Nothing Special   »   [go: up one dir, main page]

Gut Microbiota and Acute Kidney Injury - Immunological Crosstalk Link

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

International Urology and Nephrology (2024) 56:1345–1358

https://doi.org/10.1007/s11255-023-03760-5

NEPHROLOGY - REVIEW

Gut microbiota and acute kidney injury: immunological crosstalk link


Asmaa Ali1,2,3 · Liang Wu4 · Sameh Samir Ali5,6

Received: 14 April 2023 / Accepted: 14 August 2023 / Published online: 26 September 2023
© The Author(s), under exclusive licence to Springer Nature B.V. 2023

Abstract
The gut microbiota, often called the "forgotten organ," plays a crucial role in bidirectional communication with the host
for optimal physiological function. This communication helps regulate the host’s immunity and metabolism positively and
negatively. Many factors influence microbiota homeostasis and subsequently lead to an immune system imbalance. The
correlation between an unbalanced immune system and acute diseases such as acute kidney injury is not fully understood,
and the role of gut microbiota in disease pathogenesis is still yet uncovered. This review summarizes our understanding of
gut microbiota, focusing on the interactions between the host’s immune system and the microbiome and their impact on
acute kidney injury.

Keyword Gut microbiota · Innate and adaptive immune response · Acute kidney injury

Introduction diet, and host-related aspects such as genetics, age, gender,


immune system, and pathological disease [4, 5].
The gut microbiota plays a vital role in human health; it The active gut microbial community interacts with the
includes more than 100 trillion microbes to form a complex host and does many valuable functions. Hence, it is involved
ecosystem [1, 2]. Its stability and function are determined by in energy production and storage through many particular
the cooperation between the host and inhabited microbes [3]. metabolic pathways and enzymes, so these properties press
The composition and structure of the gut microbiota com- on nominating the bacteria as human symbionts [6]. Addi-
munity changed in response to related environmental factors, tionally, the gut microbiota influences the normal develop-
ment of the gut through its impact on the proliferation and
apoptosis of host epithelial cells. Even though the innate
interaction between host cell and microbiota are still undis-
* Asmaa Ali covered, a major pathway via short-chain fatty acid (SCFA)
asmaa.ali81@yahoo.com
has a significant anti-inflammatory role. Moreover, short-
* Liang Wu chain fatty acid (SCFA), which results from the fermentation
wl_ujs@163.com
of indigestible polysaccharides (fibers), such as butyrate,
Sameh Samir Ali acetate, and propionate, supports intestinal homeostasis
samh@ujs.edu.cn; samh_samir@science.tanta.edu.eg
in the normal colon by aiding intestinal repair through the
1
Department of Laboratory Medicine, School of Medicine, encouragement of proliferation and differentiation of the
Jiangsu University, Zhenjiang 212013, China cells [7–9].
2
Department of Pulmonary Medicine, Abbassia Chest Among the beneficial role of gut microbiota is stimulating
Hospital, MOH, Cairo, Egypt the development of the specific and non-specific immune
3
Department of Respiratory Allergy, A Al-Rashed Allergy system; hence, the predominant component of the micro-
Center, Ministry of Health, Kuwait, Kuwait biota is anaerobes organisms which prevent the process of
4
Yizheng Hospital, Nanjing Drum Tower Hospital Group, translocation of aerobic/facultatively anaerobic bacteria and
Yizheng 210008, China the consecutive systemic infections in immune-deficient
5
School of the Environment and Safety Engineering, Biofuels individuals. Significantly, some GIT microbiota, such as
Institute, Jiangsu University, Zhenjiang 212013, China Escherichia coli and Bacteroides fragilis, are involved in
6
Botany Department, Faculty of Science, Tanta University, synthesizing vitamins such as B1, B2, B5, B6, B12, K, folic
Tanta 31527, Egypt

13
Vol.:(0123456789)
1346 International Urology and Nephrology (2024) 56:1345–1358

acid, and biotin. The GIT microbiota, such as B. fragilis and similar protein or metabolite profiles [19, 20]. This informa-
Fusobacterium species, can degrade xenobiotics and ster- tion is essential and may be the key to understanding the
ols and perform biliary acids deconjugation [10]. All these shape of microbial communities in different diseases and
values occur when the eubiosis state is present; however, opening the door for emerging novel therapeutic approaches.
alteration of microbiota homeostasis, which is recognized as
dysbiosis, will drive to infectious and non-infectious disease, Development of GI microbiota
each characterized by specific microbiota signature that trig-
gered its pathological mechanisms [6]. A few studies have been conducted on gut microbiota devel-
This review emphasizes the different immunological opment throughout a person’s lifetime, starting from birth
mechanisms by which the gut microbiota modulates the [21, 22]. Changes in microbiota composition can be linked
usual health status and highlights some of the immune- to various factors such as mode of birth, illness, antibiotic
pathological pathways involved in acute kidney injury. use, and diet changes [22, 23]. Infants delivered by cesarean
section have been found to have depleted levels of the Bac-
Structure and composition of human gut microbiota teroides genus and increased levels of facultative anaerobes
such as Clostridium species [24, 25]. Conversely, infants
The interface of human GIT is the largest one between the delivered vaginally have a high number of lactobacilli in
host and environmental factors; hence it is estimated to be their gut microbiota due to direct transfer from the mother’s
250–400 ­m2, and its primary role is the protection of gut vaginal flora [26, 27].
integrity [11]. A collection of organisms inhabit the gut, During the early stages of development, gut microbiota
termed "gut microbiota," that interact and form a beneficial diversity is low and dominated by two main phyla: Actino-
relationship with the host [12, 13]. The estimated number of bacteria and Proteobacteria [22, 28]. However, gut micro-
gut microbiota reached 1014, while its amount of genomic biota diversity increases within the first year of life and
structure exceeds the amount of human one by 100 times converges towards an adult-like pattern by 2.5 years [29].
[14, 15]. Despite this, gut microbiota composition and function can
As a result of the improvement of culture-independent still be subject to changes in adulthood [30]. Older individu-
approaches, the ability to study the scope of gut microbiota als have been found to have a shift in gut microbiota diver-
has significantly progressed [11]. The popular technique sity towards an increased abundance of the Bacteroidetes
targets the bacterial 16S ribosomal RNA (rRNA) gene, phylum and Clostridium cluster IV, compared to younger
which is present in all archaea and bacteria and contains individuals where cluster XIVa is more common [31]. While
nine highly variable regions (V1–V9) that allow species to one study found no difference in gut microbiota composi-
be easily differentiated. Earlier techniques concentrated on tion between both age groups [32], aging has been linked
sequencing the entire 16S rRNA gene and then on analyzing to changes in short-chain fatty acid (SCFA) and amylolysis
the gene’s short sub-regions but in more depth. However, the production by gut microbiota, as well as an increase in pro-
bias and insensitivity of earlier methods and errors of read- teolytic products [33], which highlights the immunomodula-
ing in the subsequent one gave more data about microbiota tory effect of SCFA on gut inflammation and aging [34–37].
composition [16, 17]. Another more advanced technology
provides a reliable estimate of microbiota diversity; whole Biogeography of gut microbiota
genome shotgun metagenomics, characterized by high reso-
lution and sensitivity in reading [18, 19]. The composition and density of microbiota respect the
Previous research showed that 2172 species were isolated physiological properties of the gut region [38]; the below
from humans and categorized into 12 phyla. The majority diagram (Fig. 1) highlights the idea that, hence, changes in
of these species, 93.5%, belong to Proteobacteria, Firmi- chemical, nutritional, and immunological gradients along the
cutes, Actinobacteria, and Bacteroidetes. Only three of the gut affect the composition and diversity of microbiota [39].
12 phyla contained only one species isolated from humans, Moreover, the variability in composition within the same
with Akkermansia muciniphila being the sole representa- individuals was reported [40, 41]. Hence, the abundance of
tive of the Verrucomicrobia phyla [18]. Approximately 386 Bacteroidetes is higher in fecal/luminal samples than in the
of the identified human species are anaerobic and located mucosa. Contrary, Firmicutes, specifically Clostridium clus-
in mucosal areas like the oral cavity and the digestive tract ter XIVa, are augmented in the mucus layer compared to the
[19]. lumen [40, 42].
So many factors, such as diet and hosts genetic aspects, Recently, individual microbiota could be classified into
controlled the gut microbiota composition; therefore, the community types based on background reference [43]. Three
country-specific configuration signatures. So, microbiotas enterotypes were identified as a result of a multi-nationality
that differ in structure may share some functional lay-off and study and depend on a variation of the level of one of three

13
International Urology and Nephrology (2024) 56:1345–1358 1347

However, the elderly and infants have less diverse and less
stable gut microbiomes than healthy adults [48].

