Вы находитесь на странице: 1из 12

Seminars in Immunology 32 (2017) 62–73

Contents lists available at ScienceDirect

Seminars in Immunology
journal homepage: www.elsevier.com/locate/ysmim

Review

The influence of the commensal microbiota on distal tumor-promoting MARK


inflammation

Claire M. Buchta Rosean, Melanie R. Rutkowski
University of Virginia, Department of Microbiology, Immunology and Cancer Biology, 345 Crispell Drive, Carter Harrison Research Building, Room G526, Charlottesville,
VA, United States

A R T I C L E I N F O A B S T R A C T

Keywords: Commensal microbes inhabit barrier surfaces, providing a first line of defense against invading pathogens, aiding
Commensal microbiota in metabolic function of the host, and playing a vital role in immune development and function. Several recent
Cancer studies have demonstrated that commensal microbes influence systemic immune function and homeostasis. For
Inflammation patients with extramucosal cancers, or cancers occurring distal to barrier surfaces, the role of commensal mi-
Metabolism
crobes in influencing tumor progression is beginning to be appreciated. Extrinsic factors such as chronic in-
Dysbiosis
flammation, antibiotics, and chemotherapy dysregulate commensal homeostasis and drive tumor-promoting
systemic inflammation through a variety of mechanisms, including disruption of barrier function and bacterial
translocation, release of soluble inflammatory mediators, and systemic changes in metabolic output. Conversely,
it has also been demonstrated that certain immune therapies, immunogenic chemotherapies, and checkpoint
inhibitors rely on the commensal microbiota to facilitate anti-tumor immune responses. Thus, it is evident that
the mechanisms associated with commensal microbe facilitation of both pro- and anti-tumor immune responses
are context dependent and rely upon a variety of factors present within the tumor microenvironment and sys-
temic periphery. The goal of this review is to highlight the various contexts during which commensal microbes
orchestrate systemic immune function with a focus on describing possible scenarios where the loss of microbial
homeostasis enhances tumor progression.

1. Introduction homeostasis, or commensal dysbiosis, can lead to increased inflamma-


tion and immune pathology that ultimately affects the systemic per-
Commensal microbes, comprised of bacteria, archaea, viruses, and iphery. In this context, alterations to commensal equilibrium induce
eukaryotes, inhabit all mucosal barrier surfaces, providing a physical pathological inflammation that is supportive of tumor growth. Al-
barrier in defense against invading pathogens. Additionally, commensal though we do not yet have a firm understanding of the precise micro-
microbes play essential roles in the maintenance of local tissue and bial populations that associate with tumor-promoting inflammation, it
immune homeostasis within the gastrointestinal tract [1–5], the skin is evident that commensal microbes influence the outcome of extra-
[6,7], the urogenital tract [8,9] and the oral/respiratory tract [10–13]. mucosal tumors.
Colonization with commensal microbes at birth is critical for the Over three decades ago, scientists began to observe that certain
postnatal development and function of mucosal immunity [14,15]. gram-negative commensal species influence myelopoiesis and the
However, commensal-mediated immune conditioning extends beyond emergence of granulocyte precursors from the bone marrow [16–18],
mucosal surfaces, impacting both systemic immune function and suggesting that commensal microbes influence immune function
homeostasis. Changes in commensal homeostasis are dynamic and through undefined interactions with distal sites such as the bone
occur gradually during aging or from changes in diet. Acute dis- marrow. Germ-free mice have a deficit in the myeloid compartment of
turbances resulting from antibiotic usage, infection, or chemotherapy bone marrow, resulting in increased susceptibility to infection with
can also drastically alter established commensal equilibrium, rapidly Listeria. However, restoration of immune function is achieved through
culminating in a loss of immune homeostasis. Loss of commensal recolonization of germ-free mice with fecal contents from conventional

Abbreviations: LPS, lipopolysaccharide; TLR, Toll-like receptor; NKT cells, natural killer T cells; NOD, nucleotide-binding oligomerization domain–containing protein; SNP, single
nucleotide polymorphism; MDSC, myeloid-derived suppressor cell; PSA, polysaccharide A; RIG-I, retinoic acid-inducible gene-I; SCFA, short-chain fatty acid; CDC, Centers for Disease
Control and Prevention; PGE2, prostaglandin E2

Corresponding author.
E-mail address: mr2ee@virginia.edu (M.R. Rutkowski).

http://dx.doi.org/10.1016/j.smim.2017.06.002
Received 1 March 2017; Received in revised form 29 May 2017; Accepted 20 June 2017
Available online 04 July 2017
1044-5323/ © 2017 Elsevier Ltd. All rights reserved.
C.M. Buchta Rosean, M.R. Rutkowski Seminars in Immunology 32 (2017) 62–73

mice, enabling clearance of Listeria [19]. Additional studies demon- 2.1. Inflammation, dysbiosis, and cancer
strated that commensal products, such as lipopolysaccharide (LPS) or
peptidoglycan provide a tonic level of stimulation through Toll-like There are several studies linking dysbiosis with cancer and in-
receptors (TLR) and other innate receptors expressed by myeloid cells, flammation, although it remains relatively undefined whether dysbiosis
driving myelopoiesis [20] and enhancing myeloid clearance of bacterial directly impacts tumor progression or serves as a biomarker of onco-
[21] and viral pathogens [22,23]. Commensal microbes are also asso- genesis. Dysbiosis has been demonstrated in patients with advanced
ciated with the development of mucosal-associated and peripheral breast cancer, with breast tumors having reduced microbial diversity
lymphocytes such as Foxp3+ regulatory T cells [24–26], IL-17-produ- compared to normal breast tissue [34]. In these patients, reduced di-
cing αβ T cells [27,28] and γδ T cells [29], and invariant NKT cells versity of tumor-associated commensal species corresponds with re-
[30,31]. Data from the Human Functional Genomics Project supports duced expression of inflammatory innate signaling receptors such as
much of what has been elucidated in mice, demonstrating that distinct TLR2, TLR5, and nucleotide-binding oligomerization do-
commensal or metabolic signatures are associated with both innate and main–containing protein (NOD)1 and NOD2 [34]. These innate re-
adaptive cytokine response patterns [32]. These studies underscore the cognition receptors may serve a protective role in breast tissue as TLR5
complex immunoregulatory influence that commensal microbes have signaling and activation of MAIP1S has been shown to inhibit breast
on local and systemic immune homeostasis in healthy individuals. tumor growth through the induction of autophagy and tumor cell death
Cancer is a systemic disease: inflammatory immune cells, chemo- [48,49]. These studies suggest that dysbiosis within the breast tissue
kines and cytokines distally influence tumor growth and metastatic may occur through dysregulation of innate signaling receptors, pro-
progression. Cancer can impact the composition of commensal mi- moting the outgrowth of inflammatory or DNA-damaging bacterial
crobes locally within affected tissues [33–36] or distally within the species. Indeed, Urbaniak et al. determined that Escherichia coli and
intestines [37,38], altering the immune environment in favor of tumor Staphylococcus epidermidis isolates from dysbiotic breast tissues are able
growth and global immune suppression [39]. The relationship between to directly induce DNA damage in a tumor cell line [50]. Microbial
commensal microbes, inflammation, and oncogenesis is well-docu- sequencing of an additional cohort of breast tissue specimens found a
mented for colorectal cancer [40–42], which is locally influenced by tumor-specific increase in Fusobacterium [51], a genus of bacteria which
dysregulation of commensal homeostasis as a result of chronic anti- harbors the species F. nucleatum, a bacterium directly associated with
biotic exposure, diet, age, infection, and/or genetic polymorphisms that driving inflammation and carcinogenesis in colorectal cancer [52,53].
drive inflammation and oncogenesis. Cancer patients may also have Changes in the composition of commensal microbes within the re-
disruptions in commensal homeostasis as a result of chemotherapy, productive tract are also associated with increased inflammation.
administration of antibiotics, whole body irradiation, cachexia, and/or Women with endometrial cancer have dysbiosis in the vagina, cervix,
systemic tumor-promoting inflammation (Fig. 1). Several recent studies and endometrium that is associated with malignant progression [54].
have begun to link changes within the composition of commensal mi- Protein analysis of cervical samples revealed that severe cervical dys-
crobes to global modulation of tumor-promoting inflammatory cyto- biosis correlates with elevated levels of both proinflammatory cytokines
kines [39] and have identified certain microbes that facilitate en- and enzymes associated with proteolysis and alterations to the cervical
hancement of anti-tumor immune responses during immunotherapy mucosa and cytoskeleton [55]. Furthermore, alterations in the com-
[43–45] and chemotherapy [43,46]. Thus, patients with extramucosal position of bacteria within the reproductive tract are associated with
tumors, occurring distal to mucosal surfaces, are also influenced by increased levels of GM-CSF, TNFα, IFNγ, and IL-1β [56]: cytokines that
alterations in commensal composition. In this review, we will highlight promote myeloid infiltration within tumor beds. Together, these studies
mechanisms associated with commensal-induced pathological in- suggest that severe dysbiosis within the reproductive tract leads to an
flammation with a focus on detailing how microbes, microbial pro- increase in inflammation and pathology, resulting in damage to mu-
ducts, and/or disruptions in commensal homeostasis impact extra- cosal surfaces. Importantly, these studies highlight that the location,
mucosal cancer progression. composition and function of the normal microflora within each unique
niche serves a specific homeostatic role.

2. Alterations to the microbiome associated with inflammation 2.2. Diet-induced dysbiosis and cancer
and cancer
Diet-induced dysbiosis and obesity are prevalent health issues in
In healthy adults, the abundance of certain commensal species is developed countries. Morbidities associated with obesity include in-
associated with the functional ability of both myeloid and lymphoid sulin resistance, cardiovascular disease, and an increased incidence of
cells to produce inflammatory cytokines such as TNFα, IL-6, IL-1β, several types of cancers [57–60]. In a healthy individual, commensal
IFNγ, IL-17 and IL-22 [32]. These cytokines are all capable of influen- metabolic byproducts help to stabilize commensal equilibrium, pre-
cing tumor progression through multiple mechanisms, including the venting the growth of inflammatory species which compete with the
promotion of tumor growth through the recruitment of suppressive host for nutrients. In homeostatic conditions, commensal microbes also
immune cells into the tumor microenvironment via TNFα, IL-6, IL-1β or salvage potentially toxic nutritional byproducts such as bile, which can
facilitating enhanced tumor immune surveillance via IFNγ and IL-17. accumulate, cause toxicity, and in some instances, induce DNA damage
However, microbial populations associated with inflammation may be leading to oncogenic transformation [61]. A previous study comparing
altered during states of dysbiosis, defined as an imbalance of com- the bacterial composition of obese and non-obese individuals found that
mensal homeostasis and resultant outgrowth of pathological microbial obese individuals have significantly reduced gene richness and com-
species. It is well-accepted that commensal dysbiosis can drive positional diversity within their microbiota, with a predominance of
pathologies within barrier surfaces, but dysbiosis also results in sys- Bacteroides spp. occurring within the dysbiotic gastrointestinal tract
temic damage to distal organs due to aberrant inflammation and me- [62]. Functionally, this study found that commensal bacteria from
tabolic dysregulation [47]. Dysbiosis can be induced by multiple me- obese individuals have microbial gene signatures associated with in-
chanisms, including diet or genetic-induced dysbiosis, antibiotic- flammation and mucosal damage, including increased proportions of
induced dysbiosis, and dysbiosis due to tumor-promoting chronic in- inflammatory bacterial species, a reduced capacity to produce immune
flammation, all of which associate with a more unfavorable outcome regulatory butyrate, an increase in mucus degrading proteins, and an
during cancer. increased capacity to handle oxidative stress [62]. Diet-induced
changes in microbial diversity can therefore result in systemic in-
flammation and a disruption in homeostasis due to altered metabolite

63
C.M. Buchta Rosean, M.R. Rutkowski Seminars in Immunology 32 (2017) 62–73

Fig. 1. Loss of commensal equilibrium results in systemic


inflammation and context-dependent modulation of extra-
mucosal tumor progression. Factors such as antibiotic usage,
diet, or genetics (black arrows) can effectively decrease the
biodiversity of the commensal microflora, leading to a higher
incidence of chronic inflammation. In tumor-bearing in-
dividuals, cancer therapy and tumor-derived inflammation
(red arrows) can also modulate the composition of the com-
mensal ecosystem. A reduction in commensal biodiversity
(dysbiosis), antibiotics, and chemotherapy can directly da-
mage the integrity of the mucosal epithelium, resulting in
bacterial translocation, commensal shedding of inflammatory
products, such as LPS, PSA, and flagellin, and a change in
metabolic output from the commensal ecosystem. This can
lead to an enhancement in tumor-promoting inflammation
due to the induction of IL-6, GM-CSF, and TGFβ: factors
which enhance the recruitment of immune suppressive mye-
loid-derived suppressor cells and regulatory T cells into the
tumor microenvironment. In the presence of these cells, the
ability of T cells to effectively eliminate the tumor are in-
hibited. Translocation of commensals from mucosal surfaces
can also induce the activation of IFNγ-producing Th17 cells,
which in certain contexts can facilitate T cell-mediated killing
of tumors.

output and/or a disruption in mucosal integrity, leading to bacterial during tumor progression. The effects of butyrate will be discussed in
translocation and systemic distribution of shed microbial products further detail in section 4.1.
(Fig. 1), which we will discuss in greater detail below. Diets high in fat and low in fiber also result in the accumulation of
In mice, a high-fat diet results in changes within the composition of metabolites associated with increased toxicity. For example, obesity has
commensal microbes, promoting decreased production of im- been linked to increased systemic levels of deoxycholic acid [61]. Me-
munoregulatory metabolites and localized inflammation within the chanistically, circulation of deoxycholic acid through the hepatic portal
stomach epithelium, leading to cancer [63]. Fecal transplant experi- vein induces an inflammatory and pro-tumorigenic senescence-asso-
ments have demonstrated that transfer of the microbiome from mice fed ciated secretory phenotype in hepatic stellate cells, resulting in in-
a high-fat diet was sufficient to increase cancer incidence in recipient flammation-induced damage of adjacent hepatic cells and resultant
animals [63], providing evidence that diet-induced changes to the oncogenesis [65]. Supporting a role for this DNA-damage inducing
composition of commensal microbes is sufficient to influence cancer metabolite, rats fed a diet containing high amounts of deoxycholic acid
progression independent of the physiologic manifestations associated developed pre-neoplastic liver lesions [66]. Alterations in commensal
with obesity. Administration of the immune regulatory short-chain fatty microbe composition resulting from dietary changes may also influence
acid butyrate, which was reduced in people and animals with diet-in- additional bacterial metabolites as highlighted in Table 1.
duced changes to commensal equilibrium [62,63], was also sufficient to
reduce histopathology and tumor progression in animals fed a high-fat 2.3. Antibiotic-induced dysbiosis
diet [63]. Butyrate directly inhibits the growth of multiple tumor types
through inhibition of histone deacetylases and potent anti-in- Although antibiotics have significant health benefits for individuals
flammatory activity [64], suggesting that dietary intervention through with microbial infections, excessive antibiotic exposure can negatively
consumption of fiber-rich foods could provide therapeutic benefit impact commensal biodiversity and immune function [67–69].

