods: Three groups of mice were compared to reveal the differences, i.e., the control, AOM/DSS-induced CRC and AOM/DSS-FUSO infection groups. Results: Both the AOM/DSS and AOM/DSS-FUSO groups exhibited a significantly reduced body weight and increased tumor numbers than the control group, and AOM/DSS mice with F. nucleatum infection showed the highest tumor formation ratio among the three groups. Moreover, the colon pathology was the most serious in the AOM/DSS-FUSO group. We found that the structure of the colon microbiota changed considerably after F. nucleatum infection; striking differences in mucosal microbial population patterns were observed between the AOM/DSS-FUSO and AOM/DSS groups, and inflammation-inducing bacteria were enriched in the mucosal microbiota in the AOM/DSS-FUSO group. By comparing intestinal transcriptomics data from AOM vs AOM/DSS-FUSO mice, we showed that transcriptional activity was strongly affected by dysbiosis of the gut microbiota. The most microbiota-sensitive genes were oncogenes in the intestine, and the cyclic adenosine monophosphate signaling pathway, neuroactive ligand-receptor interaction, PPAR signaling pathway, retinol metabolism, mineral absorption and drug metabolism were highly enriched in the AOM/DSS-FUSO group. Additionally, we showed that microbial dysbiosis driven by F. nucleatum infection enriched eight taxa belonging to Proteobacteria, which correlates with increased expression of oncogenic genes. Conclusion: Our study demonstrated that F. nucleatum infection altered the colon mucosal microbiota by enriching pathogens related to the development of CRC, providing new insights into the role of F. nucleatum in the oncogenic microbial environment of the colon.
Article
Full-text available
Background: N6-methyladenosine (m6A) is the most common and abundant mRNA modification and it plays crucial roles in many biological processes. However, as a key RNA demethylase, alkylation repair homolog protein 5 (ALKBH5) has not been well studied in human osteosarcoma. The present study sought to explore ALKBH5-mediated m6A modification and the underlying mechanisms in human osteosarcoma. Methods: The expression of ALKBH5 and its correlation with clinicopathological features were examined by bioinformatics analysis and tissue microarrays. Cellular proliferation was detected by CCK8 assays. Cell cycle and apoptosis were analyzed by TUNEL and Flow cytometry assay. Finally, investigation of the regulatory mechanism of ALKBH5 in human osteosarcoma was performed by MeRIP assay, RNA-sequencing, dual luciferase reporter assay, RNA pull-down and RNA stability assay. Tumor xenograft models were established for in vivo experiments. Findings: Our data showed that low expression of ALKBH5 was associated with worse overall survival for osteosarcoma patients. Reducing m6A mRNA levels in human osteosarcoma cells through ALKBH5 up-regulation lead to cell proliferation inhibition, cell apoptosis and cycle arrest. We identified SOCS3, a negative regulator of STAT3, as a downstream target of ALKBH5-mediated m6A modification. And the m6A modified SOCS3 mRNA was recognized by YTHDF2, which promotes the decay of SOCS3. Mechanistically, our data revealed that ALKBH5 inactivated STAT3 pathway by increasing SOCS3 expression via an m6A-YTHDF2-dependent manner. Interpretation: M6A methylation is rising as a pathway affecting tumorigenicity and tumor progression. Our findings illuminate the clinical significance of ALKBH5-mediated m6A modification in human osteosarcoma and the regulatory mechanisms underlying tumor proliferation and growth, suggesting that ALKBH5 is a potential biomarker for treatment in human osteosarcoma. Funding: This work was supported by and Science and Technology foundation of Hubei, China (Grant No.2017CFB762); the Tongji hospital foundation (Grant No.2201103013); and the National Natural Science Foudation of China (No.82002849).
Article
Full-text available
N6-methyladenine (m6A) is the most predominant RNA modification, which has been shown to be related to many types of cancers. However, understanding of its role in prostate cancer (PCa) is largely unknown. Here, we report an upregulation of METTL14 that was correlated with poor prognosis in PCa patients. Functionally, knocking down METTL14 inhibited tumor proliferation both in vitro and in vivo. Mechanically, RNA-seq and MeRIP-seq analyses identified THBS1 as the downstream target of METTL14 in PCa. METTL14 downregulated THBS1 expression in an m6A-dependent manner, which resulted in the recruitment of YTHDF2 to recognize and degrade Thrombospondin 1 (THBS1) mRNA. Thus, our findings revealed that METTL14 acted as an oncogene by inhibiting THBS1 expression via an m6A-YTHDF2-dependent manner. METTL14 could be a potential prognosis marker and a therapeutic target.