Role of healthy gut microbiota

As a result of the unique distribution of gut microbiota of


every individual, the definite composition of healthy gut
microbiota has yet to be identified [49]. The variability of
microbial diversity and composition per individual has been
reported as a consequence of alteration in microbial gene
structure (microbiome) and changes in environmental factors
and host genetics [50–52].
The core gut microbiome was stable during healthy adult-
hood [48]. Production of SCFAs is a complex process, which
exhibits through fermentation and synthesis of specific lip-
polysaccharides (LPS) in addition to some essential amino
acids and vitamins by mature, healthy gut microbiota [51,
53, 54] (Fig. 2). On the other hand, qualitative and quan-
Fig. 1  Microbiota composition and density across the gut region titative alteration of microbial diversity has been reported
in different diseases, such as obesity and immune-related
inflammatory disorders [55–58].
genera: Bacteroides (enterotype 1), Prevotella (enterotype
2), and Ruminococcus (enterotype 3) [44, 45]. However, Role of microbiota and its metabolites in health
evidence nearby the existence and formation of these ente- and disease
rotypes is controversial [46].
The relationship between host and microbiota is mutual;
hence, the microbiota modulates gut physiology and extra-
Factors influencing the composition and diversity intestinal function, in addition to regulating energy metabo-
of microbiota lism. Microbiota depends on energy from the host to main-
tain its growth. In return, it supplies the host with energy
The composition of the gut microbiome changes along through the liberation of different enzymes and metabolites
with the age of the host; Table 1 summarizes these factors; such as SCFAs, bile acids (BAs), proteas B (CLpB), and
Microbial richness reaches its optimal level in adulthood lipopolysaccharide (LPS) [59]. Moreover, due to substantial
[47]. While there may be some variations between individu- genomic content and metabolic complements, the gut micro-
als, the gut microbiome of healthy adults is generally stable. biota provides the host with valuable benefits. Some help

Table 1  Factors affecting gut Factors Microbial composition Age group


microbiota composition all
through host age [47] Gestational age Actinobacteria Infant (up to 3 years)
Mode of delivery (Vaginal Vs caesarian) Proteobacteria, Firmicutes
Feeding type (breast Vs formula) Bacteroidetes
Malnutrition
Infant hospitalization
Antibiotic treatment
Age of solid food introduction
Diet Firmicutes, Bacteroidetes Actino- Adulthood
Changes in life style bacteria, Proteobacteria
Travelling
Illness
Antibiotic and other drug uses
Hormonal changes
Comorbid associated illness Firmicutes, Actinobacteria Bacte- Elderly (above 65)
Several medication use roidetes, Proteobacteria
Infection and inflammation susceptibility
Changes in diet and daily habits

13
1348 International Urology and Nephrology (2024) 56:1345–1358

maintain the integrity of the mucosal barrier, while others


protect against pathogens. Moreover, the interaction between
commensal microbiota and the mucosal immune system is
vital for proper immune function [11].
The proportions of propionate, butyrate, and acetate, the
three predominant SCFAs, are 1:1:3 [60]. Gut epithelial cells
rapidly absorb these fatty acids to promote multiple func-
tions [61–68] (Fig. 3).
Other microbial metabolites rather than SCFAs have
been reported to influence epithelial proliferation, keeping
the intestinal barrier functioning and sharing in the immune
system response [69]. As well gut microbiota are responsible
for some essential vitamin synthesis. Hence, a lactic acid
bacterium was the key to vitamin B12 production, which
cannot be formed by any other sources [70, 71]. Moreo-
ver, folate production was produced by Bifidobacteria and
is essential for host metabolic process as DNA synthesis
and repair [72]. Gut microbiota also shares in the synthe-
Fig. 2  Healthy microbiota response to diet rich in fibers. Foods rich sis of Vitamin K, biotin, nicotinic acid, pantothenic acid,
in fibers contain an adjustable amount of protein and fats under the riboflavin, thiamin, and pyridoxine [11]. Several studies dis-
digestion process by host enzymes, then transformed into indigestible cussed the impact of alteration of any of these products in
but fermentable polysaccharides. The microbiota of the large intestine host health, as in DM type 2 and obesity, in which changes
metabolizes and ferments it to produce a collection of compounds
and to stimulate a thick intestinal mucus layer and vital barrier func- in bile acid co-metabolisms, choline, niacin, and phenolic
tions. Microbial production of SCFAs supplies an additional energy compounds have been reported [11, 73, 74].
source for intestinal cells and causes a decrease in luminal pH. The Other studies reported the role of microbiota in cell
SCFAs acetate, butyrate, and propionate can bind to G protein-cou- renewal and wound healing through the promotion of mucin
pled receptor (GPCR)-41/43, which are expressed on entero-endo-
crine L cells, and subsequently encourage secretion of glucagon-like glycosylation and glycosyl transferase expression [75–82].
peptide 1 (GLP-1) and peptide YY (PYY) that contribute to improved
energy expenditure, abridged food intake and enhanced glucose Immunological interaction between host
metabolism and insulin secretion. Additionally, butyrate activates and microbiome
peroxisome proliferator-activated receptor-γ (PPARγ) and stimu-
lates β-oxidation and oxygen utilization in the gut, which upholds an
anaerobic environment in the gut lumen Microbiota and the host's immune system communicate
through different receptors on cells that present antigens in