64
C.M. Buchta Rosean, M.R. Rutkowski Seminars in Immunology 32 (2017) 62–73

Table 1
Bacteria and associated metabolites that influence cancer and anti-tumor immunity.

Bacterial metabolite Bacteria that produces Function in context of cancer Reference

Short-chain fatty acids: butyrate, Butyrate: Firmicutes Pro-tumor: HDAC inhibitors. Suppresses inflammation. [146,148,151,163–166,168,170–174,176,177,209]
propionate, and acetate Induces Tregs, eliminates activated T cells through
upregulation of Fas. Induces production of PGE2.
Propionate and acetate: Anti-tumor: anti-proliferative and pro-apoptotic effects on
Bacteroidetes. cancer lines. Inhibits angiogenesis. Suppresses CCL2
production.
Deoxycholic acid Many Pro-tumor: DNA damage [182,183,210]
Superoxide E. faecalis Pro-tumor: DNA damage [184]
Colibactin E. coli Pro-tumor: DNA damage [186,187]
Hydrogen sulfide Sulfate-reducing Pro-tumor: DNA damage [211]
bacteria
Polyamines Many Pro-tumor: suppression of lymphocyte proliferation and IL-2 [188,189,191,195–198,212]
production. Promote cancer cell proliferation.
Anti-tumor: protect against DNA damage.
β-glucuronidase Many Pro-tumor: Increases estrogen levels, pro-tumor effects on [202,213,214]
both estrogen-receptor positive and negative cancers.
Fragilysin B. fragilis Pro-tumor: Stimulates cellular proliferation and E-cadherin [215,216]
degradation, degrades intestinal barrier function.
Heptose-1,7-bisphosphate (HBP) Neisseria Pro-tumor: Activates NF-κB pathway and promotes [217]
inflammation.
Kynurenine Indole-positive bacteria Pro-tumor: Tryptophan catabolite, suppresses T cell function. [218,219]

According to the CDC, greater than 5 out of 6 individuals are prescribed of recombinant IL-17 [76], demonstrating that the systemic influence of
antibiotics annually in the United States [70], often unnecessarily. commensal microbes on the induction of immunoregulatory IL–17 T
Additionally, cancer patients are frequently prescribed prophylactic cells can serve a protective role in the establishment of anti-tumor
antibiotics due to severe immune suppression and susceptibility to in- immunity. However, other studies have demonstrated that antibiotic
fectious diseases. Several studies have demonstrated associations be- treatment induces the production of immune-suppressive and tumor-
tween chronic/prolonged antibiotic use and increased cancer risk. In promoting mediators through the induction of dysbiosis. Antibiotic-
one study, women prescribed 11 or more courses of the same antibiotics induced dysbiosis promotes the outgrowth of Candida species in the
had an increased association with developing breast cancer [71]. While lungs, resulting in an increase in prostaglandin E2 (PGE2) production
a large-scale retrospective study of over 2 million women found only a and polarization of macrophages into an M2 suppressive phenotype
slight increase in hazard risk of breast cancer in women using all classes [77]. These studies highlight the contextual relationship between
of antibiotics for more than 101 days, women prescribed 3 or more commensal microbes and the immune system. By further understanding
courses of tetracyclines over a span of more than 101 days have a sig- the functional consequences of alterations to microbial diversity, we
nificantly greater risk of breast cancer than women given other types of may be able to develop unique probiotic cocktails to attenuate various
antibiotics [72,73]. Similarly, in a large multi-cancer health database pathological conditions, especially in immune compromised patients
survey, it was found that the risk for lung, esophageal, gastric and renal that require antibiotic treatment.
cancers significantly increases in individuals prescribed penicillins
more than 5 times. The risk for lung and renal cancers is significantly
associated with multiple courses of macrolides [73]. This study also 2.4. Genetic mutations associated with effects on bacterial composition/
confirmed that long-term exposure to tetracyclines increases the risk of tumorigenesis
breast cancer [73]. Overall, these retrospective studies suggest that
although a single exposure to antibiotics does not significantly influ- Analysis of data from the Human Microbiome Project demonstrates
ence cancer risk, multiple and long-term exposures to single classes of a significant association between certain single nucleotide polymorph-
antibiotics increase the incidence of numerous cancers. Although the isms (SNP) and the composition of commensal microbes at several
mechanisms linking antibiotic use and cancer development and pro- barrier sites [78]. Polymorphisms associated with immunological
gression are unknown, one might speculate that chronic antibiotic ex- pathways and commensal-driven diseases significantly impact micro-
posure drives prolonged dysbiosis, inflammation, and tissue damage bial composition [78]. This human data supports several mouse studies
that leads to an increased risk for oncogenic transformation in suscep- demonstrating that single genes associated with microbial sensing are
tible tissues. However, undefined host or environmental factors may significantly able to impact the composition of commensal microbes
result in the need for frequent antibiotic courses and may also be as- and overall host physiology [79–81].
sociated with an increased risk of cancer. Further studies are necessary Approximately 7% of the general population harbors a dominant-
to determine whether there is a direct relationship between antibiotics negative SNP mutation within the flagellin-binding domain of TLR5,
and cancer. resulting in ablation of TLR5 signaling [82]. This polymorphism was
Antibiotics, especially tetracyclines [74], are also capable of directly shown to have an influence on the outcome and survival of patients
influencing tumor progression through the induction of mitochondrial with various extramucosal malignancies [39]. Mechanistically, TLR5
dysfunction, increased production of reactive oxygen species (ROS), signaling through interactions with commensal microbes results in
and corresponding DNA damage [75]. Antibiotics have been shown to systemic elevation of IL-6 and the recruitment of myeloid-derived
directly influence immune cell function in addition to modulating the suppressor cells (MDSC) into the tumor microenvironment. MDSCs se-
composition of commensal microbes, resulting in the attenuation of creting adenosine induce galectin-1 expression in γδ T cells, resulting in
immune surveillance. Metastatic growth of Lewis lung carcinoma and cumulative suppression of tumor-associated immune responses and
B16F10 melanoma is enhanced in the lungs of antibiotic-treated mice more rapid progression of ovarian tumors and sarcomas [39]. This
which is mediated by a reduction in lung-protective IL-17-producing γδ study was the first to suggest that genetic polymorphisms present
T cells. This increase in metastatic growth was lost upon administration within the human population, mediated through interactions with
commensal microbes and the immune system, could influence the

65
C.M. Buchta Rosean, M.R. Rutkowski Seminars in Immunology 32 (2017) 62–73

outcome of extramucosal cancers. injury and the induction of acute pancreatitis in mice [104,105]. This
A recent study has demonstrated that retinoic acid-inducible gene-I inflammation accelerates the development of pancreatic cancer in mice
(RIG-I), a viral RNA receptor capable of recognizing double-stranded with a genetic driver mutation in K-ras [106,107]. As mutated K-ras has
RNA viruses, regulates the composition of commensal microbes through been identified in a substantial proportion of healthy individuals, in-
the production of IgA and the downstream induction of Reg3γ, an an- creased levels of circulating endotoxin may synergize with K-ras in-
timicrobial peptide important in controlling the composition of com- duced transformation, driving tumorigenesis [108,109]. Furthermore,
mensal microbes [83]. RIG-I-deficient mice have increased dysbiosis systemic exposure to LPS enhances metastasis to both the lungs and
and emergence to colorectal cancer upon treatment with AOM/DSS. liver in several different tumor models [110–112]. LPS may act directly
Clinically, these findings are relevant, as human colorectal cancer on tumor cells, eliciting production of mediators that influence epi-
specimens have significantly reduced levels of RIG-I, suggesting that thelial mesenchymal transition and promote enhanced metastasis
downregulation of this receptor results in dysbiosis and susceptibility to through the induction of cell adhesion molecules that facilitate inva-
colorectal cancer [83]. Expression of NOD2 on the colonic mucosa was sion, as demonstrated using in vitro cell culture spheroid models
also found to influence the composition of commensal microbes and the [113,114]. Additionally, TLR4 expression associates with metastasis to
inflammatory microenvironment within the intestines, leading to an distal lymph nodes in patients with breast cancer [115]. In vitro sti-
increased incidence of colitis-associated cancer in NOD2-deficient mice mulation of breast tumor cell lines with LPS induces a significant in-
[84]. Although most studies have focused upon the role of NOD re- crease in tumor-specific production of matrix metalloproteinases and
ceptors in modulating intestinal malignancies, a study using NOD-de- cytokines associated with angiogenesis, such as VEGF and IL-6. In im-
ficient breast tumor cell lines demonstrated that breast tumors lacking munocompromised mice, this ultimately results in increased metastatic
NOD were more resistant to TNFα-induced apoptosis and were more dissemination to the liver [115].
sensitive to estrogen-mediated tumor growth [85]. Thus, similar to Another bacterial product associated with systemic inflammatory
what has been reported in colorectal cancer, NOD expression on the effects is flagellin. The only known ligand for TLR5, flagellin is the main
tumor epithelium may have a host-protective role in extramucosal protein component of flagellum on flagellated bacteria. Though it has
cancers, such as breast cancer. However, the attenuation of tumor been reported to be detectable in healthy human serum, systemic
growth in this model is primarily mediated through NOD-dependent concentrations of flagellin in the serum increase in the context of in-
activation of caspase 8, resulting in apoptosis [85,86]. Because these flammation and injury [116–118]. Flagellin has been reported to have
studies were performed using in vitro models, this mechanism may be both pro- and anti-tumor effects. Multiple publications have demon-
mediated independently of commensal microbiota within the breast strated immunosuppressive functions of flagellin and TLR5 signaling
tissue. through the induction of Th2 responses [119,120]. Dendritic cells sti-
mulated with flagellin secrete low levels of Th1-supportive cytokines,
3. Microbial products associated with inflammation and tumor such as IL-12p40 and p70, IL-6 and TNFα, resulting in Th2 polarization
progression of CD4T cells [119]. Additionally, flagellin stimulation of myeloid
precursors induces the generation of CXCR4-expressing MDSCs [120].
While most bacteria comprising the human microbiota are com- These studies suggest that depending upon the cell type (mature DC
partmentalized on the skin and in the gastrointestinal and female re- versus immature myeloid precursor) and the immunological context of
productive tracts, recent studies have demonstrated that additional sites flagellin activation, stimulation through TLR5 may have varying im-
contain regional microbiota. Organs previously considered to be sterile mune modulatory effects. Flagellated bacteria are a common com-
in healthy individuals, such as lymph nodes [87–89] and the bladder mensal type found on murine skin, and the presence of flagellin ex-
[90–93], are now known to contain bacteria under homeostatic con- acerbates tumor development in a model of skin cancer [7,121]. In
ditions without any observable pathological consequence. Additionally, vitro, flagellin enhances proliferation and migration of several types of
some bacterial-derived products reach systemic circulation [94,95], cancer cell lines, including multiple myeloma, gastric cancer, and
which may influence tumor progression. For example, disruption in the salivary gland adenocarcinoma [122–124]. However, other studies
compartmentalization of commensal microbes or enhanced microbial have shown that flagellin promotes anti-tumor activity through sup-
shedding of inflammatory ligands during pathological conditions has pression of cancer cell proliferation and migration [48,125,126]. These
the potential to drive systemic tumor-promoting inflammation, induce contrasting findings may depend on several factors, including the an-
DNA damage, and promote cellular proliferation: all of which promote tigenicity of the tumor, the timing of the flagellin stimulus [127], the
the progression of extramucosal tumors. cytokine and immune composition of the tumor microenvironment, and
perhaps even the stage of tumor initiation or progression.
3.1. Bacterial components circulating in the periphery Two additional bacterial components, peptidoglycan and poly-
saccharide A (PSA), affect systemic inflammation through the ligation
Bacterial LPS, also known as endotoxin, is the best characterized of TLR2. Peptidoglycan is a bacterial cell wall component that is shed
bacterial component found in systemic circulation. A cell wall compo- through cell division. Present in human plasma, peptidoglycan has been
nent of gram negative bacteria, LPS is the major ligand for TLR4 and shown to promote invasiveness, adhesion, and pro-inflammatory cyto-
stimulates proinflammatory cytokine production through binding the kine production of cancer cells in vitro [128,129]. PSA, a capsule
receptor. Additionally, LPS has pro-angiogenic effects [96–98]. In the component of Bacteroides fragilis, drives the differentiation of im-
context of wound healing, pro-inflammatory and pro-angiogenic func- munosuppressive IL-10-producing Foxp3+ regulatory T cells both in
tions of acute LPS exposure are considered beneficial, but chronic LPS- mice and humans [130–132]. The broad expression of TLRs 2, 4, and 5
mediated inflammation could be detrimental during tumorigenesis and on cells of various human cancer types underline the potential effects of
metastasis. Importantly, though gram negative bacteria shed LPS as a bacteria and/or their components on the tumor microenvironment
normal part of cell division, antibiotics further promote LPS shedding (reviewed in [133]).
through their bactericidal effects [99,100], suggesting that during
dysbiosis, LPS levels may be elevated in the periphery. 3.2. Bacterial translocation
In addition to promoting proinflammatory cytokine production, LPS
also drives myeloid cells to produce ROS [101,102]. The superoxide While resident bacterial communities are present in extramucosal
radicals produced by this process cause direct DNA damage, potentially organs of healthy individuals, intestinal barrier permeability is ad-
contributing to mutations leading to cellular transformation [103]. The ditionally increased in the context of inflammation, enhancing trans-
resultant inflammation from LPS exposure contributes both to hepatic location of bacteria and bacterial components from the gut to distal