Article
N6-methyladenosine (m6A), the most prevalent internal modification in mammalian mRNAs, is involved in many pathological processes. METTL16 is a recently identified m6A methyltransferase. However, its role in leukemia has yet to be investigated. Here, we show that METTL16 is a highly essential gene for the survival of acute myeloid leukemia (AML) cells via CRISPR-Cas9 screening and experimental validation. METTL16 is aberrantly overexpressed in human AML cells, especially in leukemia stem cells (LSCs) and leukemia-initiating cells (LICs). Genetic depletion of METTL16 dramatically suppresses AML initiation/development and maintenance and significantly attenuates LSC/LIC self-renewal, while moderately influencing normal hematopoiesis in mice. Mechanistically, METTL16 exerts its oncogenic role by promoting expression of branched-chain amino acid (BCAA) transaminase 1 (BCAT1) and BCAT2 in an m6A-dependent manner and reprogramming BCAA metabolism in AML. Collectively, our results characterize the METTL16/m6A/BCAT1-2/BCAA axis in leukemogenesis and highlight the essential role of METTL16-mediated m6A epitranscriptome and BCAA metabolism reprograming in leukemogenesis and LSC/LIC maintenance.
Article
RNA N⁶-methyladenosine (m⁶A) modification regulates the cell stress response and homeostasis, but whether titanium dioxide nanoparticle (nTiO2)-induced acute pulmonary injury is associated with the m⁶A epitranscriptome and the underlying mechanisms remain unclear. Here, the potential association between m⁶A modification and the bioeffects of several engineered nanoparticles (nTiO2, nAg, nZnO, nFe2O3, and nCuO) were verified thorough in vitro experiments. nFe2O3, nZnO, and nTiO2 exposure significantly increased the global m⁶A level in A549 cells. Our study further revealed that nTiO2 can induce m⁶A-mediated acute pulmonary injury. Mechanistically, nTiO2 exposure promoted methyltransferase-like 3 (METTL3)-mediated m⁶A signal activation and thus mediated the inflammatory response and IL-8 release through the degeneration of anti-Mullerian hormone (AMH) and Mucin 5 B (MUC5B) mRNAs in a YTH m⁶A RNA-binding protein 2 (YTHDF2)-dependent manner. Moreover, nTiO2 exposure stabilized METTL3 protein by the lipid reactive oxygen species (ROS)-activated ERK1/2 pathway. The scavenging of ROS with ferrostatin-1 (Fer-1) alleviates the ERK1/2 activation, m⁶A upregulation, and the inflammatory response caused by nTiO2 both in vitro and in vivo. In conclusion, our study demonstrates that m⁶A is a potential intervention target for alleviating the adverse effects of nTiO2-induced acute pulmonary injury in vitro and in vivo, which has far-reaching implications for protecting human health and improving the sustainability of nanotechnology.
Article
Metabolic abnormalities are one of the main hallmarks of cancer and are associated with chemoresistance. Therefore, targeting the metabolic reprogramming of cancer cells has the potential to overcome chemoresistance. Probiotic-derived extracellular vesicles (EVs) play important roles in biological function and intracellular communication. However, the inhibitory effect of Lactobacillus plantarum-derived EVs (LpEVs) on colorectal cancer (CRC) cells has not yet been elucidated. This study clearly revealed that increased glycolysis in 5-fluorouracil (5-FU)-resistant CRC cells (CRC/5FUR) is directly related to chemoresistance and that the metabolic shift reversed by LpEVs inhibits cancer cell proliferation and eventually leads to apoptosis. Pyruvate dehydrogenase kinase 2 (PDK2), one of the crucial enzymes for enhancing glycolysis, was upregulated in CRC/5FUR cells. In our study, LpEVs sensitized CRC/5FUR cells to 5-FU by attenuating PDK2 expression in p53-p21-dependent metabolic signaling, thereby circumventing 5-FU resistance. We demonstrated the effect of cellular responses to 5-FU by modifying the PDK2 expression level in both 5-FU-sensitive parental CRC and 5-FU resistant CRC cell lines. Finally, we revealed that the PDK2 signaling pathway can potentially be targeted using LpEVs treatment to overcome chemoresistant CRC, thereby providing a potential strategy for CRC treatment by intervening in tumor metabolism.