Fig. 3  Major functions of


SCFAs and its predominant
types

13
International Urology and Nephrology (2024) 56:1345–1358 1349

the gut lining. This communication activates both the innate can also influence the types of T cells generated, such as
and adaptive immune response. In eubiosis, the immune sys- regulatory T cells, which play a crucial role in maintain-
tem responds to the gut bacteria [83], but the exact way they ing immune tolerance to commensal bacteria [173]; fur-
communicate is still an area of research. This review cov- thermore, the adaptive immune system can also influence
ers the latest research on how gut bacteria and the human the composition of the gut microbiome. For example, when
immune system interact, which could also have implications harmful bacteria are detected, the adaptive immune sys-
for acute kidney injury. tem can produce specific antibodies which can target and
eliminate those bacteria [174]. This interaction can lead to
Crosstalk between the innate and adaptive immune changes in the composition of the gut microbiome, as the
system and the microbiota immune response may positively or negatively impact the
presence of certain bacteria [175, 176].
The innate immune system and ecosystem work together Briefly, understanding reciprocal relationship of the gut
to maintain body health, and disruptions in this balance microbiota and the adaptive immune system is essential to
can lead to diseases. The specific mechanisms of this com- know how the gut microbiome can impact the health and
munication are still being studied and understood, but it is disease status of the host.
believed to play a crucial role in maintaining overall health.
In Table 2, we summarized the role of some immunological Microbiota and acute kidney injury
cells and their interaction with gut microbiota. Additionally,
the proper role and mechanism of interaction between the Acute kidney injury (AKI) is now used instead of acute renal
microbiome and adaptive immune system was enlightened failure (ARF), and reflects that minor decrease in kidney
too [84–164]. function even without organ failure could influence the clini-
The gut microbiota can interact with the innate immune cal consequence which increases the likelihood of morbidity
system in several ways; first, commensal bacteria can stimu- and mortality [177]. Multiple definitions have been created
late the production of cytokines, signaling molecules that for AKI to establish a consistent understanding; these defini-
help regulate the immune response [165]; second, the gut tions rely solely on serum creatinine and urine output levels.
microbiome can help to educate immune cells, lick T-cells They are mainly used to recognize AKI in epidemiological
and dendritic cells, about the presence of benign bacteria, and outcome based studies. However, it is yet to be con-
thus reducing the likelihood of an unnecessary immune firmed whether these definitions and staging systems are
response [166]; finally, the gut microbiota can also directly helpful in clinically evaluating and treating patients with
modulate the function of macrophages, which play an essen- AKI.
tial role in the innate immune response [167]. According to the Kidney Disease Improving Global
On the other hand, the innate immune system can also Guidelines (KDIGO), AKI should be classified into three
influence the gut microbiome. For example, when harmful stages based on the level of serum creatine and urine output.
bacteria are detected, the innate immune system can respond Additionally, it is suggested to assess patients for their risk
by releasing antimicrobial peptides, which can kill bacteria of AKI and monitor the serum creatinine level and urine
and shape gut microbiota composition [168]. In addition, the output regularly in those at risk and those with established
innate immune system can also modulate gut permeability, AKI [178]. From that instant, AKI is a sudden decrease in
affecting bacteria' access to the host's immune system [169]. kidney function that causes the fanfare of urea and other
The gut commensal and the innate immune system have nitrogenous waste products in the body. It also disrupts the
a symbiotic relationship, continuously interacting and influ- balance of extracellular volume and electrolytes [177].
encing each other. Disruptions in this relationship can lead Acute kidney injury (AKI) impacts various body systems,
to inflammation and an increased risk of disease, including resulting in fluid retention and impaired toxin excretion. This
conditions such as irritable bowel syndrome, inflammatory disease can lead to respiratory or circulatory failure, gas-
bowel disease, and other gastrointestinal disorders [170]. trointestinal dysfunction, neurocognitive defects, anemia,
The gut microbiota also interacts with the adaptive immunodeficiency, and systemic inflammation (Fig. 4);
immune system. The adaptive immune system, which kidney cell necrosis releases debris into the bloodstream,
includes T and B cells, can recognize and respond to spe- causing damage to the lungs, microvascular injury, throm-
cific pathogens and create immunological memory [171]. bosis, and in some cases, acute respiratory distress syndrome
The gut microbiome can influence this system in several [179].
ways; first, commensal bacteria can stimulate the develop- The immune and renal systems have a close relation-
ment of T and B cells in the gut-associated lymphoid tissue, ship, with the kidneys contributing to the immune sys-
which is vital for the maturation and education of the adap- tem's balance by removing toxins and monitoring blood-
tive immune system [172]; in addition, the gut microbiota borne proteins [180]. The kidneys support the stability of

13
1350 International Urology and Nephrology (2024) 56:1345–1358

Table 2  Innate and adaptive immune cells and structure and their interaction with gut microbiota
Immune cells and structure Role and mechanism of action Reference

Mucus barrier cells It protects the gut by two ways: physically blocks harmful microbes and influences immune cells [84, 86]
called dendritic cells to remain in an anti-inflammatory state; Secretory antibodies and antimi-
crobial peptides also help maintain the barrier's effectiveness
Tight junctions Upregulation of tight junctions and associated cytoskeletal proteins was promoted through micro- [87]
bial protein signals as indole, which endorses reinforcing the epithelial barrier
Paneth cells Cells in the small intestine mucosa were responsible for secreting large amounts of intestinal [88, 89]
Antimicrobial peptides (AMPs). Subsequently, AMPs interact with microbiota; it is important
in its shaping and configuration
Toll-like receptors (TLRs) One of the pattern recognition receptors (PRRs) that responsible for initiating microbial signals [90–95]
during infection. It is involved in defense mechanisms against pathogens and regulating com-
mensal abundance to maintain tissue integrity. Intestinal mucosa expresses different types
of TLR, each of which has cell type-specific and temporal patterns. For example, TLR5 was
defined in gut microbiota shaping
Polysaccharide A (PSA) Produced by commensal Bacteroides fragilis and are responsible for host immune system educa- [96–100]
tion. PSA is recognized by the TLR2/TLR1 heterodimer in collaboration with Dectin-1 Down-
stream to TLR1/TLR2 and Dectin-1 signaling, the phosphoinositide 3-kinase (PI3K) pathway
is activated, leading to the inactivation of glycogen synthase kinase 3β (GSK3β), which in turn
induces cAMP response element binding protein (CREB)-dependent expression of anti-inflam-
matory genes
Dectin-1 It has a recognition role, also it may regulate the immune response via Treg cell differentiation [101]
through modification of microbiota configuration
NOD-like receptors (NLRs) Pattern recognition receptors, it shapes the gut microbiota. NOD1) serves as an innate sensor [102–104]
assisting the generation of adaptive lymphoid tissues and maintaining intestinal homeostasis.
NOD2 prevents inflammation of the small intestine by restricting the growth of the commensal
Bacteroides vulgates. Stimulation of NOD2 by commensal bacteria promotes gut epithelial stem
cell survival and epithelial regeneration
MyD88 Reorganize microbial signals and signal pathways. It controls the epithelial expression of several [95, 105–107]
AMPs, including RegIIIγ, which limits the number of surface-associated gram-positive bacteria
and the activation of adaptive immunity. regulates T-cell differentiation, promotes microbiota
homeostasis through stimulation of IgA, and controls the expansion of Th17 cells by restricting
the growth of SFB
Inflammasomes NLRP6 inflammasome; it plays a crucial role in activating the inflammatory caspases, which [108–119]
leads to the maturation of IL-1β and IL-18 and the promotion of pyroptosis; regulate the
microbiome's composition and maintaining intestinal homeostasis; plays a role in regulating the
innate antiviral immunity in the intestine
NLRP3 inflammasome; anti-commensal IgG stimulates gut-resident FcγR-expressing mac-
rophages to produce the pro-inflammatory cytokine IL-1β through NLRP3- and reactive oxygen
species-dependent mechanisms
Proteus mirabilis stimulate monocytes to induce NLRP3-dependent IL-1β release and cause
intestinal inflammation upon injury
AIM2 inflammasome regulates intestinal homeostasis through the IL-18/IL-22/STAT3 pathway
Mammalian peptidogly- protect the host from colitis by promoting balanced microbiota configuration and by preventing [119]
can recognition proteins production of IFNγ by NK cells in response to injury
(PGRPs)
IPAF NOD‐LRR family of proteins. It recognizes intracellular flagellin and activates inflammasomes, [120]
stimulates caspase‐1, and promotes IL‐1β production in a TLR5‐independent manner in
Salmonella-infected macrophages
RIG-I-like receptors (RLRs) Protects against enteric bacterial infection by activating epithelial inflammasome signaling, and [121–123]
and OAS-like receptors thus promoting DC-driven Th1 and Th17 immunity
(OLRs)
Monocytes and macrophages A large microbiota-derived polysaccharide has been shown to induce an anti-inflammatory gene [124–127]
signature in murine intestinal macrophages
Butyrate can drive monocyte-to-macrophage differentiation through histone deacetylase 3
(HDAC3) inhibitions
Trimethylamine N-oxide (TMAO) can drive murine macrophage polarization in an NLRP3
inflammasome-dependent manner

13
International Urology and Nephrology (2024) 56:1345–1358 1351

Table 2  (continued)
Immune cells and structure Role and mechanism of action Reference