66
C.M. Buchta Rosean, M.R. Rutkowski Seminars in Immunology 32 (2017) 62–73

locations [134]. For example, intestinal permeability and systemic le- coupled receptors, and butyrate and propionate exert their anti-in-
vels of LPS are enhanced in cases of alcohol abuse and are often de- flammatory effects through their actions as histone deacetylase in-
tected in patients with metabolic disorders influenced by obesity hibitors. These effects include the modulation of NF-κB pathways,
[95,135–137]. Considering the strong correlations between obesity, leukocyte trafficking, and suppression of various cytokines and che-
alcohol use, and increased cancer risk, bacterial translocation and/or mokines [145–153]. Additionally, SCFAs promote tight junction in-
systemic bacterial products likely play a large role in promoting the tegrity in the intestines, reducing translocation of bacteria and bacterial
systemic inflammation seen in these conditions. Cancer treatments such products [154–156]. Germ-free mice have very low concentrations of
as chemotherapy, radiation, and checkpoint blockade also result in SCFAs in their intestines, demonstrating the importance of commensal
mucosal damage, increasing intestinal permeability [94]. Total body microbes in the production of these metabolites [157]. SCFAs have
irradiation, used to lymphodeplete recipients of both bone marrow mainly been investigated for their anti-inflammatory effects on colitis
transplants and adoptive T cell therapy, results in elevated serum LPS [158]. However, as they reach systemic circulation after their deriva-
and proinflammatory cytokines in mice [94,138]. Thus, while bacteria tion in the colon, SCFAs may have effects in the context of extramucosal
may play a role in tumorigenesis, cancer treatment may additionally cancers [159–161].
exacerbate this effect by increasing bacterial translocation to extra- One well-known function of SCFAs is the effect they exert on po-
mucosal sites. pulations of T cells. Butyrate promotes both the induction of regulatory
Bacterial translocation is not always associated with tumor-pro- T cells and their resultant production of IL-10 in addition to driving the
moting inflammation, and may also be associated with the enhance- elimination of activated T cells through the upregulation of Fas
ment of anti-tumor immunity. Cyclophosphamide induces im- [162–166]. In the context of intestinal inflammation, these effects
munogenic cell death in cancer cells and promotes the differentiation of would be beneficial to the host. However, in the context of cancer, an
Th17 and Th1 cells which enhance therapeutic efficacy [139,140]. increase in Tregs and concurrent decrease in effector T cells would fa-
Cyclophosphamide also results in increased intestinal permeability and cilitate a reduction in tumor immune surveillance and anti-tumor im-
the translocation of bacteria from the intestines to lymph organs. Un- munity. This effect of butyrate extends beyond the gut, as mice on a
expectedly, cyclophosphamide-induced bacterial translocation of gram high-fiber or SFCA-supplemented diet showed both suppressed colonic
positive bacterial species results in the generation of Th1 memory T inflammation as well as diminished allergic airway disease as a result of
cells and the differentiation of IFNγ-producing Th17 cells (Fig. 1) which increased suppressive activity of Tregs in the lungs [167]. Additionally,
are critical for anti-tumor immune responses during therapy [46]. Be- SCFAs induce the production of PGE2, a potent tumor-promoting
cause this study demonstrates that antibiotics diminish the efficacy of mediator, from human monocytes. This effect is enhanced in the pre-
cyclophosphamide-induced tumor immune surveillance, it is important sence of LPS, demonstrating the complex effects of commensal bacteria
to consider how antibiotic administration during treatment with certain on systemic inflammation [168].
chemotherapies may impair therapeutic efficacy. Treatment with the However, other studies have shown anti-tumor effects of butyrate
checkpoint inhibitor anti-CTLA-4 is also known to induce mucosal pa- and other SCFAs. For example, oral administration of dietary fiber as a
thology and disrupt commensal homeostasis. However, Vetizou et al. prebiotic reduced the incidence of carcinogen-induced mammary
demonstrated a clear relationship between colonization with certain cancer [169]. Several potential mechanisms of SCFAs may account for
Bacteroides species, such as B. fragilis and B. thetaiotaomicron, and re- this effect. Butyrate exerts anti-proliferative and pro-apoptotic effects
duced mucosal pathology during treatment with a-CTLA-4. Ad- on several cancer cell lines in vitro [170–175], primarily through the
ditionally, they showed that T cells recognizing these bacterial species induction of oxidative stress within tumor cells and modulation of the
associate with responsiveness to therapy in both murine models and in expression of genes associated with cellular proliferation and growth.
patients with melanoma [45]. As T cells recognizing specific bacterial Additionally, butyrate inhibits angiogenesis through downregulation of
species mediate the efficacy of anti-CTLA-4 therapy, it is possible that VEGF gene expression [176]. Another potential anti-tumor effect of
shared antigens exist between commensal species and tumor neoanti- butyrate is its ability to suppress CCL2 production, both in vivo and in
gens. vitro from tumor cell lines, through impairment of phosphorylation of
the ERK1/2 and Akt inflammatory signaling pathways [177–179]. As
4. Systemic effects of commensal metabolites during cancer CCL2 has been implicated in tumor progression and metastasis through
recruitment of tumor-associated macrophages, SCFAs may inhibit this
Another mechanism by which bacteria contribute to systemic in- process [180]. Finally, it was demonstrated that butyrate in combina-
flammation is through the production of metabolites. The microbiome, tion with 5-azacytidine, a DNA methyltransferase inhibitor, reduces the
defined as the combined genetic material of all microbes within the growth of breast cancer stem cells [181], suggesting that using pro-
human body, outnumbers human genes by roughly 150-fold [141]. This biotics to enrich for butyrate-producing bacteria or increasing the
vast quantity of genetic material codes for a wide variety of different dietary consumption of fiber may have a protective role against certain
proteins that impact bacterial metabolism. Commensal bacteria have types of cancers.
key roles in human digestion, including the extraction of nutrients and
the synthesis of biologically relevant metabolites (reviewed in Ref. 4.2. Metabolites that influence DNA damage
[142]). Several groups have shown striking differences in the con-
centration and composition of circulating metabolites in germ-free In the context of gastrointestinal cancers, some bacterial metabo-
compared to conventional mice, demonstrating the contributions of lites impact tumorigenesis through direct DNA damage. Commensal
commensal microbes to systemic metabolic products [143,144]. This bacteria convert primary bile acids produced by the liver to secondary
could be relevant in the development of immune function as germ-free bile acids. As previously mentioned, the secondary bile acid deoxy-
animals have poorly developed lymphoid structures and a defect in cholic acid causes DNA damage through the induction of ROS
granulopoiesis, as discussed previously [19,20]. [182,183]. Additional bacterial products, such as Enterococcus faecalis-
derived superoxide and Escherichia coli-derived colibactin also induce
4.1. Short-chain fatty acids DNA strand breaks [184–187]. Repetitive DNA damage leads to the
acquisition of mutations that can drive cellular transformation and the
One of the best-studied families of microbial metabolites is the development of cancer.
short-chain fatty acids (SCFA), comprised of butyrate, propionate, and Polyamines, such as putrescine, spermidine, and spermine, are ad-
acetate. Produced by anaerobic bacterial fermentation of dietary fiber, ditional metabolites that are produced by commensal bacteria. Unlike
SCFAs are broadly anti-inflammatory. SCFAs are ligands for G protein- the metabolites mentioned above, polyamines actively protect against

67
C.M. Buchta Rosean, M.R. Rutkowski Seminars in Immunology 32 (2017) 62–73

DNA damage by scavenging free radicals and influencing the structure influencing the composition of commensal microbes. Chemotherapy
of DNA [188–192]. However, polyamines have also been implicated in influences microbial composition and induces translocation of com-
the suppression of anti-tumor immune responses. Polyamines are in- mensal microbes from barrier surfaces due to damage of the pro-
creased in the urine and serum of cancer patients and have been shown liferating cells within the mucosal epithelium. Depending upon the
to promote cancer cell proliferation [193–195]. Additionally, they chemotherapeutic agent and the mechanisms associated with tumor-
suppress lymphocyte proliferation and IL-2 production, presumably progression, commensal microbes may facilitate tumor regression or
through metabolic constraints on activated T cells, eventually resulting result in toxicity, as discussed previously. Multiple studies have found
in decreased anti-tumor immunity [196]. This effect can be inhibited, that certain immune therapies enhance tumor immune surveillance
as depletion of polyamines restricted tumor growth in a T cell-depen- through triggering of innate receptors and inflammasomes with mi-
dent mechanism [197]. Antibiotic treatment decreases polyamine crobial products, such as LPS, complexed DNA, and cellular RNA,
concentrations, demonstrating the impact of commensal bacteria on whereas inhibition of TLR3 and AIM2 inflammasome activation are
this metabolic product [198]. Polyamines can also directly enhance the protective against radiation-induced cytotoxicity [208]. Thus, it will be
production of tumor-derived proteases and matrix metalloproteinases, critical to understand the relevant contexts in which commensal mi-
resulting in increased extravasation and invasion of tumor cells. Com- crobes and the activation of relevant signaling pathways influence
bined with their function in immune suppression, polyamines promote tumor progression and anti-tumor immunity.
enhanced tumor progression and metastasis [199]. Commensal disequilibrium results in a range of pathologies, and in
the context of cancer can have a direct influence on tumor proliferation
4.3. Metabolites and hormone production and cytokine secretion (Fig. 1). Systemically, commensal products and
metabolic function provoke changes to immune function that may re-
Microbes also play a significant role in the metabolism of hormones. sult in either tumor-promoting inflammation or enhanced anti-tumor
This is particularly important in the context of hormone-receptor po- immunity. The role of commensal microbes during cancer is dependent
sitive cancers. Estrogen, specifically, is one driving factor in the de- upon many factors, demonstrating that a more comprehensive under-
velopment of hormone-receptor positive breast cancer and promotes standing of the effects that individual microbes have on tumor growth
tumor growth [200,201]. Estrogens are conjugated with glucuronic and immune function within the tumor microenvironment is needed.
acid in the liver, allowing them to be excreted. Some bacteria produce Conversely, it will be important to understand how tumors impact
an enzyme, β-glucuronidase, that deconjugates estrogen from glu- commensal homeostasis. Several studies have already identified that
curonic acid, promoting its reabsorption into circulation, thereby en- certain commensal microbes are required to facilitate cancer therapy:
hancing estrogen levels in the host [202]. In humans, differences in through checkpoint inhibition of PD-1 [44] and CTLA-4 [45] and
estrogen metabolism correlate with variability in gut microbial di- during treatment with immunostimulatory immune therapy [43] or
versity, and antibiotic treatment associates with an increase in excre- chemotherapy [46]. These and future studies could pave the way for
tion of conjugated estrogens [203–205]. Despite its significance in engineering probiotic cocktails associated with the restoration of anti-
hormone-receptor positive cancer, estrogen has also recently been tumor immune responses.
shown to affect the progression of different estrogen-insensitive tumor
models. Svoronos et al. showed that estrogen drives MDSC mobilization Acknowledgments
as well as their suppressive activity, enhancing tumor progression
[206]. This demonstrates that hormones directly impact both tumor This work was supported by the American Cancer Society IRG grant
growth and immune function. 81-001-29 and the National Cancer Institute P30 award
Additional bacterial metabolites have potential roles in promoting 4P30CA044579-25. CBR is supported by the National Institutes of
systemic inflammation and tumor progression. These are described in HealthT32 AI007496-22. We would like to thank Raegan R. Bostic and
Table 1. Tzu-Yu Feng for critical reading and editing of the manuscript.

5. Conclusions and future perspectives References

The microbiome is often thought of as the “forgotten organ” [207]. [1] S. Rakoff-Nahoum, J. Paglino, F. Eslami-Varzaneh, S. Edberg, R. Medzhitov,
Work from the last twenty years has demonstrated critical roles for Recognition of commensal microflora by toll-like receptors is required for in-
testinal homeostasis, Cell 118 (2) (2004) 229–241.
commensal microbes in the development and progression of many [2] S.K. Mazmanian, J.L. Round, D.L. Kasper, A microbial symbiosis factor prevents
disease states, including cancer. The widespread use of antibiotics in intestinal inflammatory disease, Nature 453 (7195) (2008) 620–625.
our society makes this topic relevant in the context of cancer, as cancer [3] M.J. Silva, M.B. Carneiro, B. dos Anjos Pultz, D. Pereira Silva, M.E. Lopes, L.M. dos
Santos, The multifaceted role of commensal microbiota in homeostasis and gas-
patients are frequently prescribed prophylactic antibiotics to combat trointestinal diseases, J. Immunol. Res. 2015 (2015) 321241.
infections associated with chemotherapy-induced immunosuppression. [4] D.A. Hill, C. Hoffmann, M.C. Abt, Y. Du, D. Kobuley, T.J. Kirn, F.D. Bushman,
While chronic antibiotic exposure increases the risk of tumorigenesis in D. Artis, Metagenomic analyses reveal antibiotic-induced temporal and spatial
changes in intestinal microbiota with associated alterations in immune cell
multiple cancer types, antibiotic use also impacts commensal home- homeostasis, Mucosal Immunol. 3 (2) (2010) 148–158.
ostasis, decreasing circulating bacterial components and metabolites [5] C. Zaph, Y. Du, S.A. Saenz, M.G. Nair, J.G. Perrigoue, B.C. Taylor, A.E. Troy,
and shaping systemic immune function. As discussed previously, many D.E. Kobuley, R.A. Kastelein, D.J. Cua, Y. Yu, D. Artis, Commensal-dependent
expression of IL-25 regulates the IL-23-IL-17 axis in the intestine, J. Exp. Med. 205
secreted bacterial factors act in context-dependent mechanisms, and
(10) (2008) 2191–2198.
depending upon the type and stage of cancer, the class of antibiotics [6] S. Nakamizo, G. Egawa, T. Honda, S. Nakajima, Y. Belkaid, K. Kabashima,
prescribed, the duration of antibiotic exposure, and the initial compo- Commensal Bacteria and Cutaneous Immunity Commensal Bacteria and Cutaneous
sition of commensal microorganisms, antibiotic use during cancer Immunity, Springer, 2015, pp. 73–80.
[7] S. Naik, N. Bouladoux, C. Wilhelm, M.J. Molloy, R. Salcedo, W. Kastenmuller,
therapy has the potential to both promote and to inhibit cancer pro- C. Deming, M. Quinones, L. Koo, S. Conlan, S. Spencer, J.A. Hall, A. Dzutsev,
gression. It was recently hypothesized that the impact of persistent use H. Kong, D.J. Campbell, G. Trinchieri, J.A. Segre, Y. Belkaid, Compartmentalized
of antibiotics is compounded across generations, resulting in the gra- control of skin immunity by resident commensals, Science 337 (6098) (2012)
1115–1119.
dual loss of diversity and emergence into dysbiotic states [67]. Thus, [8] S.A. Whiteside, H. Razvi, S. Dave, G. Reid, The microbiome of the urinary tract —
further understanding of the potential direct effects of antibiotics a role beyond infection, Nat Rev Urol. 12 (2) (2015) 81–90.
during tumor progression is required to better inform clinical decisions [9] P. Mirmonsef, D. Gilbert, M.R. Zariffard, The effects of commensal bacteria on
innate immune responses in the female genital tract, Am. J. Reprod. Immunol. 65
for the treatment of cancer patients. (3) (2011) 190–195.
In tumor-bearing individuals, antibiotics are not the only factor

68
C.M. Buchta Rosean, M.R. Rutkowski Seminars in Immunology 32 (2017) 62–73

[10] M. Avila, D.M. Ojcius, Ö. Yilmaz, The oral microbiota: living with a permanent (10) (2013) e79017.
guest, DNA Cell Biol. 28 (8) (2009) 405–411. [37] J.J. Goedert, G. Jones, X. Hua, X. Xu, G. Yu, R. Flores, R.T. Falk, M.H. Gail, J. Shi,
[11] P. Belda-Ferre, L.D. Alcaraz, R. Cabrera-Rubio, The oral metagenome in health and J. Ravel, H.S. Feigelson, Investigation of the association between the fecal mi-
disease, ISME J. 6 (1) (2012) 46–56. crobiota and breast cancer in postmenopausal women: a population-based case-
[12] M. Hilty, C. Burke, H. Pedro, P. Cardenas, A. Bush, C. Bossley, J. Davies, A. Ervine, control pilot study, J. Natl. Cancer Inst. 107 (8) (2015).
L. Poulter, L. Pachter, M.F. Moffatt, W.O. Cookson, Disordered microbial com- [38] W. Cozen, G. Yu, M.H. Gail, V.K. Ridaura, B.N. Nathwani, A.E. Hwang,
munities in asthmatic airways, PLoS One 5 (1) (2010) e8578. A.S. Hamilton, T.M. Mack, J.I. Gordon, J.J. Goedert, Fecal microbiota diversity in
[13] T.B. Clarke, Early innate immunity to bacterial infection in the lung is regulated survivors of adolescent/young adult Hodgkin lymphoma: a study of twins, Br. J.
systemically by the commensal microbiota via nod-like receptor ligands, Infect. Cancer 108 (5) (2013) 1163–1167.
Immun. 82 (11) (2014) 4596–4606. [39] M.R. Rutkowski, T.L. Stephen, N. Svoronos, M.J. Allegrezza, A.J. Tesone,
[14] M.A. Koch, G.L. Reiner, K.A. Lugo, L.S. Kreuk, A.G. Stanbery, E. Ansaldo, A. Perales-Puchalt, E. Brencicova, X. Escovar-Fadul, J.M. Nguyen, M.G. Cadungog,
T.D. Seher, W.B. Ludington, G.M. Barton, Maternal IgG and IgA antibodies R. Zhang, M. Salatino, J. Tchou, G.A. Rabinovich, J.R. Conejo-Garcia, Microbially
dampen mucosal t helper cell responses in early life, Cell 165 (4) (2016) 827–841. driven TLR5-dependent signaling governs distal malignant progression through
[15] R.A. Dimmitt, E.M. Staley, G. Chuang, The role of postnatal acquisition of the tumor-promoting inflammation, Cancer Cell 27 (1) (2015) 27–40.
intestinal microbiome in the early development of immune function, J. Pediatr. 51 [40] S. Grivennikov, K. Wang, D. Mucida, C. Stewart, B. Schnabl, D. Jauch,
(3) (2010) 262–273. K. Taniguchi, G.-Y. Yu, C. Osterreicher, K. Hung, C. Datz, Y. Feng, E. Fearon,
[16] H. Goris, D.F. Boer, V.D. der Waaij, Myelopoiesis in experimentally contaminated M. Oukka, L. Tessarollo, V. Coppola, F. Yarovinsky, H. Cheroutre, L. Eckmann,
specific-pathogen-free and germfree mice during oral administration of poly- G. Trinchieri, M. Karin, Adenoma-linked barrier defects and microbial products
myxin, Infect. Immun. 50 (2) (1985) 437–441. drive IL-23/IL-17-mediated tumour growth, Nature 491 (7423) (2012) 254–258.
[17] T. Tada, S. Yamamura, Y. Kuwano, T. Abo, Level of myelopoiesis in the bone [41] S. Grivennikov, E. Karin, J. Terzic, D. Mucida, G.Y. Yu, S. Vallabhapurapu,
marrow is influenced by intestinal flora, Cell. Immunol. 173 (1) (1996) 155–161. J. Scheller, S. Rose-John, H. Cheroutre, L. Eckmann, M. Karin, IL-6 and Stat3 are
[18] A.G.P. Nicaise, C. Sandre, F. Forestier, R. Kergot, A.-M. Quero, C. Labarre, required for survival of intestinal epithelial cells and development of colitis-as-
Influence of intestinal microflora on murine bone marrow and spleen macrophage sociated cancer, Cancer Cell 15 (2) (2009) 103–113.
precursors, Scand. J. Immunol. 48 (1998) 585–591. [42] M.G. Rooks, P. Veiga, L.H. Wardwell-Scott, T. Tickle, N. Segata, M. Michaud,
[19] A. Khosravi, A. Yáñez, J.G. Price, A. Chow, M. Merad, Gut microbiota promote C.A. Gallini, C. Beal, J.E. van Hylckama-Vlieg, S.A. Ballal, X.C. Morgan,
hematopoiesis to control bacterial infection, Cell Host Microbe 15 (3) (2014) J.N. Glickman, D. Gevers, C. Huttenhower, W.S. Garrett, Gut microbiome com-
374–381. position and function in experimental colitis during active disease and treatment-
[20] M.L. Balmer, C.M. Schürch, Y. Saito, Microbiota-derived compounds drive steady- induced remission, ISME J. 8 (7) (2014) 1403–1417.
state granulopoiesis via MyD88/TICAM signaling, J. Immunol. 193 (10) (2014) [43] N. Iida, A. Dzutsev, C.A. Stewart, L. Smith, N. Bouladoux, R.A. Weingarten,
5273–5283. D.A. Molina, R. Salcedo, T. Back, S. Cramer, R.M. Dai, H. Kiu, M. Cardone, S. Naik,
[21] T.B. Clarke, K.M. Davis, E.S. Lysenko, A.Y. Zhou, Y. Yu, J.N. Weiser, Recognition A.K. Patri, E. Wang, F.M. Marincola, K.M. Frank, Y. Belkaid, G. Trinchieri,
of peptidoglycan from the microbiota by Nod1 enhances systemic innate im- R.S. Goldszmid, Commensal bacteria control cancer response to therapy by
munity, Nat. Med. 16 (2) (2010) 228–231. modulating the tumor microenvironment, Science 342 (6161) (2013) 967–970.
[22] M. Abt, L. Osborne, L. Monticelli, T. Doering, T. Alenghat, G. Sonnenberg, [44] A. Sivan, L. Corrales, N. Hubert, J.B. Williams, K. Aquino-Michaels, Z.M. Earley,
M. Paley, M. Antenus, K. Williams, J. Erikson, E. Wherry, D. Artis, Commensal F.W. Benyamin, Y.M. Lei, B. Jabri, M.L. Alegre, E.B. Chang, T.F. Gajewski,
bacteria calibrate the activation threshold of innate antiviral immunity, Immunity Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-
37 (1) (2012) 158–170. PD-L1 efficacy, Science 350 (6264) (2015) 1084–1089.
[23] T. Ichinohe, I.K. Pang, Y. Kumamoto, D.R. Peaper, J.H. Ho, T.S. Murray, [45] M. Vetizou, J.M. Pitt, R. Daillere, P. Lepage, N. Waldschmitt, C. Flament,
A. Iwasaki, Microbiota regulates immune defense against respiratory tract influ- S. Rusakiewicz, B. Routy, M.P. Roberti, C.P. Duong, V. Poirier-Colame, A. Roux,
enza A virus infection, Proc. Natl. Acad. Sci. U. S. A. 108 (13) (2011) 5354–5359. S. Becharef, S. Formenti, E. Golden, S. Cording, G. Eberl, A. Schlitzer, F. Ginhoux,
[24] S. Lathrop, S. Bloom, S. Rao, K. Nutsch, C.-W. Lio, N. Santacruz, D. Peterson, S. Mani, T. Yamazaki, N. Jacquelot, D.P. Enot, M. Berard, J. Nigou, P. Opolon,
T. Stappenbeck, C.-S. Hsieh, Peripheral education of the immune system by co- A. Eggermont, P.L. Woerther, E. Chachaty, N. Chaput, C. Robert, C. Mateus,
lonic commensal microbiota, Nature 478 (7368) (2011) 250–254. G. Kroemer, D. Raoult, I.G. Boneca, F. Carbonnel, M. Chamaillard, L. Zitvogel,
[25] N. Arpaia, C. Campbell, X. Fan, S. Dikiy, J. van der Veeken, P. Deroos, H. Liu, Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota,
J. Cross, K. Pfeffer, P. Coffer, A. Rudensky, Metabolites produced by commensal Science 350 (6264) (2015) 1079–1084.
bacteria promote peripheral regulatory T-cell generation, Nature 504 (7480) [46] S. Viaud, F. Saccheri, G. Mignot, T. Yamazaki, R. Daillere, D. Hannani, D.P. Enot,
(2013) 451–455. C. Pfirschke, C. Engblom, M.J. Pittet, A. Schlitzer, F. Ginhoux, L. Apetoh,
[26] Y. Furusawa, Y. Obata, S. Fukuda, T. Endo, G. Nakato, D. Takahashi, Y. Nakanishi, E. Chachaty, P.L. Woerther, G. Eberl, M. Berard, C. Ecobichon, D. Clermont,
C. Uetake, K. Kato, T. Kato, M. Takahashi, N. Fukuda, S. Murakami, E. Miyauchi, C. Bizet, V. Gaboriau-Routhiau, N. Cerf-Bensussan, P. Opolon, N. Yessaad,
S. Hino, K. Atarashi, S. Onawa, Y. Fujimura, T. Lockett, J. Clarke, D. Topping, E. Vivier, B. Ryffel, C.O. Elson, J. Dore, G. Kroemer, P. Lepage, I.G. Boneca,
M. Tomita, S. Hori, O. Ohara, T. Morita, H. Koseki, J. Kikuchi, K. Honda, K. Hase, F. Ghiringhelli, L. Zitvogel, The intestinal microbiota modulates the anticancer
H. Ohno, Commensal microbe-derived butyrate induces the differentiation of co- immune effects of cyclophosphamide, Science 342 (6161) (2013) 971–976.
lonic regulatory T cells, Nature 504 (7480) (2013) 446–450. [47] D.R. Littman, E.G. Pamer, Role of the commensal microbiota in normal and pa-
[27] M.H. Shaw, N. Kamada, Y.-G.G. Kim, G. Núñez, Microbiota-induced IL-1β, but not thogenic host immune responses, Cell Host Microbe 10 (4) (2011) 311–323.
IL-6, is critical for the development of steady-state TH17 cells in the intestine, J. [48] Z. Cai, A. Sanchez, Z. Shi, T. Zhang, M. Liu, D. Zhang, Activation of Toll-like
Exp. Med. 209 (2) (2012) 251–258. receptor 5 on breast cancer cells by flagellin suppresses cell proliferation and
[28] I. Ivanov, K. Atarashi, N. Manel, E. Brodie, T. Shima, U. Karaoz, D. Wei, tumor growth, Cancer Res. 71 (7) (2011) 2466–2475.
K. Goldfarb, C. Santee, S. Lynch, T. Tanoue, A. Imaoka, K. Itoh, K. Takeda, [49] M. Shi, Y. Yao, F. Han, Y. Li, Y. Li, MAP1S controls breast cancer cell TLR5 sig-
Y. Umesaki, K. Honda, D. Littman, Induction of intestinal Th17 cells by segmented naling pathway and promotes TLR5 signaling-based tumor suppression, PLoS One
filamentous bacteria, Cell 139 (3) (2009) 485–498. 9 (1) (2014) e86839.
[29] J. Duan, H. Chung, E. Troy, D.L. Kasper, Microbial colonization drives expansion [50] C. Urbaniak, G.B. Gloor, M. Brackstone, L. Scott, M. Tangney, G. Reid, The mi-
of IL-1 receptor 1-expressing and IL-17-producing γ/δ t cells, Cell Host Microbe 7 crobiota of breast tissue and its association with breast cancer, Appl. Environ.
(2) (2010) 140–150. Microbiol. 82 (16) (2016) 5039–5048.
[30] T. Olszak, D. An, S. Zeissig, M. Vera, J. Richter, A. Franke, J.N. Glickman, [51] T.J. Hieken, J. Chen, T.L. Hoskin, M. Walther-Antonio, S. Johnson, S. Ramaker,
R. Siebert, R.M. Baron, D.L. Kasper, R.S. Blumberg, Microbial exposure during J. Xiao, D.C. Radisky, K.L. Knutson, K.R. Kalari, J.Z. Yao, L.M. Baddour, N. Chia,
early life has persistent effects on natural killer t cell function, Science 336 (6080) A.C. Degnim, The microbiome of aseptically collected human breast tissue in be-
(2012) 489–493. nign and malignant disease, Sci. Rep. 6 (2016) 30751.
[31] B. Wei, G. Wingender, D. Fujiwara, D. Chen, M. McPherson, S. Brewer, [52] A. Bashir, A.Y. Miskeen, Y.M. Hazari, S. Asrafuzzaman, K.M. Fazili, Fusobacterium
J. Borneman, M. Kronenberg, J. Braun, Commensal microbiota and C.D8+ T cells nucleatum, inflammation, and immunity: the fire within human gut, Tumour Biol.
shape the formation of invariant NKT cells, J. Immunol. 184 (3) (2010) 37 (3) (2016) 2805–2810.
1218–1226. [53] A.D. Kostic, E. Chun, L. Robertson, J.N. Glickman, C.A. Gallini, M. Michaud,
[32] M. Schirmer, S.P. Smeekens, H. Vlamakis, M. Jaeger, M. Oosting, E.A. Franzosa, T.E. Clancy, D.C. Chung, P. Lochhead, G.L. Hold, E.M. El-Omar, D. Brenner,
T. Jansen, L. Jacobs, M.J. Bonder, A. Kurilshikov, J. Fu, L.A. Joosten, C.S. Fuchs, M. Meyerson, W.S. Garrett, Fusobacterium nucleatum potentiates in-
A. Zhernakova, C. Huttenhower, C. Wijmenga, M.G. Netea, R.J. Xavier, Linking testinal tumorigenesis and modulates the tumor-immune microenvironment, Cell
the human gut microbiome to inflammatory cytokine production capacity, Cell Host Microbe 14 (2) (2013) 207–215.
167 (4) (2016) 1125–1930850304. [54] M.R. Walther-Antonio, J. Chen, F. Multinu, A. Hokenstad, T.J. Distad, E.H. Cheek,
[33] J. Gagniere, J. Raisch, J. Veziant, N. Barnich, R. Bonnet, E. Buc, M.A. Bringer, G.L. Keeney, D.J. Creedon, H. Nelson, A. Mariani, N. Chia, Potential contribution
D. Pezet, M. Bonnet, Gut microbiota imbalance and colorectal cancer, World J. of the uterine microbiome in the development of endometrial cancer, Genome
Gastroenterol. 22 (2) (2016) 501–518. Med. 8 (1) (2016) 122.
[34] C. Xuan, J.M. Shamonki, A. Chung, M.L. DiNome, Microbial dysbiosis is associated [55] H. Borgdorff, R. Gautam, S.D. Armstrong, D. Xia, G.F. Ndayisaba, N.H. van
with human breast cancer, PLoS One 9 (1) (2014) e83744. Teijlingen, T.B. Geijtenbeek, J.M. Wastling, J.H. van de Wijgert, Cervicovaginal
[35] D. Chase, A. Goulder, F. Zenhausern, B. Monk, The vaginal and gastrointestinal microbiome dysbiosis is associated with proteome changes related to alterations of
microbiomes in gynecologic cancers: a review of applications in etiology, symp- the cervicovaginal mucosal barrier, Mucosal Immunol. 9 (3) (2016) 621–633.
toms and treatment, Gynecologic 138 (1) (2015) 190–200. [56] B.L. Anderson, S. Cu-Uvin, C.A. Raker, C. Fitzsimmons, S.L. Hillier, Subtle per-
[36] X.T. Zeng, A.P. Deng, C. Li, L.Y. Xia, Y.M. Niu, W.D. Leng, Periodontal disease and turbations of genital microflora alter mucosal immunity among low-risk pregnant
risk of head and neck cancer: a meta-analysis of observational studies, PLoS One 8 women, Acta Obstet. Gynecol. Scand. 90 (5) (2011) 510–515.