Innate lymphoid cells (ILCs) Rapid secretion of polarized cytokines and chemokines to combat infection and promote mucosal [128–136]
tissue repair
Type 3 ILCs mediate immune surveillance of microbiota configuration to facilitate early coloniza-
tion resistance through a transcriptional regulator ID2-dependent regulation of IL-22
NCR + ILC3 cells is essential for maintaining cecal homeostasis in mice during Citrobacter
rodentium infection
Ruminococcus gnavus is responsible for allergic disease in children, and induced infiltration of
the colon and lung parenchyma by eosinophils and mast cells in mice via a cascade implicating
type 2 ILCs, which suggest the role of ILCs in immune tolerance
B cells Maintain gut homeostasis by producing many secretory IgA antibodies that respond to com- [136–141]
mensal bacteria with T cell-dependent IgA playing a more significant role in shaping the gut
microbial community
Secretory IgA antibodies coat colitogenic bacteria, preventing disruptions to enteric homeostasis
and inflammation
Without B cells or IgA, the gut activates its inherent immune defense mechanisms through
interferon-inducible response pathways, leading to changes in microbiome composition and
impaired intestinal absorption and metabolism
T cells In response to intestinal bacteria, CD4 + T cells support homeostasis [142–164]
Microbiota is involved in Th17 differentiation in the intestine and skin, but oral barrier Th17 cell
development seems to be largely independent from microbial colonization
CD8 + T cells require priming by APCs and amplification by CD4 + T cell signaling, but their
memory potential is dependent on microbiota-derived short chain fatty acids
Microbiota-derived secondary bile acids regulate gut RORγ + regulatory T cell homeostasis
Follicular helper T (Tfh) cells assist B cells and maintain microbiota homeostasis, while Tfh cell
differentiation is impaired in GF mice and can be restored by TLR2 agonists
SFB-induced Tfh cell differentiation can boost autoantibody production and exacerbate arthritis
DCs shape immune responses, with a Syk kinase signaling pathway critical for microbiota
induced production of IL-17 and IL-22
NIK mediates mucosal DC function and altered enteric IgA secretion and microbiota homeostasis
Invariant natural killer T cells (iNKTs) are affected by the gut microbiota, with iNKTs from GF
animals showing decreased activation by antigens

Fig. 4  Pathogenesis of acute


kidney injury

13
1352 International Urology and Nephrology (2024) 56:1345–1358

the immune system by filtering out circulating cytokines in current review we spotlight on the mechanism by which
and bacterial toxins, such as lipopolysaccharide (LPS), and gut microbiota interacts with kidney immune cells, which
by constantly monitoring blood-borne proteins. Emerging is complex and unclear, however, short-chain fatty acids
evidence shows that renal tubular epithelial cells, various (SCFAs) appear to play important roles (Fig. 5) [184].
immune system components, and various molecular factors Gut microbiota produces SCFAs, which lead to ligation
contribute to the development of intrinsic AKI [181]. Vari- and activation of various G-protein coupled receptors such
ous factors contribute to the immunological development as GPR109a, free fatty acid receptor 2, free fatty acid recep-
of AKI, including pro-inflammatory damage-associated tor 3, and olfactory receptor-78, by which they modulate
molecular patterns (DAMPs), pathogen-associated molec- immune-cell function. SCFAs can further modulate the
ular patterns (PAMPs), toll-like receptors (TLRs), oxida- activity of histone acetyl-transferase and deacetylase and
tive stress, hypoxia-inducible factor (HIF), the complement the hypoxia-inducible factor. Exogenous administration of
system, adhesion molecules, cell death, resident renal den- SCFAs improved kidney function during experimental AKI
dritic cells (DCs), T and B lymphocytes, neutrophils, mac- and contrast-induced nephropathy [184].
rophages, natural killer T (NKT) cells, secreted cytokines, Dysregulation in gut microbiota and microbiome-associ-
and chemokines [182, 183]. If the immunological processes ated metabolites likely affect T-cell functions and antibody-
continue, it can lead to kidney fibrosis and chronic kidney mediated humoral immunity. Acetate treatment ameliorated
disease. sepsis-induced AKI by inhibiting NADPH-oxidase signaling
In the same line, the gut microbiota plays bidirectional in T cells, suggesting that SCFAs act through immune-cell
role in protecting inflammatory process and promoting regulation [185]. Furthermore, in vitro SCFA treatment was
kidney repair, as well as influencing the pro-inflammatory found to modulate the inflammatory process by decreasing
cascade through its impact in immune system. Therefore, dendritic-cell maturation and inhibiting CD4 and CD8 T-cell

Fig. 5  Microbiota-immune cell interaction in AKI. Short-chain fatty flammatory cytokines, such as IL-12 and IL-6, and skew the differ-
acids (SCFAs), produced from gut microbiota, such as acetate, pro- entiation of naive CD4 T cells and the maturation of B cells. Th17-
pionate, and butyrate, interact with multiple G-protein coupled recep- inducing bacteria may promote Th17 immunity via IL-17A/IL- 17F
tors on kidney epithelial cells. Similar interactions are likely involved induction, which may involve signaling mediated by the Toll-like
in immune cells that modulate their number, immune function, and receptor ligands. Additionally, IgA produced by plasma cells resid-
metabolism in the kidney. Normal gut microbiota promotes the anti- ing in the gut epithelium modulates response to colonization by spe-
inflammatory milieu by increasing T helper 2 (Th2) cells, regulatory cific commensal bacteria. SCFAs also regulate cytokine expression in
T (Treg) cells, and M2 macrophage populations that protect kidneys T cells and the generation of Treg cells through histone deacetylase
from AKI. However, AKI-induced dysbiosis and translocation of bac- inhibition. Therapeutic and supplemental use of pre-, pro-, and post-
terial products across the leaky intestine promote a proinflammatory biotics could help normalize bacterial composition in the gut and pro-
immune environment, such as increased Th1 cells, M1 macrophages, tect the kidneys from AKI. NKT, natural killer T cell
and activated dendritic cells (DCs). Activated DCs secrete proin-