69
C.M. Buchta Rosean, M.R. Rutkowski Seminars in Immunology 32 (2017) 62–73

[57] C. Scholz, U. Andergassen, P. Hepp, C. Schindlbeck, T.W.P. Friedl, N. Harbeck, (2003) 1563–1572.
M. Kiechle, H. Sommer, H. Hauner, K. Friese, Obesity as an independent risk factor [83] H. Zhu, W.Y. Xu, Z. Hu, H. Zhang, Y. Shen, S. Lu, C. Wei, Z.G. Wang, RNA virus
for decreased survival in node-positive high-risk breast cancer, Breast Cancer Res. receptor Rig-I monitors gut microbiota and inhibits colitis-associated colorectal
Treat. 151 (3) (2015) 569–576. cancer, J. Exp. Clin. Cancer Res. 36 (1) (2017) 2.
[58] Z. Liu, T.T. Zhang, J.J. Zhao, S.F. Qi, P. Du, D.W. Liu, Q.B. Tian, The association [84] A. Couturier-Maillard, T. Secher, A. Rehman, S. Normand, A. De Arcangelis,
between overweight, obesity and ovarian cancer: a meta-analysis, Jpn. J. Clin. R. Haesler, L. Huot, T. Grandjean, A. Bressenot, A. Delanoye-Crespin, O. Gaillot,
Oncol. 45 (12) (2015) 1107–1115. S. Schreiber, Y. Lemoine, B. Ryffel, D. Hot, G. Nunez, G. Chen, P. Rosenstiel,
[59] M.A. Onstad, R.E. Schmandt, K.H. Lu, Addressing the role of obesity in en- M. Chamaillard, NOD2-mediated dysbiosis predisposes mice to transmissible co-
dometrial cancer risk, prevention, and treatment, J. Clin. Oncol. 34 (35) (2016) litis and colorectal cancer, J. Clin. Invest. 123 (2) (2013) 700–711.
4225–4230. [85] J. da Correia, Y. Miranda, N. Austin-Brown, J. Hsu, J. Mathison, R. Xiang, H. Zhou,
[60] C. Jochem, M. Leitzmann, Obesity and colorectal cancer, Recent Results Cancer Q. Li, J. Han, R.J. Ulevitch, Nod1-dependent control of tumor growth, Proc. Natl.
Res. 208 (2016) 17–41. Acad. Sci. U. S. A. 103 (6) (2006) 1840–1845.
[61] J.M. Ridlon, D.-J.J. Kang, P.B. Hylemon, Bile salt biotransformations by human [86] J. da Silva Correia, Y. Miranda, N. Leonard, J. Hsu, R.J. Ulevitch, Regulation of
intestinal bacteria, J. Lipid Res. 47 (2) (2006) 241–259. Nod1-mediated signaling pathways, Cell Death Differ. 14 (4) (2007) 830–839.
[62] L.E. Chatelier, T. Nielsen, J. Qin, E. Prifti, F. Hildebrand, Richness of human gut [87] T. Obata, Y. Goto, J. Kunisawa, S. Sato, M. Sakamoto, H. Setoyama, T. Matsuki,
microbiome correlates with metabolic markers, Nature 500 (7464) (2013) K. Nonaka, N. Shibata, M. Gohda, Y. Kagiyama, T. Nochi, Y. Yuki, Y. Fukuyama,
541–546. A. Mukai, S. Shinzaki, K. Fujihashi, C. Sasakawa, H. Iijima, M. Goto, Y. Umesaki,
[63] M.D. Schulz, C. Atay, J. Heringer, F.K. Romrig, S. Schwitalla, B. Aydin, Y. Benno, H. Kiyono, Indigenous opportunistic bacteria inhabit mammalian gut-
P.K. Ziegler, J. Varga, W. Reindl, C. Pommerenke, G. Salinas-Riester, A. Bock, associated lymphoid tissues and share a mucosal antibody-mediated symbiosis,
C. Alpert, M. Blaut, S.C. Polson, L. Brandl, T. Kirchner, F.R. Greten, S.W. Polson, Proc. Natl. Acad. Sci. U. S. A. 107 (16) (2010) 7419–7424.
M.C. Arkan, High-fat-diet-mediated dysbiosis promotes intestinal carcinogenesis [88] G.F. Sonnenberg, L.A. Monticelli, T. Alenghat, T.C. Fung, N.A. Hutnick,
independently of obesity, Nature 514 (7523) (2014) 508–512. J. Kunisawa, N. Shibata, S. Grunberg, R. Sinha, A.M. Zahm, M.R. Tardif,
[64] R. Canani, M. Costanzo, L. Leone, The epigenetic effects of butyrate: potential T. Sathaliyawala, M. Kubota, D.L. Farber, R.G. Collman, A. Shaked, L.A. Fouser,
therapeutic implications for clinical practice, Clin. Epigenet. 4 (1) (2012) 4. D.B. Weiner, P.A. Tessier, J.R. Friedman, H. Kiyono, F.D. Bushman, K.M. Chang,
[65] S. Yoshimoto, T.M. Loo, K. Atarashi, H. Kanda, S. Sato, S. Oyadomari, Y. Iwakura, D. Artis, Innate lymphoid cells promote anatomical containment of lymphoid-re-
K. Oshima, H. Morita, M. Hattori, K. Honda, Y. Ishikawa, E. Hara, N. Ohtani, sident commensal bacteria, Science (New York, N.Y.) 336 (6086) (2012)
Obesity-induced gut microbial metabolite promotes liver cancer through senes- 1321–1325.
cence secretome, Nature 499 (7456) (2013) 97–101. [89] M.Y. Zeng, D. Cisalpino, S. Varadarajan, J. Hellman, H.S. Warren, M. Cascalho,
[66] S. Kitazawa, A. Denda, M. Tsutsumi, T. Tsujiuchi, K. Hasegawa, K. Tamura, N. Inohara, G. Nunez, Gut microbiota-Induced immunoglobulin G controls sys-
H. Maruyama, Y. Konishi, Enhanced preneoplastic liver lesion development under temic infection by symbiotic bacteria and pathogens, Immunity 44 (3) (2016)
‘selection pressure' conditions after administration of deoxycholic or lithocholic 647–658.
acid in the initiation phase in rats, Carcinogenesis 11 (8) (1990) 1323–1328. [90] Q. Dong, D.E. Nelson, E. Toh, L. Diao, X. Gao, J.D. Fortenberry, B. Van der Pol, The
[67] M.J. Blaser, Antibiotic use and its consequences for the normal microbiome, microbial communities in male first catch urine are highly similar to those in
Science 352 (6285) (2016) 544–545. paired urethral swab specimens, PLoS One 6 (5) (2011) e19709.
[68] K.M. Keeney, S. Yurist-Doutsch, M.C. Arrieta, B.B. Finlay, Effects of antibiotics on [91] D.E. Fouts, R. Pieper, S. Szpakowski, H. Pohl, S. Knoblach, M.J. Suh, S.T. Huang,
human microbiota and subsequent disease, Annu. Rev. Microbiol. 68 (2014) I. Ljungberg, B.M. Sprague, S.K. Lucas, M. Torralba, K.E. Nelson, S.L. Groah,
217–235. Integrated next-generation sequencing of 16S rDNA and metaproteomics differ-
[69] C.T. Peterson, V. Sharma, L. Elmén, S.N. Peterson, Immune homeostasis, dysbiosis entiate the healthy urine microbiome from asymptomatic bacteriuria in neuro-
and therapeutic modulation of the gut microbiota, Clin. Exp. Immunol. 179 (3) pathic bladder associated with spinal cord injury, J. Transl. Med. 10 (2012) 174.
(2015) 363–377. [92] E.E. Hilt, K. McKinley, M.M. Pearce, A.B. Rosenfeld, M.J. Zilliox, E.R. Mueller,
[70] L.A. Hicks, M.G. Bartoces, R.M. Roberts, K.J. Suda, R.J. Hunkler, T.H. Taylor, L. Brubaker, X. Gai, A.J. Wolfe, P.C. Schreckenberger, Urine is not sterile: use of
S.J. Schrag, US outpatient antibiotic prescribing variation according to geography, enhanced urine culture techniques to detect resident bacterial flora in the adult
patient population, and provider specialty in 2011, Clin. Infect. Dis. 60 (9) (2015) female bladder, J. Clin. Microbiol. 52 (3) (2014) 871–876.
1308–1316. [93] D.A. Lewis, R. Brown, J. Williams, P. White, S.K. Jacobson, J.R. Marchesi,
[71] C.M. Velicer, S.R. Heckbert, J.W. Lampe, J.D. Potter, C.A. Robertson, S.H. Taplin, M.J. Drake, The human urinary microbiome; bacterial DNA in voided urine of
Antibiotic use in relation to the risk of breast cancer, JAMA 291 (7) (2004) asymptomatic adults, Front. Cell. Infect. Microbiol. 3 (2013) 41.
827–835. [94] C.M. Paulos, C. Wrzesinski, A. Kaiser, C.S. Hinrichs, M. Chieppa, L. Cassard,
[72] G.D. Friedman, N. Oestreicher, J. Chan, C.P. Quesenberry Jr., N. Udaltsova, D.C. Palmer, A. Boni, P. Muranski, Z. Yu, L. Gattinoni, P.A. Antony,
L.A. Habel, Antibiotics and risk of breast cancer: up to 9 years of follow-up of 2.1 S.A. Rosenberg, N.P. Restifo, Microbial translocation augments the function of
million women, Cancer Epidemiol. Biomark. Prev. 15 (11) (2006) 2102–2106. adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling, J.
[73] B. Boursi, R. Mamtani, K. Haynes, Y.X. Yang, Recurrent antibiotic exposure may Clin. Invest. 117 (8) (2007) 2197–2204.
promote cancer formation–another step in understanding the role of the human [95] H. Fukui, B. Brauner, J.C. Bode, C. Bode, Plasma endotoxin concentrations in
microbiota? Eur. J. Cancer 51 (17) (2015) 2655–2664. patients with alcoholic and non-alcoholic liver disease: reevaluation with an im-
[74] N. Moullan, L. Mouchiroud, X. Wang, D. Ryu, E.G. Williams, A. Mottis, proved chromogenic assay, J. Hepatol. 12 (2) (1991) 162–169.
V. Jovaisaite, M.V. Frochaux, P.M. Quiros, B. Deplancke, R.H. Houtkooper, [96] W.W. Li, G. Grayson, J. Folkman, P.A. D'Amore, Sustained-release endotoxin. A
J. Auwerx, Tetracyclines disturb mitochondrial function across eukaryotic models: model for inducing corneal neovascularization, Invest. Ophthalmol. Vis. Sci. 32
a call for caution in biomedical research, Cell Rep. (2015). (11) (1991) 2906–2911.
[75] S. Kalghatgi, C.S. Spina, J.C. Costello, M. Liesa, J.R. Morones-Ramirez, [97] I. Mattsby-Baltzer, A. Jakobsson, J. Sorbo, K. Norrby, Endotoxin is angiogenic, Int.
S. Slomovic, A. Molina, O.S. Shirihai, J.J. Collins, Bactericidal antibiotics induce J. Exp. Pathol. 75 (3) (1994) 191–196.
mitochondrial dysfunction and oxidative damage in Mammalian cells, Sci. Transl. [98] J.H. Harmey, C.D. Bucana, W. Lu, A.M. Byrne, S. McDonnell, C. Lynch,
Med. 5 (192) (2013) 192ra85. D. Bouchier-Hayes, Z. Dong, Lipopolysaccharide-induced metastatic growth is
[76] M. Cheng, L. Qian, G. Shen, G. Bian, T. Xu, W. Xu, G. Shen, S. Hu, Microbiota associated with increased angiogenesis, vascular permeability and tumor cell in-
modulate tumoral immune surveillance in lung through a gammadeltaT17 im- vasion, Int. J. Cancer 101 (5) (2002) 415–422.
mune cell-dependent mechanism, Cancer Res. 74 (15) (2014) 4030–4041. [99] S. Tanabe, M. Yoshioka, D. Hinode, D. Grenier, Subinhibitory concentrations of
[77] Y.-G.G. Kim, K.G. Udayanga, N. Totsuka, J.B. Weinberg, G. Núñez, A. Shibuya, Gut tetracyclines induce lipopolysaccharide shedding by Porphyromonas gingivalis
dysbiosis promotes M2 macrophage polarization and allergic airway inflammation and modulate the host inflammatory response, J. Periodontal Res. 49 (5) (2014)
via fungi-induced PGE2, Cell Host Microbe 15 (1) (2014) 95–102. 603–608.
[78] R. Blekhman, J.K. Goodrich, K. Huang, Q. Sun, R. Bukowski, J.T. Bell, [100] R.H. Eng, S.M. Smith, P. Fan-Havard, T. Ogbara, Effect of antibiotics on endotoxin
T.D. Spector, A. Keinan, R.E. Ley, D. Gevers, A.G. Clark, Host genetic variation release from gram-negative bacteria, Diagn. Microbiol. Infect. Dis. 16 (3) (1993)
impacts microbiome composition across human body sites, Genome Biol. 16 185–189.
(2015) 191. [101] H.Y. Hsu, M.H. Wen, Lipopolysaccharide-mediated reactive oxygen species and
[79] M. Vijay-Kumar, J.D. Aitken, F.A. Carvalho, T.C. Cullender, S. Mwangi, signal transduction in the regulation of interleukin-1 gene expression, J. Biol.
S. Srinivasan, S.V. Sitaraman, R. Knight, R.E. Ley, A.T. Gewirtz, Metabolic syn- Chem. 277 (25) (2002) 22131–22139.
drome and altered gut microbiota in mice lacking toll-like receptor 5, Science 328 [102] F. Simon, R. Fernandez, Early lipopolysaccharide-induced reactive oxygen species
(5975) (2010) 228–231. production evokes necrotic cell death in human umbilical vein endothelial cells, J.
[80] J. Henao-Mejia, E. Elinav, C. Jin, L. Hao, W.Z. Mehal, T. Strowig, C.A. Thaiss, Hypertens. 27 (6) (2009) 1202–1216.
A.L. Kau, S.C. Eisenbarth, M.J. Jurczak, J.P. Camporez, G.I. Shulman, J.I. Gordon, [103] I.L. Glukhov, N.P. Sirota, E.A. Kuznetsova, DNA damage in human mononuclear
H.M. Hoffman, R.A. Flavell, Inflammasome-mediated dysbiosis regulates pro- cells induced by bacterial endotoxin, Bull. Exp. Biol. Med. 146 (3) (2008)
gression of NAFLD and obesity, Nature 482 (7384) (2012) 179–185. 301–303.
[81] W.S. Garrett, G.M. Lord, S. Punit, G. Lugo-Villarino, S.K. Mazmanian, S. Ito, [104] K. Monden, S. Arii, S. Itai, T. Sasaoki, Y. Adachi, N. Funaki, H. Higashitsuji,
J.N. Glickman, L.H. Glimcher, Communicable ulcerative colitis induced by T-bet T. Tobe, Enhancement and hepatocyte-modulating effect of chemical mediators
deficiency in the innate immune system, Cell 131 (1) (2007) 33–45. and monokines produced by hepatic macrophages in rats with induced sepsis, Res.
[82] T.R. Hawn, A. Verbon, K.D. Lettinga, L.P. Zhao, S.S. Li, R.J. Laws, S.J. Skerrett, Exp. Med. (Berl.) 191 (3) (1991) 177–187.
B. Beutler, L. Schroeder, A. Nachman, A. Ozinsky, K.D. Smith, A. Aderem, A [105] S.P. Ding, J.C. Li, C. Jin, A mouse model of severe acute pancreatitis induced with
common dominant TLR5 stop codon polymorphism abolishes flagellin signaling caerulein and lipopolysaccharide, World J. Gastroenterol. 9 (3) (2003) 584–589.
and is associated with susceptibility to legionnaires' disease, J. Exp. Med. 198 (10) [106] A. Ochi, A.H. Nguyen, A.S. Bedrosian, H.M. Mushlin, S. Zarbakhsh, R. Barilla,

70
C.M. Buchta Rosean, M.R. Rutkowski Seminars in Immunology 32 (2017) 62–73

C.P. Zambirinis, N.C. Fallon, A. Rehman, Y. Pylayeva-Gupta, S. Badar, C.H. Hajdu, cancer cells, PLoS One 5 (5) (2010) e10850.
A.B. Frey, D. Bar-Sagi, G. Miller, MyD88 inhibition amplifies dendritic cell capa- [130] J.L. Round, S.M. Lee, J. Li, G. Tran, B. Jabri, T.A. Chatila, S.K. Mazmanian, The
city to promote pancreatic carcinogenesis via Th2 cells, J. Exp. Med. 209 (9) Toll-like receptor 2 pathway establishes colonization by a commensal of the
(2012) 1671–1687. human microbiota, Science (New York, N.Y.) 332 (6032) (2011) 974–977.
[107] J. Daniluk, Y. Liu, D. Deng, J. Chu, H. Huang, S. Gaiser, Z. Cruz-Monserrate, [131] J.L. Round, S.K. Mazmanian, Inducible Foxp3+ regulatory T-cell development by
H. Wang, B. Ji, C.D. Logsdon, An NF-kappaB pathway-mediated positive feedback a commensal bacterium of the intestinal microbiota, Proc. Natl. Acad. Sci. U. S. A.
loop amplifies Ras activity to pathological levels in mice, J. Clin. Invest. 122 (4) 107 (27) (2010) 12204–12209.
(2012) 1519–1528. [132] K.M. Telesford, W. Yan, J. Ochoa-Reparaz, A. Pant, C. Kircher, M.A. Christy,
[108] L. Yan, C. McFaul, N. Howes, J. Leslie, G. Lancaster, T. Wong, J. Threadgold, S. Begum-Haque, D.L. Kasper, L.H. Kasper, A commensal symbiotic factor derived
J. Evans, I. Gilmore, H. Smart, M. Lombard, J. Neoptolemos, W. Greenhalf, from Bacteroides fragilis promotes human CD39(+)Foxp3(+) T cells and Treg
Molecular analysis to detect pancreatic ductal adenocarcinoma in high-risk function, Gut Microbes 6 (4) (2015) 234–242.
groups, Gastroenterology 128 (7) (2005) 2124–2130. [133] Y. Sato, Y. Goto, N. Narita, D.S. Hoon, Cancer cells expressing toll-like receptors
[109] M.S. Yakubovskaya, V. Spiegelman, F.C. Luo, S. Malaev, A. Salnev, I. Zborovskaya, and the tumor microenvironment, Cancer Microenviron. 2 (Suppl. 1) (2009)
A. Gasparyan, B. Polotsky, Z. Machaladze, A.C. Trachtenberg, et al., High fre- 205–214.
quency of K-ras mutations in normal appearing lung tissues and sputum of patients [134] F. Hietbrink, M.G. Besselink, W. Renooij, M.B. de Smet, A. Draisma, H. van der
with lung cancer, Int. J. Cancer 63 (6) (1995) 810–814. Hoeven, P. Pickkers, Systemic inflammation increases intestinal permeability
[110] L. Yan, Q. Cai, Y. Xu, The ubiquitin-CXCR4 axis plays an important role in acute during experimental human endotoxemia, Shock 32 (4) (2009) 374–378.
lung infection-enhanced lung tumor metastasis, Clin. Cancer Res. 19 (17) (2013) [135] C. Bode, V. Kugler, J.C. Bode, Endotoxemia in patients with alcoholic and non-
4706–4716. alcoholic cirrhosis and in subjects with no evidence of chronic liver disease fol-
[111] G.P. Pidgeon, J.H. Harmey, E. Kay, M. Da Costa, H.P. Redmond, D.J. Bouchier- lowing acute alcohol excess, J. Hepatol. 4 (1) (1987) 8–14.
Hayes, The role of endotoxin/lipopolysaccharide in surgically induced tumour [136] J.M. Moreno-Navarrete, F. Ortega, M. Serino, E. Luche, A. Waget, G. Pardo,
growth in a murine model of metastatic disease, Br. J. Cancer 81 (8) (1999) J. Salvador, W. Ricart, G. Fruhbeck, R. Burcelin, J.M. Fernandez-Real, Circulating
1311–1317. lipopolysaccharide-binding protein (LBP) as a marker of obesity-related insulin
[112] F. Vidal-Vanaclocha, A. Alvarez, A. Asumendi, B. Urcelay, P. Tonino, resistance, Int. J. Obesity (2005) 36 (11) (2012) 442–449.
C.A. Dinarello, Interleukin 1 (IL-1)-dependent melanoma hepatic metastasis in [137] B. Jayashree, Y.S. Bibin, D. Prabhu, C.S. Shanthirani, K. Gokulakrishnan,
vivo; increased endothelial adherence by IL-1-induced mannose receptors and B.S. Lakshmi, V. Mohan, M. Balasubramanyam, Increased circulatory levels of li-
growth factor production in vitro, J. Natl. Cancer Inst. 88 (3–4) (1996) 198–205. popolysaccharide (LPS) and zonulin signify novel biomarkers of proinflammation
[113] N. Simiantonaki, C. Jayasinghe, C.J. Kirkpatrick, Effect of pro-inflammatory sti- in patients with type 2 diabetes, Mol. Cell. Biochem. 388 (1–2) (2014) 203–210.
muli on tumor cell-mediated induction of endothelial cell adhesion molecules in [138] L. Gattinoni, S.E. Finkelstein, C.A. Klebanoff, P.A. Antony, D.C. Palmer, P.J. Spiess,
vitro, Exp. Mol. Pathol. 73 (1) (2002) 46–53. L.N. Hwang, Z. Yu, C. Wrzesinski, D.M. Heimann, C.D. Surh, S.A. Rosenberg,
[114] N. Simiantonaki, C. Jayasinghe, C.J. Kirkpatrick, Differential endothelial CAM- N.P. Restifo, Removal of homeostatic cytokine sinks by lymphodepletion enhances
expression after stimulation with supernatants of LPS- and cytokine-stimulated the efficacy of adoptively transferred tumor-specific CD8+ T cells, J. Exp. Med.
HT-29 and ST-ML-12 tumor cells growing as monolayer cultures and multicellular 202 (7) (2005) 907–912.
spheroids, Anticancer Res. 22 (5) (2002) 2641–2649. [139] A. Sistigu, S. Viaud, N. Chaput, L. Bracci, E. Proietti, L. Zitvogel,
[115] H. Yang, B. Wang, T. Wang, L.J. Xu, C.Y. He, H.Y. Wen, J. Yan, H.H. Su, X.M. Zhu, Immunomodulatory effects of cyclophosphamide and implementations for vaccine
Toll-Like receptor 4 prompts human breast cancer cells invasiveness via lipopo- design, Semin. Immunopathol. 33 (4) (2011) 369–383.
lysaccharide stimulation and is overexpressed in patients with lymph node me- [140] S. Viaud, C. Flament, M. Zoubir, P. Pautier, A. LeCesne, V. Ribrag, J.C. Soria,
tastasis, PLoS One 9 (10) (2014). V. Marty, P. Vielh, C. Robert, N. Chaput, L. Zitvogel, Cyclophosphamide induces
[116] J. Svard, D. Paquin-Proulx, M. Buggert, K. Noyan, B. Barqasho, A. Sonnerborg, differentiation of Th17 cells in cancer patients, Cancer Res. 71 (3) (2011)
P. Nowak, Role of translocated bacterial flagellin in monocyte activation among 661–665.
individuals with chronic HIV-1 infection, Clin. Immunol. (Orlando Fla.) 161 (2) [141] J. Qin, R. Li, J. Raes, M. Arumugam, K.S. Burgdorf, C. Manichanh, T. Nielsen,
(2015) 180–189. N. Pons, F. Levenez, T. Yamada, D.R. Mende, J. Li, J. Xu, S. Li, D. Li, J. Cao,
[117] T.R. Ziegler, M. Luo, C.F. Estivariz, D.A. Moore 3rd, S.V. Sitaraman, L. Hao, B. Wang, H. Liang, H. Zheng, Y. Xie, J. Tap, P. Lepage, M. Bertalan, J.M. Batto,
N. Bazargan, J.M. Klaroth, J. Tian, J.R. Galloway, L.M. Leader, D. Jones, T. Hansen, D. Le Paslier, A. Linneberg, H.B. Nielsen, E. Pelletier, P. Renault,
A.T. Gewirtz, Detectable serum flagellin and lipopolysaccharide and upregulated T. Sicheritz-Ponten, K. Turner, H. Zhu, C. Yu, S. Li, M. Jian, Y. Zhou, Y. Li,
anti-flagellin and lipopolysaccharide immunoglobulins in human short bowel X. Zhang, S. Li, N. Qin, H. Yang, J. Wang, S. Brunak, J. Dore, F. Guarner,
syndrome, Am. J. Physiol. 294 (2) (2008) R402–R410. K. Kristiansen, O. Pedersen, J. Parkhill, J. Weissenbach, P. Bork, S.D. Ehrlich,
[118] L. Grimes, A. Doyle, A.L. Miller, R.B. Pyles, G. Olah, C. Szabo, S. Hoskins, T. Eaves- J. Wang, A human gut microbial gene catalogue established by metagenomic se-
Pyles, Intraluminal flagellin differentially contributes to gut dysbiosis and sys- quencing, Nature 464 (7285) (2010) 59–65.
temic inflammation following burn injury, PLoS One 11 (12) (2016) e0166770. [142] J.R. Brestoff, D. Artis, Commensal bacteria at the interface of host metabolism and
[119] A. Didierlaurent, I. Ferrero, L.A. Otten, B. Dubois, M. Reinhardt, H. Carlsen, the immune system, Nat. Immunol. 14 (7) (2013) 676–684.
R. Blomhoff, S. Akira, J.P. Kraehenbuhl, J.C. Sirard, Flagellin promotes myeloid [143] W.R. Wikoff, A.T. Anfora, J. Liu, P.G. Schultz, S.A. Lesley, E.C. Peters, G. Siuzdak,
differentiation factor 88-dependent development of Th2-type response, J. Metabolomics analysis reveals large effects of gut microflora on mammalian blood
Immunol. 172 (11) (2004) 6922–6930. metabolites, Proc. Natl. Acad. Sci. U. S. A. 106 (10) (2009) 3698–3703.
[120] N. Rieber, A. Brand, A. Hector, U. Graepler-Mainka, M. Ost, I. Schafer, I. Wecker, [144] V.R. Velagapudi, R. Hezaveh, C.S. Reigstad, P. Gopalacharyulu, L. Yetukuri,
D. Neri, A. Wirth, L. Mays, S. Zundel, J. Fuchs, R. Handgretinger, M. Stern, S. Islam, J. Felin, R. Perkins, J. Boren, M. Oresic, F. Backhed, The gut microbiota
M. Hogardt, G. Doring, J. Riethmuller, M. Kormann, D. Hartl, Flagellin induces modulates host energy and lipid metabolism in mice, J. Lipid Res. 51 (5) (2010)
myeloid-derived suppressor cells: implications for Pseudomonas aeruginosa in- 1101–1112.
fection in cystic fibrosis lung disease, J. Immunol. 190 (3) (2013) 1276–1284. [145] A.J. Brown, S.M. Goldsworthy, A.A. Barnes, M.M. Eilert, L. Tcheang, D. Daniels,
[121] E. Hoste, E.N. Arwert, R. Lal, A.P. South, J.C. Salas-Alanis, D.F. Murrell, G. Donati, A.I. Muir, M.J. Wigglesworth, I. Kinghorn, N.J. Fraser, N.B. Pike, J.C. Strum,
F.M. Watt, Innate sensing of microbial products promotes wound-induced skin K.M. Steplewski, P.R. Murdock, J.C. Holder, F.H. Marshall, P.G. Szekeres,
cancer, Nat. Commun. 6 (2015) 5932. S. Wilson, D.M. Ignar, S.M. Foord, A. Wise, S.J. Dowell, The Orphan G protein-
[122] H.Y. Cho, S.W. Lee, TLR5 activation by flagellin induces doxorubicin resistance via coupled receptors GPR41 and GPR43 are activated by propionate and other short
interleukin-6 (IL-6) expression in two multiple myeloma cells, Cell. Immunol. 289 chain carboxylic acids, J. Biol. Chem. 278 (13) (2003) 11312–11319.
(1-2) (2014) 27–35. [146] L.E. Willemsen, M.A. Koetsier, S.J. van Deventer, E.A. van Tol, Short chain fatty
[123] E.J. Song, M.J. Kang, Y.S. Kim, S.M. Kim, S.E. Lee, C.H. Kim, D.J. Kim, J.H. Park, acids stimulate epithelial mucin 2 expression through differential effects on
Flagellin promotes the proliferation of gastric cancer cells via the Toll-like receptor prostaglandin E(1) and E(2) production by intestinal myofibroblasts, Gut 52 (10)
5, Int. J. Mol. Med. 28 (1) (2011) 115–119. (2003) 1442–1447.
[124] J.H. Park, H.E. Yoon, D.J. Kim, S.A. Kim, S.G. Ahn, J.H. Yoon, Toll-like receptor 5 [147] C. Nastasi, M. Candela, C.M. Bonefeld, C. Geisler, M. Hansen, T. Krejsgaard,
activation promotes migration and invasion of salivary gland adenocarcinoma, J. E. Biagi, M.H. Andersen, P. Brigidi, N. Odum, T. Litman, A. Woetmann, The effect
Oral Pathol. Med. 40 (2) (2011) 187–193. of short-chain fatty acids on human monocyte-derived dendritic cells, Sci. Rep. 5
[125] S.H. Rhee, E. Im, C. Pothoulakis, Toll-like receptor 5 engagement modulates tumor (2015) 16148.
development and growth in a mouse xenograft model of human colon cancer, [148] M. Usami, K. Kishimoto, A. Ohata, M. Miyoshi, M. Aoyama, Y. Fueda, J. Kotani,
Gastroenterology 135 (2) (2008) 518–528. Butyrate and trichostatin A attenuate nuclear factor kappaB activation and tumor
[126] H. Zhou, J.H. Chen, J. Hu, Y.Z. Luo, F. Li, L. Xiao, M.Z. Zhong, High expression of necrosis factor alpha secretion and increase prostaglandin E2 secretion in human
Toll-like receptor 5 correlates with better prognosis in non-small-cell lung cancer: peripheral blood mononuclear cells, Nutr. Res. (New York, N.Y.) 28 (5) (2008)
an anti-tumor effect of TLR5 signaling in non-small cell lung cancer, J. Cancer Res. 321–328.
Clin. Oncol. 140 (4) (2014) 633–643. [149] M.A. Vinolo, H.G. Rodrigues, E. Hatanaka, F.T. Sato, S.C. Sampaio, R. Curi,
[127] L. Sfondrini, A. Rossini, D. Besusso, A. Merlo, E. Tagliabue, S. Menard, A. Balsari, Suppressive effect of short-chain fatty acids on production of proinflammatory
Antitumor activity of the TLR-5 ligand flagellin in mouse models of cancer, J. mediators by neutrophils, J. Nutr. Biochem. 22 (9) (2011) 849–855.
Immunol. (Baltim., Md.: 1950) 176 (11) (2006) 6624–6630. [150] M.A. Vinolo, G.J. Ferguson, S. Kulkarni, G. Damoulakis, K. Anderson,
[128] X.L. Xu, R.T. Lee, H.M. Fang, Y.M. Wang, R. Li, H. Zou, Y. Zhu, Y. Wang, Bacterial Y.M. Bohlooly, L. Stephens, P.T. Hawkins, R. Curi, SCFAs induce mouse neutrophil
peptidoglycan triggers Candida albicans hyphal growth by directly activating the chemotaxis through the GPR43 receptor, PLoS One 6 (6) (2011) e21205.
adenylyl cyclase Cyr1p, Cell Host Microbe 4 (1) (2008) 28–39. [151] P.V. Chang, L. Hao, S. Offermanns, R. Medzhitov, The microbial metabolite bu-
[129] W. Xie, Y. Huang, W. Xie, A. Guo, W. Wu, Bacteria peptidoglycan promoted breast tyrate regulates intestinal macrophage function via histone deacetylase inhibition,
cancer cell invasiveness and adhesiveness by targeting toll-like receptor 2 in the Proc. Natl. Acad. Sci. U. S. A. 111 (6) (2014) 2247–2252.