13
International Urology and Nephrology (2024) 56:1345–1358 1353

proliferation, and could be a potential mechanism through 7. Lazar V, Ditu LM, Pircalabioru GG, Gheorghe I, Curutiu C,
which gut microbiota interact and modulate T-cell functions Holban AM, Picu A, Petcu L, Chifiriuc MC (2018) Aspects of
gut microbiota and immune system interactions in infectious dis-
in vivo [185]. eases, immunopathology, and cancer. Front Immunol 15(9):1830
Recent data demonstrated that diet could induce post- 8. Scheppach W (1994) Effects of short chain fatty acids on gut
translational modifications to the microbial proteome that, in morphology and function. Gut 35(1):S35–S38
turn, affect microbial metabolite production and, ultimately, 9. Heerdt BG, Houston MA, Augenlicht LH (1997) Short-chain
fatty acid-initiated cell cycle arrest and apoptosis of colonic
kidney function [186]. The lack of microbes in GF mice has epithelial cells is linked to mitochondrial function. Cell Growth
also been shown to affect the differentiation of regulatory T Diff-Publ Am Assoc Cancer Res. 8(5):523–532
cells due to the absence of SCFA production [143]. 10. Gérard P (2014) Metabolism of cholesterol and bile acids by the
gut microbiota. Pathogens 3(1):14–24
11. Thursby E, Juge N (2017) Introduction to the human gut micro-
biota. Biochem J 474(11):1823–1836
Conclusion and prospective 12. Neish AS (2009) Microbes in gastrointestinal health and disease.
Gastroenterol 136(1):65–80
13. Bäckhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI
The gut microbiota has a magnificent role in human health. (2005) Host-bacterial mutualism in the human intestine. Science
The microbiome can be changed through dietary modifica- 307(5717):1915–1920
tions and administration of pre-, pro-, and postbiotics, pro- 14. Gill SR, Pop M, DeBoy RT, Eckburg PB, Turnbaugh PJ, Sam-
viding unique opportunities to develop novel therapeutics for uel BS, Gordon JI, Relman DA, Fraser-Liggett CM, Nelson KE
(2006) Metagenomic analysis of the human distal gut microbi-
different disease as AKI. Moreover, the immune mechanisms ome. Science 312(5778):1355–1359
by which the potential SCFA-based therapeutic products can 15. Sender R, Fuchs S, Milo R (2016) Revised estimates for the
interfere with distant organ damage are still undiscovered. number of human and bacteria cells in the body. PLoS Biol
Therefore, it is a promising field for more research. 14(8):e1002533
16. Poretsky R, Rodriguez-R LM, Luo C, Tsementzi D, Konstan-
tinidis KT (2014) Strengths and limitations of 16S rRNA gene
amplicon sequencing in revealing temporal microbial community
Author contributions All authors contributed to the study concep- dynamics. PLoS ONE 9(4):e93827
tion and design. Data collection was performed by Asmaa Ali and Wu 17. Mizrahi-Man O, Davenport ER, Gilad Y (2013) Taxonomic
Liang. The first draft of the manuscript was written by Asmaa Ali. classification of bacterial 16S rRNA genes using short sequenc-
Asmaa Ali, Wu Liang and Sameh Samir Ali wrote the second draft, all ing reads: evaluation of effective study designs. PLoS ONE
authors commented on previous versions of the manuscript. All authors 8(1):e53608
read and approved the final manuscript. 18. Hugon P, Dufour JC, Colson P, Fournier PE, Sallah K, Raoult D
(2015) A comprehensive repertoire of prokaryotic species identi-
Funding The authors declare that no funds, grants, or other support fied in human beings. Lancet Infect Dis 15(10):1211–1219
were received during the preparation of this manuscript. 19. Li J, Jia H, Cai X, Zhong H, Feng Q, Sunagawa S, Arumugam M,
Kultima JR, Prifti E, Nielsen T, Juncker AS (2014) An integrated
Declarations catalog of reference genes in the human gut microbiome. Nat
Biotechnol 32(8):834–841
Conflict of interest The authors have no relevant financial or non-fi- 20. Moya A, Ferrer M (2016) Functional redundancy-induced stabil-
nancial interests to disclose. ity of gut microbiota subjected to disturbance. Trends Microbiol
24(5):402–413
21. Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, Versalovic
J (2014) The placenta harbors a unique microbiome. Sci Trans
References Med. 6(237):237–265
22. Rodríguez JM, Murphy K, Stanton C, Ross RP, Kober OI, Juge
1. Xu C, Zhu H, Qiu P (2019) Aging progression of human gut N, Avershina E, Rudi K, Narbad A, Jenmalm MC, Marchesi
microbiota. BMC Microbiol 19(1):1 JR (2015) The composition of the gut microbiota throughout
2. O’Hara AM, Shanahan F (2006) The gut flora as a forgotten life, with an emphasis on early life. Microb Ecol Health Dis
organ. EMBO Rep 7(7):688–693 26(1):26050
3. Shreiner AB, Kao JY, Young VB (2015) The gut microbiome in 23. Koenig JE, Spor A, Scalfone N, Fricker AD, Stombaugh J,
health and in disease. Curr Opin Gastroenterol 31(1):69 Knight R, Angenent LT, Ley RE (2011) Succession of micro-
4. Griffin NW, Ahern PP, Cheng J, Heath AC, Ilkayeva O, New- bial consortia in the developing infant gut microbiome. Proc Natl
gard CB, Fontana L, Gordon JI (2017) Prior dietary practices Acad Sci 108(1):4578–4585
and connections to a human gut microbial metacommunity alter 24. Jakobsson HE, Abrahamsson TR, Jenmalm MC, Harris K,
responses to diet interventions. Cell Host Microbe 21(1):84–96 Quince C, Jernberg C, Björkstén B, Engstrand L, Andersson AF
5. Lewis JD, Chen EZ, Baldassano RN, Otley AR, Griffiths AM, (2014) Decreased gut microbiota diversity, delayed Bacteroidetes
Lee D, Bittinger K, Bailey A, Friedman ES, Hoffmann C, Alben- colonisation and reduced Th1 responses in infants delivered by
berg L (2015) Inflammation, antibiotics, and diet as environmen- caesarean section. Gut 63(4):559–566
tal stressors of the gut microbiome in pediatric Crohn’s disease. 25. Salminen S, Gibson GR, McCartney AL, Isolauri E (2004) Influ-
Cell Host Microbe 18(4):489–500 ence of mode of delivery on gut microbiota composition in seven
6. Clemente JC, Ursell LK, Parfrey LW, Knight R (2012) The year old children. Gut 53(9):1388–1389
impact of the gut microbiota on human health: an integrative 26. Avershina E, Storrø O, Øien T, Johnsen R, Pope P, Rudi K (2014)
view. Cell 148(6):1258–1270 Major faecal microbiota shifts in composition and diversity with

13
1354 International Urology and Nephrology (2024) 56:1345–1358

age in a geographically restricted cohort of mothers and their 44. Falony G, Joossens M, Vieira-Silva S, Wang J, Darzi Y, Faust
children. FEMS Microbiol Ecol 87(1):280–290 K, Kurilshikov A, Bonder MJ, Valles-Colomer M, Vandeputte
27. Aagaard K, Riehle K, Ma J, Segata N, Mistretta TA, Coarfa D, Tito RY (2016) Population-level analysis of gut microbiome
C, Raza S, Rosenbaum S, Van den Veyver I, Milosavljevic A, variation. Science 352(6285):560–564
Gevers D (2012) A metagenomic approach to characterization 45. Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T,
of the vaginal microbiome signature in pregnancy. PLoS ONE Mende DR, Fernandes GR, Tap J, Bruls T, Batto JM, Bertalan
7(6):e36466 M (2011) Enterotypes of the human gut microbiome. Nature
28. Bäckhed F (2011) Programming of host metabolism by the gut 473(7346):174–180
microbiota. Ann Nutr Metab 58(2):44–52 46. Jeffery IB, Claesson MJ, Otoole PW, Shanahan F (2012) Cat-
29. Palmer C, Bik EM, DiGiulio DB, Relman DA, Brown PO (2007) egorization of the gut microbiota: enterotypes or gradients?
Development of the human infant intestinal microbiota. PLoS Nat Rev Microbiol 10(9):591–592
Biol 5(7):e177 47. Abubucker S, Segata N, Goll J, Schubert AM, Izard J, Cantarel
30. Dethlefsen L, Relman DA (2011) Incomplete recovery and BL, Rodriguez-Mueller B, Zucker J, Thiagarajan M, Henrissat
individualized responses of the human distal gut micro- B, White O (2012) Metabolic reconstruction for metagenomic
biota to repeated antibiotic perturbation. Proc Natl Acad Sci data and its application to the human microbiome. PLoS Com-
108(1):4554–4561 put Biol 8(6):e1002358
31. Claesson MJ, Cusack S, O’Sullivan O, Greene-Diniz R, de Weerd 48. D’Argenio V, Salvatore F (2015) The role of the gut microbi-
H, Flannery E, Marchesi JR, Falush D, Dinan T, Fitzgerald G, ome in the healthy adult status. Clin Chim Acta 7(451):97–102
Stanton C (2011) Composition, variability, and temporal stabil- 49. Korem T, Zeevi D, Suez J, Weinberger A, Avnit-Sagi T,
ity of the intestinal microbiota of the elderly. Proc Natl Acad Sci Pompan-Lotan M, Matot E, Jona G, Harmelin A, Cohen N,
108(1):4586–4591 Sirota-Madi A (2015) Growth dynamics of gut microbiota in
32. Xiao X, Wu ZC, Chou KC (2011) A multi-label classifier for health and disease inferred from single metagenomic samples.
predicting the subcellular localization of gram-negative bac- Science 349(6252):1101–1106
terial proteins with both single and multiple sites. PLoS ONE 50. Zeevi D, Korem T, Godneva A, Bar N, Kurilshikov A, Lotan-
6(6):e20592 Pompan M, Weinberger A, Fu J, Wijmenga C, Zhernakova A,
33. Woodmansey EJ, McMurdo ME, Macfarlane GT, Macfarlane S Segal E (2019) Structural variation in the gut microbiome asso-
(2004) Comparison of compositions and metabolic activities of ciates with host health. Nature 568(7750):43–48
fecal microbiotas in young adults and in antibiotic-treated and 51. Human Microbiome Project Consortium (2012) Structure,
non-antibiotic-treated elderly subjects. Appl Environ Microbiol function and diversity of the healthy human microbiome.
70(10):6113–6122 Nature 486(7402):207
34. Biagi E, Candela M, Turroni S, Garagnani P, Franceschi C, Bri- 52. Rothschild D, Weissbrod O, Barkan E, Kurilshikov A, Korem
gidi P (2013) Ageing and gut microbes: perspectives for health T, Zeevi D, Costea PI, Godneva A, Kalka IN, Bar N, Shilo S
maintenance and longevity. Pharmacol Res 69(1):11–20 (2018) Environment dominates over host genetics in shaping
35. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez- human gut microbiota. Nature 555(7695):210–215
Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, 53. Flint HJ, Scott KP, Louis P, Duncan SH (2012) The role of the
Anokhin AP, Heath AC (2012) Human gut microbiome viewed gut microbiota in nutrition and health. Nat Rev Gastroenterol
across age and geography. Nature 486(7402):222–227 Hepatol 9(10):577–589
36. Stewart CJ, Ajami NJ, O’Brien JL, Hutchinson DS, Smith 54. Shafquat A, Joice R, Simmons SL, Huttenhower C (2014)
DP, Wong MC, Ross MC, Lloyd RE, Doddapaneni H, Met- Functional and phylogenetic assembly of microbial com-
calf GA, Muzny D (2018) Temporal development of the gut munities in the human microbiome. Trends Microbiol
microbiome in early childhood from the TEDDY study. Nature 22(5):261–266
562(7728):583–588 55. Severance EG, Yolken RH, Eaton WW (2016) Autoimmune
37. Laursen MF, Bahl MI, Michaelsen KF, Licht TR (2017) First diseases, gastrointestinal disorders and the microbiome
foods and gut microbes. Front Microbiol 6(8):356 in schizophrenia: more than a gut feeling. Schizophr Res
38. Macpherson AJ, McCoy KD (2013) Stratification and compart- 176(1):23–35
mentalisation of immunoglobulin responses to commensal intes- 56. Schnabl B, Brenner DA (2014) Interactions between the
tinal microbes. Sem Immunolo 25:358–363 intestinal microbiome and liver diseases. Gastroenterology
39. Donaldson GP, Lee SM, Mazmanian SK (2016) Gut biogeogra- 146(6):1513–1524
phy of the bacterial microbiota. Nat Rev Microbiol 14(1):20–32 57. Clarke SF, Murphy EF, Nilaweera K, Ross PR, Shanahan F,
40. Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlef- O’Toole PW, Cotter PD (2012) The gut microbiota and its
sen L, Sargent M, Gill SR, Nelson KE, Relman DA (2005) relationship to diet and obesity: new insights. Gut microbes
Diversity of the human intestinal microbial flora. Science 3(3):186–202
308(5728):1635–1638 58. Cox AJ, West NP, Cripps AW (2015) Obesity, inflammation, and
41. Lavelle A, Lennon G, O’sullivan O, Docherty N, Balfe A, the gut microbiota. Lancet Diabetes Endocrinol 3(3):207–215
Maguire A, Mulcahy HE, Doherty G, Odonoghue D, Hyland 59. Fetissov SO (2017) Role of the gut microbiota in host appetite
J, Ross RP (2015) Spatial variation of the colonic microbiota control: bacterial growth to animal feeding behaviour. Nat Rev
in patients with ulcerative colitis and control volunteers. Gut Endocrinol 13(1):11–25
64(10):1553–1561 60. Louis P, Hold GL, Flint HJ (2014) The gut microbiota, bac-
42. Van den Abbeele P, Belzer C, Goossens M, Kleerebezem M, De terial metabolites and colorectal cancer. Nat Rev Microbiol
Vos WM, Thas O, De Weirdt R, Kerckhof FM, Van de Wiele 12(10):661–672
T (2013) Butyrate-producing Clostridium cluster XIVa species 61. Corrêa-Oliveira R, Fachi JL, Vieira A, Sato FT, Vinolo MA
specifically colonize mucins in an in vitro gut model. ISME J (2016) Regulation of immune cell function by short-chain fatty
7(5):949–961 acids. Clin Translat Immunol 5(4):e73
43. Ding T, Schloss PD (2014) Dynamics and associations of 62. Koh A, De Vadder F, Kovatcheva-Datchary P, Bäckhed F (2016)
microbial community types across the human body. Nature From dietary fiber to host physiology: short-chain fatty acids as
509(7500):357–360 key bacterial metabolites. Cell 165(6):1332–1345