71
C.M. Buchta Rosean, M.R. Rutkowski Seminars in Immunology 32 (2017) 62–73

[152] N. Singh, M. Thangaraju, P.D. Prasad, P.M. Martin, N.A. Lambert, T. Boettger, chain fatty acids on human colon cancer cell phenotype are associated with his-
S. Offermanns, V. Ganapathy, Blockade of dendritic cell development by bacterial tone hyperacetylation, J. Nutr. 132 (5) (2002) 1012–1017.
fermentation products butyrate and propionate through a transporter (Slc5a8)- [173] Y.H. Kim, J.W. Park, J.Y. Lee, T.K. Kwon, Sodium butyrate sensitizes TRAIL-
dependent inhibition of histone deacetylases, J. Biol. Chem. 285 (36) (2010) mediated apoptosis by induction of transcription from the DR5 gene promoter
27601–27608. through Sp1 sites in colon cancer cells, Carcinogenesis 25 (10) (2004) 1813–1820.
[153] A. Trompette, E.S. Gollwitzer, K. Yadava, A.K. Sichelstiel, N. Sprenger, C. Ngom- [174] J. Qiu, Z. Gao, H. Shima, Growth of human prostate cancer cells is significantly
Bru, C. Blanchard, T. Junt, L.P. Nicod, N.L. Harris, B.J. Marsland, Gut microbiota suppressed in vitro with sodium butyrate through apoptosis, Oncol. Rep. 27 (1)
metabolism of dietary fiber influences allergic airway disease and hematopoiesis, (2012) 160–167.
Nat. Med. 20 (2) (2014) 159–166. [175] M. Mandal, R. Kumar, Bcl-2 expression regulates sodium butyrate-induced apop-
[154] M. Kinoshita, Y. Suzuki, Y. Saito, Butyrate reduces colonic paracellular perme- tosis in human MCF-7 breast cancer cells, Cell Growth Differ. 7 (3) (1996)
ability by enhancing PPARgamma activation, Biochem. Biophys. Res. Commun. 311–318.
293 (2) (2002) 827–831. [176] S. Prasanna Kumar, G. Thippeswamy, M.L. Sheela, B.T. Prabhakar, B.P. Salimath,
[155] S. Fukuda, H. Toh, K. Hase, K. Oshima, Y. Nakanishi, K. Yoshimura, T. Tobe, Butyrate-induced phosphatase regulates VEGF and angiogenesis via Sp1, Arch.
J.M. Clarke, D.L. Topping, T. Suzuki, T.D. Taylor, K. Itoh, J. Kikuchi, H. Morita, Biochem. Biophys. 478 (1) (2008) 85–95.
M. Hattori, H. Ohno, Bifidobacteria can protect from enteropathogenic infection [177] S.R. Pulliam, S.T. Pellom Jr., A. Shanker, S.E. Adunyah, Butyrate regulates the
through production of acetate, Nature 469 (7331) (2011) 543–547. expression of inflammatory and chemotactic cytokines in human acute leukemic
[156] C.J. Kelly, L. Zheng, E.L. Campbell, B. Saeedi, C.C. Scholz, A.J. Bayless, cells during apoptosis, Cytokine 84 (2016) 74–87.
K.E. Wilson, L.E. Glover, D.J. Kominsky, A. Magnuson, T.L. Weir, S.F. Ehrentraut, [178] E.C. Aguilar, A.J. Leonel, L.G. Teixeira, A.R. Silva, J.F. Silva, J.M. Pelaez,
C. Pickel, K.A. Kuhn, J.M. Lanis, V. Nguyen, C.T. Taylor, S.P. Colgan, Crosstalk L.S. Capettini, V.S. Lemos, R.A. Santos, J.I. Alvarez-Leite, Butyrate impairs
between microbiota-derived short-chain fatty acids and intestinal epithelial HIF atherogenesis by reducing plaque inflammation and vulnerability and decreasing
augments tissue barrier function, Cell Host Microbe 17 (5) (2015) 662–671. NFkappaB activation, Nutr. Metab. Cardiovasc. Dis. 24 (6) (2014) 606–613.
[157] T. Hoverstad, T. Midtvedt, Short-chain fatty acids in germfree mice and rats, J. [179] R. Simeoli, G. Mattace Raso, C. Pirozzi, A. Lama, A. Santoro, R. Russo, T. Montero-
Nutr. 116 (9) (1986) 1772–1776. Melendez, R. Berni Canani, A. Calignano, M. Perretti, R. Meli, An orally ad-
[158] K.M. Maslowski, A.T. Vieira, A. Ng, J. Kranich, F. Sierro, D. Yu, H.C. Schilter, ministered butyrate-releasing derivative reduces neutrophil recruitment and in-
M.S. Rolph, F. Mackay, D. Artis, R.J. Xavier, M.M. Teixeira, C.R. Mackay, flammation in dextran sulphate sodium-induced murine colitis, Br. J. Pharmacol.
Regulation of inflammatory responses by gut microbiota and chemoattractant (2016).
receptor GPR43, Nature 461 (7268) (2009) 1282–1286. [180] B.Z. Qian, J. Li, H. Zhang, T. Kitamura, J. Zhang, L.R. Campion, E.A. Kaiser,
[159] C.M. van der Beek, J.G. Bloemen, M.A. van den Broek, K. Lenaerts, K. Venema, L.A. Snyder, J.W. Pollard, CCL2 recruits inflammatory monocytes to facilitate
W.A. Buurman, C.H. Dejong, Hepatic uptake of rectally administered butyrate breast-tumour metastasis, Nature 475 (7355) (2011) 222–225.
prevents an increase in systemic butyrate concentrations in humans, J. Nutr. 145 [181] R. Pathania, S. Ramachandran, G. Mariappan, P. Thakur, H. Shi, J.-H. Choi,
(9) (2015) 2019–2024. S. Manicassamy, R. Kolhe, P.D. Prasad, S. Sharma, Combined inhibition of DNMT
[160] A.C. Nilsson, E.M. Ostman, K.E. Knudsen, J.J. Holst, I.M. Bjorck, A cereal-based and HDAC blocks the tumorigenicity of cancer stem-like cells and attenuates
evening meal rich in indigestible carbohydrates increases plasma butyrate the next mammary tumor growth, Cancer Res. 76 (11) (2016) 3224–3235.
morning, J. Nutr. 140 (11) (2010) 1932–1936. [182] H. Bernstein, C.M. Payne, C. Bernstein, J. Schneider, S.E. Beard, C.L. Crowley,
[161] E. Boets, S.V. Gomand, L. Deroover, T. Preston, K. Vermeulen, V. De Preter, Activation of the promoters of genes associated with DNA damage, oxidative stress
H.M. Hamer, G. Van den Mooter, L. De Vuyst, C.M. Courtin, P. Annaert, ER stress and protein malfolding by the bile salt, deoxycholate, Toxicol. Lett. 108
J.A. Delcour, K.A. Verbeke, Systemic availability and metabolism of colonic-de- (1) (1999) 37–46.
rived short-chain fatty acids in healthy subjects: a stable isotope study, J. Physiol. [183] P.A. Craven, J. Pfanstiel, F.R. DeRubertis, Role of reactive oxygen in bile salt
595 (2) (2017) 541–555. stimulation of colonic epithelial proliferation, J. Clin. Invest. 77 (3) (1986)
[162] N. Singh, A. Gurav, S. Sivaprakasam, E. Brady, R. Padia, H. Shi, M. Thangaraju, 850–859.
P.D. Prasad, S. Manicassamy, D.H. Munn, J.R. Lee, S. Offermanns, V. Ganapathy, [184] M.M. Huycke, D.R. Moore, In vivo production of hydroxyl radical by Enterococcus
Activation of Gpr109a, receptor for niacin and the commensal metabolite buty- faecalis colonizing the intestinal tract using aromatic hydroxylation, Free Radic.
rate, suppresses colonic inflammation and carcinogenesis, Immunity 40 (1) (2014) Biol. Med. 33 (6) (2002) 818–826.
128–139. [185] X. Wang, M.M. Huycke, Extracellular superoxide production by Enterococcus
[163] P.M. Smith, M.R. Howitt, N. Panikov, M. Michaud, C.A. Gallini, Y.M. Bohlooly, faecalis promotes chromosomal instability in mammalian cells, Gastroenterology
J.N. Glickman, W.S. Garrett, The microbial metabolites, short-chain fatty acids 132 (2) (2007) 551–561.
regulate colonic Treg cell homeostasis, Science (New York, N.Y.) 341 (6145) [186] J.P. Nougayrede, S. Homburg, F. Taieb, M. Boury, E. Brzuszkiewicz, G. Gottschalk,
(2013) 569–573. C. Buchrieser, J. Hacker, U. Dobrindt, E. Oswald, Escherichia coli induces DNA
[164] Y. Furusawa, Y. Obata, S. Fukuda, T.A. Endo, G. Nakato, D. Takahashi, double-strand breaks in eukaryotic cells, Science (New York, N.Y.) 313 (5788)
Y. Nakanishi, C. Uetake, K. Kato, T. Kato, M. Takahashi, N.N. Fukuda, (2006) 848–851.
S. Murakami, E. Miyauchi, S. Hino, K. Atarashi, S. Onawa, Y. Fujimura, T. Lockett, [187] G. Cuevas-Ramos, C.R. Petit, I. Marcq, M. Boury, E. Oswald, J.P. Nougayrede,
J.M. Clarke, D.L. Topping, M. Tomita, S. Hori, O. Ohara, T. Morita, H. Koseki, Escherichia coli induces DNA damage in vivo and triggers genomic instability in
J. Kikuchi, K. Honda, K. Hase, H. Ohno, Commensal microbe-derived butyrate mammalian cells, Proc. Natl. Acad. Sci. U. S. A. 107 (25) (2010) 11537–11542.
induces the differentiation of colonic regulatory T cells, Nature 504 (7480) (2013) [188] B.G. Feuerstein, N. Pattabiraman, L.J. Marton, Molecular mechanics of the inter-
446–450. actions of spermine with DNA: DNA bending as a result of ligand binding, Nucleic
[165] N. Arpaia, C. Campbell, X. Fan, S. Dikiy, J. van der Veeken, P. deRoos, H. Liu, Acids Res. 18 (5) (1990) 1271–1282.
J.R. Cross, K. Pfeffer, J. Coffer, A.Y. Rudensky, Metabolites produced by com- [189] S. Fujisawa, Y. Kadoma, Kinetic evaluation of polyamines as radical scavengers,
mensal bacteria promote peripheral regulatory T-cell generation, Nature 504 Anticancer Res. 25 (2a) (2005) 965–969.
(7480) (2013) 451–455. [190] K. Zahedi, J.J. Bissler, Z. Wang, A. Josyula, L. Lu, P. Diegelman, N. Kisiel,
[166] M.A. Zimmerman, N. Singh, P.M. Martin, M. Thangaraju, V. Ganapathy, C.W. Porter, M. Soleimani, Spermidine/spermine N1-acetyltransferase over-