13
International Urology and Nephrology (2024) 56:1345–1358 1355

63. Morrison DJ, Preston T (2016) Formation of short chain fatty 80. Graziani F, Pujol A, Nicoletti C, Dou S, Maresca M, Giardina
acids by the gut microbiota and their impact on human metabo- T, Fons M, Perrier J (2016) Ruminococcus gnavus E1 modu-
lism. Gut Microbes 7(3):189–200 lates mucin expression and intestinal glycosylation. J Appl
64. Derrien M, Vaughan EE, Plugge CM, de Vos WM (2004) Microbiol 120(5):1403–1417
Akkermansia muciniphila gen nov, sp nov, a human intesti- 81. Varyukhina S, Freitas M, Bardin S, Robillard E, Tavan E, Sapin
nal mucin-degrading bacterium. Int J syst Evolut Microbiol. C, Grill JP, Trugnan G (2012) Glycan-modifying bacteria-
54(5):1469–1476 derived soluble factors from Bacteroides thetaiotaomicron
65. Guarner F, Casellas F, Borruel N, Antolìn M, Videla S, Vilas- and Lactobacillus casei inhibit rotavirus infection in human
eca J (2003) Malagelada Jr Gut flora in health and disease. intestinal cells. Microbes Infect 14(3):273–278
Lancet 361:512–519 82. Freitas M, Cayuela C, Antoine JM, Piller F, Sapin C, Trugnan
66. Lin L, Zhang J (2017) Role of intestinal microbiota and metab- G (2001) A heat labile soluble factor from Bacteroides thetaio-
olites on gut homeostasis and human diseases. BMC Immunol taomicron VPI-5482 specifically increases the galactosylation
18(1):1–25 pattern of HT29-MTX cells. Cell Microbiol 3(5):289–300
67. Donohoe DR, Collins LB, Wali A, Bigler R, Sun W, Bult- 83. Konrad A, Cong Y, Duck W, Borlaza R, Elson CO (2006) Tight
man SJ (2012) The Warburg effect dictates the mechanism of mucosal compartmentation of the murine immune response
butyrate-mediated histone acetylation and cell proliferation. to antigens of the enteric microbiota. Gastroenterology
Mol Cell 48(4):612–626 130(7):2050–2059
68. Chambers ES, Morrison DJ, Frost G (2015) Control of appetite 84. Belkaid Y, Naik S (2013) Compartmentalized and systemic
and energy intake by SCFA: what are the potential underlying control of tissue immunity by commensals. Nat Immunol
mechanisms? Proce Nut Soc 74(3):328–336 14(7):646–653
69. Nagai M, Obata Y, Takahashi D, Hase K (2016) Fine-tuning 85. Shan M, Gentile M, Yeiser JR, Walland AC, Bornstein VU,
of the mucosal barrier and metabolic systems using the diet- Chen K, He B, Cassis L, Bigas A, Cols M, Comerma L (2013)
microbial metabolite axis. Int Immunopharmacol 1(37):79–86 Mucus enhances gut homeostasis and oral tolerance by deliver-
70. LeBlanc JG, Milani C, De Giori GS, Sesma F, Van Sinderen ing immunoregulatory signals. Science 342(6157):447–453
D, Ventura M (2013) Bacteria as vitamin suppliers to their 86. Peterson DA, McNulty NP, Guruge JL, Gordon JI (2007) IgA
host: a gut microbiota perspective. Curr Opin Biotechnol response to symbiotic bacteria as a mediator of gut homeosta-
24(2):160–168 sis. Cell Host Microbe 2(5):328–339
71. Martens JH, Barg H, Warren MA, Jahn D (2002) Micro- 87. Bansal T, Alaniz RC, Wood TK, Jayaraman A (2010) The
bial production of vitamin B 12. Appl Microbiol Biotechnol bacterial signal indole increases epithelial-cell tight-junction
58(3):275–285 resistance and attenuates indicators of inflammation. Proc Natl
72. Pompei A, Cordisco L, Amaretti A, Zanoni S, Matteuzzi D, Rossi Acad Sci 107(1):228–233
M (2007) Folate production by bifidobacteria as a potential pro- 88. Bevins CL, Salzman NH (2011) Paneth cells, antimicrobial
biotic property. Appl Environ Microbiol 73(1):179–185 peptides and maintenance of intestinal homeostasis. Nat Rev
73. Palau-Rodriguez M, Tulipani S, Isabel Queipo-Ortuño M, Urpi- Microbiol 9(5):356–368
Sarda M, Tinahones FJ, Andres-Lacueva C (2015) Metabolomic 89. Ehmann D, Wendler J, Koeninger L, Larsen IS, Klag T, Berger
insights into the intricate gut microbial–host interaction in the J, Marette A, Schaller M, Stange EF, Malek NP, Jensen BA
development of obesity and type 2 diabetes. Front Microbiol (2019) Paneth cell α-defensins HD-5 and HD-6 display dif-
27(6):1151 ferential degradation into active antimicrobial fragments. Proc
74. Smith K, McCoy KD, Macpherson AJ (2007) Use of axenic ani- Natl Acad Sci 116(9):3746–3751
mals in studying the adaptation of mammals to their commensal 90. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S,
intestinal microbiota. Sem Immunol 19:59–69 Medzhitov R (2004) Recognition of commensal microflora by
75. Swanson PA, Kumar A, Samarin S, Vijay-Kumar M, Kundu K, toll-like receptors is required for intestinal homeostasis. Cell
Murthy N, Hansen J, Nusrat A, Neish AS (2011) Enteric com- 118(2):229–241
mensal bacteria potentiate epithelial restitution via reactive 91. Price AE, Shamardani K, Lugo KA, Deguine J, Roberts AW,
oxygen species-mediated inactivation of focal adhesion kinase Lee BL, Barton GM (2018) A map of toll-like receptor expres-
phosphatases. Proc Natl Acad Sci 108(21):8803–8808 sion in the intestinal epithelium reveals distinct spatial, cell
76. Reunanen J, Kainulainen V, Huuskonen L, Ottman N, Belzer C, type-specific, and temporal patterns. Immunity 49(3):560–575
Huhtinen H, de Vos WM, Satokari R (2015) Akkermansia mucin- 92. Carvalho FA, Koren O, Goodrich JK, Johansson ME, Nalban-
iphila adheres to enterocytes and strengthens the integrity of the toglu I, Aitken JD, Su Y, Chassaing B, Walters WA, González
epithelial cell layer. Appl Environ Microbiol 81(11):3655–3662 A, Clemente JC (2012) Transient inability to manage proteo-
77. Chen HQ, Yang J, Zhang M, Zhou YK, Shen TY, Chu ZX, Zhang bacteria promotes chronic gut inflammation in TLR5-deficient
M, Hang XM, Jiang YQ, Qin HL (2010) Lactobacillus plantarum mice. Cell Host Microbe 12(2):139–152
ameliorates colonic epithelial barrier dysfunction by modulating 93. Vijay-Kumar M, Aitken JD, Carvalho FA, Cullender TC,
the apical junctional complex and PepT1 in IL-10 knockout mice. Mwangi S, Srinivasan S, Sitaraman SV, Knight R, Ley RE,
Am J Physiol-Gastroint Liver Physiol. 299(6):G1287–G1297 Gewirtz AT (2010) Metabolic syndrome and altered gut
78. Petersson J, Schreiber O, Hansson GC, Gendler SJ, Vel- microbiota in mice lacking Toll-like receptor 5. Science
cich A, Lundberg JO, Roos S, Holm L, Phillipson M (2011) 328(5975):228–231
Importance and regulation of the colonic mucus barrier in a 94. Ubeda C, Lipuma L, Gobourne A, Viale A, Leiner I,
mouse model of colitis. Am J Physiol-Gastroint Liver Physiol. Equinda M, Khanin R, Pamer EG (2012) Familial transmis-
300(2):G327–G333 sion rather than defective innate immunity shapes the dis-
79. Wrzosek L, Miquel S, Noordine ML, Bouet S, Chevalier-Curt tinct intestinal microbiota of TLR-deficient mice. J Exp Med
MJ, Robert V, Philippe C, Bridonneau C, Cherbuy C, Robbe- 209(8):1445–1456
Masselot C, Langella P (2013) Bacteroides thetaiotaomicron and 95. Wen L, Ley RE, Volchkov PY, Stranges PB, Avanesyan L, Stone-
Faecalibacterium prausnitzii influence the production of mucus braker AC, Hu C, Wong FS, Szot GL, Bluestone JA, Gordon JI
glycans and the development of goblet cells in the colonic epi- (2008) Innate immunity and intestinal microbiota in the develop-
thelium of a gnotobiotic model rodent. BMC Biol 11(1):1–3 ment of Type 1 diabetes. Nature 455(7216):1109–1113