J.L. Waller, H. Shi, K.D. Robertson, D.H. Munn, K. Liu, Butyrate suppresses colonic expression in kidney epithelial cells disrupts polyamine homeostasis, leads to DNA
inflammation through HDAC1-dependent Fas upregulation and Fas-mediated damage, and causes G2 arrest, Am. J. Physiol. 292 (3) (2007) C1204–15.
apoptosis of T cells, Am. J. Physiol. 302 (12) (2012) G1405–15. [191] V.M. Johansson, S.M. Oredsson, K. Alm, Polyamine depletion with two different
[167] A.N. Thorburn, C.I. McKenzie, S. Shen, D. Stanley, L. Macia, L.J. Mason, polyamine analogues causes DNA damage in human breast cancer cell lines, DNA
L.K. Roberts, C.H. Wong, R. Shim, R. Robert, N. Chevalier, J.K. Tan, E. Marino, Cell Biol. 27 (9) (2008) 511–516.
R.J. Moore, L. Wong, M.J. McConville, D.L. Tull, L.G. Wood, V.E. Murphy, [192] T. Douki, Y. Bretonniere, J. Cadet, Protection against radiation-induced degrada-
J. Mattes, P.G. Gibson, C.R. Mackay, Evidence that asthma is a developmental tion of DNA bases by polyamines, Radiat. Res. 153 (1) (2000) 29–35.
origin disease influenced by maternal diet and bacterial metabolites, Nat. [193] B.G. Durie, S.E. Salmon, D.H. Russell, Polyamines as markers of response and
Commun. 6 (2015) 7320. disease activity in cancer chemotherapy, Cancer Res. 37 (1) (1977) 214–221.
[168] M.A. Cox, J. Jackson, M. Stanton, A. Rojas-Triana, L. Bober, M. Laverty, X. Yang, [194] D.H. Russell, C.C. Levy, S.C. Schimpff, I.A. Hawk, Urinary polyamines in cancer
F. Zhu, J. Liu, S. Wang, F. Monsma, G. Vassileva, M. Maguire, E. Gustafson, patients, Cancer Res. 31 (11) (1971) 1555–1558.
M. Bayne, C.C. Chou, D. Lundell, C.H. Jenh, Short-chain fatty acids act as anti- [195] A.E. Pegg, Polyamine metabolism and its importance in neoplastic growth and a
inflammatory mediators by regulating prostaglandin E(2) and cytokines, World J. target for chemotherapy, Cancer Res. 48 (4) (1988) 759–774.
Gastroenterol. 15 (44) (2009) 5549–5557. [196] L. Chamaillard, V. Catros-Quemener, J.G. Delcros, J.Y. Bansard, R. Havouis,
[169] M. Kassayova, N. Bobrov, L. Strojny, T. Kiskova, J. Mikes, V. Demeckova, D. Desury, A. Commeurec, N. Genetet, J.P. Moulinoux, Polyamine deprivation
P. Orendas, B. Bojkova, M. Pec, P. Kubatka, A. Bomba, Preventive effects of pro- prevents the development of tumour-induced immune suppression, Br. J. Cancer
biotic bacteria Lactobacillus plantarum and dietary fiber in chemically-induced 76 (3) (1997) 365–370.
mammary carcinogenesis, Anticancer Res. 34 (9) (2014) 4969–4975. [197] C.S. Hayes, A.C. Shicora, M.P. Keough, A.E. Snook, M.R. Burns, S.K. Gilmour,
[170] B. Bonnotte, N. Favre, S. Reveneau, O. Micheau, N. Droin, C. Garrido, A. Fontana, Polyamine-blocking therapy reverses immunosuppression in the tumor micro-
B. Chauffert, E. Solary, F. Martin, Cancer cell sensitization to fas-mediated apop- environment, Cancer Immunol. Res. 2 (3) (2014) 274–285.
tosis by sodium butyrate, Cell Death Differ. 5 (6) (1998) 480–487. [198] C.H. Johnson, C.M. Dejea, D. Edler, L.T. Hoang, A.F. Santidrian, B.H. Felding,
[171] K.Y. Fung, G.V. Brierley, S. Henderson, P. Hoffmann, S.R. McColl, T. Lockett, J. Ivanisevic, K. Cho, E.C. Wick, E.M. Hechenbleikner, W. Uritboonthai, L. Goetz,
R. Head, L. Cosgrove, Butyrate-induced apoptosis in HCT116 colorectal cancer R.A. Casero Jr., D.M. Pardoll, J.R. White, G.J. Patti, C.L. Sears, G. Siuzdak,
cells includes induction of a cell stress response, J. Proteome Res. 10 (4) (2011) Metabolism links bacterial biofilms and colon carcinogenesis, Cell Metab. 21 (6)
1860–1869. (2015) 891–897.
[172] B.F. Hinnebusch, S. Meng, J.T. Wu, S.Y. Archer, R.A. Hodin, The effects of short- [199] K. Soda, The mechanisms by which polyamines accelerate tumor spread, J. Exp.

72
C.M. Buchta Rosean, M.R. Rutkowski Seminars in Immunology 32 (2017) 62–73

Clin. Cancer Res. 30 (2011). [211] G.M. Nava, F. Carbonero, J.A. Croix, E. Greenberg, H.R. Gaskins, Abundance and
[200] J. Russo, S.V. Fernandez, P.A. Russo, R. Fernbaugh, F.S. Sheriff, H.M. Lareef, diversity of mucosa-associated hydrogenotrophic microbes in the healthy human
J. Garber, I.H. Russo, 17-Beta-estradiol induces transformation and tumorigenesis colon, ISME J. 6 (1) (2012) 57–70.
in human breast epithelial cells, FASEB J. 20 (10) (2006) 1622–1634. [212] J. Schneider, V.F. Wendisch, Biotechnological production of polyamines by bac-
[201] P.A. Furth, M.C. Cabrera, E.S. Diaz-Cruz, S. Millman, R.E. Nakles, Assessing es- teria: recent achievements and future perspectives, Appl. Microbiol. Biotechnol.
trogen signaling aberrations in breast cancer risk using genetically engineered 91 (1) (2011) 17–30.
mouse models, Ann. N. Y. Acad. Sci. 1229 (2011) 147–155. [213] J. Nakamura, Y. Kubota, M. Miyaoka, T. Saitoh, F. Mizuno, Y. Benno, Comparison
[202] M. Dabek, S.I. McCrae, V.J. Stevens, S.H. Duncan, P. Louis, Distribution of beta- of four microbial enzymes in Clostridia and Bacteroides isolated from human
glucosidase and beta-glucuronidase activity and of beta-glucuronidase gene gus in feces, Microbiol. Immunol. 46 (7) (2002) 487–490.
human colonic bacteria, FEMS Microbiol. Ecol. 66 (3) (2008) 487–495. [214] A.J. McBain, G.T. Macfarlane, Ecological and physiological studies on large in-
[203] B.J. Fuhrman, H.S. Feigelson, R. Flores, M.H. Gail, X. Xu, J. Ravel, J.J. Goedert, testinal bacteria in relation to production of hydrolytic and reductive enzymes
Associations of the fecal microbiome with urinary estrogens and estrogen meta- involved in formation of genotoxic metabolites, J. Med. Microbiol. 47 (5) (1998)
bolites in postmenopausal women, J. Clin. Endocrinol. Metab. 99 (12) (2014) 407–416.
4632–4640. [215] S. Wu, K.J. Rhee, M. Zhang, A. Franco, C.L. Sears, Bacteroides fragilis toxin sti-
[204] R. Flores, J. Shi, B. Fuhrman, X. Xu, T.D. Veenstra, M.H. Gail, P. Gajer, J. Ravel, mulates intestinal epithelial cell shedding and gamma-secretase-dependent E-
J.J. Goedert, Fecal microbial determinants of fecal and systemic estrogens and cadherin cleavage, J. Cell Sci. 120 (Pt 11) (2007) 1944–1952.
estrogen metabolites: a cross-sectional study, J. Transl. Med. 10 (2012) 253. [216] R.J. Obiso Jr., A.O. Azghani, T.D. Wilkins, The Bacteroides fragilis toxin fragilysin
[205] H. Adlercreutz, M.O. Pulkkinen, E.K. Hamalainen, J.T. Korpela, Studies on the role disrupts the paracellular barrier of epithelial cells, Infect. Immunol. 65 (4) (1997)
of intestinal bacteria in metabolism of synthetic and natural steroid hormones, J. 1431–1439.
Steroid Biochem. 20 (1) (1984) 217–229. [217] R.G. Gaudet, A. Sintsova, C.M. Buckwalter, N. Leung, A. Cochrane, J. Li, A.D. Cox,
[206] N. Svoronos, A. Perales-Puchalt, M.J. Allegrezza, M.R. Rutkowski, K.K. Payne, J. Moffat, S.D. Gray-Owen, Cytosolic detection of the bacterial metabolite HBP
A.J. Tesone, J.M. Nguyen, T.J. Curiel, M.G. Cadungog, S. Singhal, E.B. Eruslanov, activates TIFA-dependent innate immunity, Science (New York, N. Y.) 348 (6240)
P. Zhang, J. Tchou, R. Zhang, J.R. Conejo-Garcia, Tumor cell-independent es- (2015) 1251–1255.
trogen signaling drives disease progression through mobilization of myeloid-de- [218] F. Fallarino, U. Grohmann, S. You, B.C. McGrath, D.R. Cavener, C. Vacca,
rived suppressor cells, Cancer Discov. 7 (1) (2017) 72–85. C. Orabona, R. Bianchi, M.L. Belladonna, C. Volpi, M.C. Fioretti, P. Puccetti,
[207] A.M. O'Hara, F. Shanahan, The gut flora as a forgotten organ, EMBO Rep. 7 (7) Tryptophan catabolism generates autoimmune-preventive regulatory T cells,
(2006) 688–693. Transpl. Immunol. 17 (1) (2006) 58–60.
[208] S. Roy, G. Trinchieri, Microbiota: a key orchestrator of cancer therapy, Nat. Rev. [219] F. Fallarino, U. Grohmann, S. You, B.C. McGrath, D.R. Cavener, C. Vacca,
Cancer (2017). C. Orabona, R. Bianchi, M.L. Belladonna, C. Volpi, P. Santamaria, M.C. Fioretti,
[209] S. Macfarlane, G.T. Macfarlane, Regulation of short-chain fatty acid production, P. Puccetti, The combined effects of tryptophan starvation and tryptophan cata-
Proc. Nutr. Soc. 62 (1) (2003) 67–72. bolites down-regulate T cell receptor zeta-chain and induce a regulatory pheno-
[210] J.M. Ridlon, D.J. Kang, P.B. Hylemon, Bile salt biotransformations by human in- type in naive T cells, J. Immunol. (Baltim. Md.: 1950) 176 (11) (2006) 6752–6761.
testinal bacteria, J. Lipid Res. 47 (2) (2006) 241–259.

73

Вам также может понравиться