13
1356 International Urology and Nephrology (2024) 56:1345–1358

96. Mazmanian SK, Round JL, Kasper DL (2008) A microbial 113. Wang P, Zhu S, Yang L, Cui S, Pan W, Jackson R, Zheng Y, Ron-
symbiosis factor prevents intestinal inflammatory disease. gvaux A, Sun Q, Yang G, Gao S (2015) Nlrp6 regulates intestinal
Nature 453(7195):620–625 antiviral innate immunity. Science 350(6262):826–830
97. Lee YK, Mehrabian P, Boyajian S, Wu WL, Selicha J, Vonder- 114. Gálvez EJ, Iljazovic A, Gronow A, Flavell R, Strowig T
fecht S, Mazmanian SK (2018) The protective role of Bacte- (2017) Shaping of intestinal microbiota in Nlrp6-and Rag2-
roides fragilis in a murine model of colitis-associated colorec- deficient mice depends on community structure. Cell Rep
tal cancer. MSphere 3(6):e00587-e618 21(13):3914–3926
98. Ramakrishna C, Kujawski M, Chu H, Li L, Mazmanian 115. Castro-Dopico T, Dennison TW, Ferdinand JR, Mathews RJ,
SK, Cantin EM (2019) Bacteroides fragilis polysaccharide Fleming A, Clift D, Stewart BJ, Jing C, Strongili K, Labzin LI,
A induces IL-10 secreting B and T cells that prevent viral Monk EJ (2019) Anti-commensal IgG drives intestinal inflam-
encephalitis. Nat Commun 10(1):1–3 mation and type 17 immunity in ulcerative colitis. Immunity
99. Erturk-Hasdemir D, Oh SF, Okan NA, Stefanetti G, Gazzaniga 50(4):1099–1114
FS, Seeberger PH, Plevy SE, Kasper DL (2019) Symbionts 116. Seo SU, Kamada N, Muñoz-Planillo R, Kim YG, Kim D, Koi-
exploit complex signaling to educate the immune system. Proc zumi Y, Hasegawa M, Himpsl SD, Browne HP, Lawley TD,
Natl Acad Sci 116(52):26157–26166 Mobley HL (2015) Distinct commensals induce interleukin-1β
100. Brown GD (2006) Dectin-1: a signalling non-TLR pattern- via NLRP3 inflammasome in inflammatory monocytes to pro-
recognition receptor. Nat Rev Immunol 6(1):33–43 mote intestinal inflammation in response to injury. Immunity
101. Tang CE, Kamiya T, Liu Y, Kadoki M, Kakuta S, Oshima 42(4):744–755
K, Hattori M, Takeshita K, Kanai T, Saijo S, Ohno N (2015) 117. Ratsimandresy RA, Indramohan M, Dorfleutner A, Stehlik C
Inhibition of dectin-1 signaling ameliorates colitis by induc- (2017) AIM2 inflammasome is a central regulator of intestinal
ing Lactobacillus-mediated regulatory T cell expansion in the homeostasis through the IL-18/IL-22/STAT3 pathway. Cell Mol
intestine. Cell Host Microbe 18(2):183–197 Immunol 14(1):127–142
102. Bouskra D, Brézillon C, Bérard M, Werts C, Varona R, Boneca 118. Saha S, Jing X, Park SY, Wang S, Li X, Gupta D, Dziarski R
IG, Eberl G (2008) Lymphoid tissue genesis induced by com- (2010) Peptidoglycan recognition proteins protect mice from
mensals through NOD1 regulates intestinal homeostasis. experimental colitis by promoting normal gut flora and prevent-
Nature 456(7221):507–510 ing induction of interferon-γ. Cell Host Microbe 8(2):147–162
103. Ramanan D, San Tang M, Bowcutt R, Cadwell K (2014) Bacte- 119. Jing X, Zulfiqar F, Park SY, Núñez G, Dziarski R, Gupta D
rial sensor Nod2 prevents inflammation of the small intestine (2014) Peptidoglycan Recognition Protein 3 and Nod2 Syner-
by restricting the expansion of the commensal Bacteroides gistically Protect Mice from Dextran Sodium Sulfate-Induced
vulgatus. Immunity 41(2):311–324 Colitis. J Immunol 193(6):3055–3069
104. Nigro G, Rossi R, Commere PH, Jay P, Sansonetti PJ (2014) 120. Franchi L, Amer A, Body-Malapel M, Kanneganti TD, Özören N,
The cytosolic bacterial peptidoglycan sensor Nod2 affords stem Jagirdar R, Inohara N, Vandenabeele P, Bertin J, Coyle A, Grant
cell protection and links microbes to gut epithelial regenera- EP (2006) Cytosolic flagellin requires Ipaf for activation of cas-
tion. Cell Host Microbe 15(6):792–798 pase-1 and interleukin 1β in salmonella-infected macrophages.
105. Janeway CA Jr, Medzhitov R (2002) Innate immune recogni- Nat Immunol 7(6):576–582
tion. Annu Rev Immunol 20(1):197–216 121. Zhu H, Xu WY, Hu Z, Zhang H, Shen Y, Lu S, Wei C, Wang ZG
106. Vaishnava S, Yamamoto M, Severson KM, Ruhn KA, Yu X, (2017) RNA virus receptor Rig-I monitors gut microbiota and
Koren O, Ley R, Wakeland EK, Hooper LV (2011) The antibac- inhibits colitis-associated colorectal cancer. J Exp Clin Cancer
terial lectin RegIIIγ promotes the spatial segregation of micro- Res 36(1):1–1
biota and host in the intestine. Science 334(6053):255–258 122. Hornung V, Hartmann R, Ablasser A, Hopfner KP (2014) OAS
107. Wang S, Charbonnier LM, Rivas MN, Georgiev P, Li N, Gerber proteins and cGAS: unifying concepts in sensing and responding
G, Bry L, Chatila TA (2015) MyD88 adaptor-dependent micro- to cytosolic nucleic acids. Nat Rev Immunol 14(8):521–528
bial sensing by regulatory T cells promotes mucosal tolerance 123. Chudnovskiy A, Mortha A, Kana V, Kennard A, Ramirez JD,
and enforces commensalism. Immunity 43(2):289–303 Rahman A, Remark R, Mogno I, Ng R, Gnjatic S, Amir EA
108. Broz P, Dixit VM (2016) Inflammasomes: mechanism of (2016) Host-protozoan interactions protect from mucosal
assembly, regulation and signalling. Nat Rev Immunol infections through activation of the inflammasome. Cell
16(7):407–420 167(2):444–456
109. Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss CA, 124. Mosser DM, Edwards JP (2008) Exploring the full spectrum of
Booth CJ, Peaper DR, Bertin J, Eisenbarth SC, Gordon JI, macrophage activation. Nat Rev Immunol 8(12):958–969
Flavell RA (2011) NLRP6 inflammasome regulates colonic 125. Danne C, Ryzhakov G, Martínez-López M, Ilott NE, Franchini
microbial ecology and risk for colitis. Cell 145(5):745–757 F, Cuskin F, Lowe EC, Bullers SJ, Arthur JS, Powrie F (2017) A
110. Levy M, Thaiss CA, Zeevi D, Dohnalová L, Zilberman-Scha- large polysaccharide produced by Helicobacter hepaticus induces
pira G, Mahdi JA, David E, Savidor A, Korem T, Herzig Y, an anti-inflammatory gene signature in macrophages. Cell Host
Pevsner-Fischer M (2015) Microbiota-modulated metabolites Microbe 22(6):733–745
shape the intestinal microenvironment by regulating NLRP6 126. Schulthess J, Pandey S, Capitani M, Rue-Albrecht KC, Arnold I,
inflammasome signaling. Cell 163(6):1428–1443 Franchini F, Chomka A, Ilott NE, Johnston DG, Pires E, McCul-
111. Wlodarska M, Thaiss CA, Nowarski R, Henao-Mejia J, Zhang lagh J (2019) The short chain fatty acid butyrate imprints an
JP, Brown EM, Frankel G, Levy M, Katz MN, Philbrick WM, antimicrobial program in macrophages. Immunity 50(2):432–445
Elinav E (2014) NLRP6 inflammasome orchestrates the colonic 127. Wu K, Yuan Y, Yu H, Dai X, Wang S, Sun Z, Wang F, Fei H, Lin
host-microbial interface by regulating goblet cell mucus secre- Q, Jiang H, Chen T (2020) The gut microbial metabolite trimeth-
tion. Cell 156(5):1045–1059 ylamine N-oxide aggravates GVHD by inducing M1 macrophage
112. Birchenough GM, Nyström EE, Johansson ME, Hansson polarization in mice. Blood 136(4):501–515
GC (2016) A sentinel goblet cell guards the colonic crypt 128. Constantinides MG, McDonald BD, Verhoef PA, Bendelac A
by triggering Nlrp6-dependent Muc2 secretion. Science (2014) A committed precursor to innate lymphoid cells. Nature
352(6293):1535–1542 508(7496):397–401

13
International Urology and Nephrology (2024) 56:1345–1358 1357

129. Gury-BenAri M, Thaiss CA, Serafini N, Winter DR, Giladi 145. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA,
A, Lara-Astiaso D, Levy M, Salame TM, Weiner A, David E, Bohlooly-y M, Glickman JN, Garrett WS (2013) The microbial
Shapiro H (2016) The spectrum and regulatory landscape of metabolites, short-chain fatty acids, regulate colonic Treg cell
intestinal innate lymphoid cells are shaped by the microbiome. homeostasis. Science 341(6145):569–573
Cell 166(5):1231–1246 146. Hegazy AN, West NR, Stubbington MJ, Wendt E, Suijker KI,
130. Sonnenberg GF, Hepworth MR (2019) Functional interactions Datsi A, This S, Danne C, Campion S, Duncan SH, Owens BM
between innate lymphoid cells and adaptive immunity. Nat Rev (2017) Circulating and tissue-resident CD4+ T cells with reactiv-
Immunol 19(10):599–613 ity to intestinal microbiota are abundant in healthy individuals
131. McDonald BD, Jabri B, Bendelac A (2018) Diverse develop- and function is altered during inflammation. Gastroenterology
mental pathways of intestinal intraepithelial lymphocytes. Nat 153(5):1320–1337
Rev Immunol 18(8):514–525 147. Miossec P, Kolls JK (2012) Targeting IL-17 and TH 17 cells in
132. Chun E, Lavoie S, Fonseca-Pereira D, Bae S, Michaud M, Hov- chronic inflammation. Nat Rev Drug Discovery 11(10):763–776
eyda HR, Fraser GL, Comeau CA, Glickman JN, Fuller MH, 148. Omenetti S, Bussi C, Metidji A, Iseppon A, Lee S, Tolaini M, Li
Layden BT (2019) Metabolite-sensing receptor Ffar2 regu- Y, Kelly G, Chakravarty P, Shoaie S, Gutierrez MG (2019) The
lates colonic group 3 innate lymphoid cells and gut immunity. intestine harbors functionally distinct homeostatic tissue-resident
Immunity 51(5):871–884 and inflammatory Th17 cells. Immunity 51(1):77–89
133. Bostick JW, Wang Y, Shen Z, Ge Y, Brown J, Zong-ming EC, 149. Atarashi K, Tanoue T, Ando M, Kamada N, Nagano Y, Narush-
Mohamadzadeh M, Fox JG, Zhou L (2019) Dichotomous regu- ima S, Suda W, Imaoka A, Setoyama H, Nagamori T, Ishikawa E
lation of group 3 innate lymphoid cells by nongastric Helico- (2015) Th17 cell induction by adhesion of microbes to intestinal
bacter species. Proc Natl Acad Sci 116(49):24760–24769 epithelial cells. Cell 163(2):367–380
134. Guo X, Liang Y, Zhang Y, Lasorella A, Kee BL, Fu YX (2015) 150. Naik S, Bouladoux N, Wilhelm C, Molloy MJ, Salcedo R, Kas-
Innate lymphoid cells control early colonization resistance tenmuller W, Deming C, Quinones M, Koo L, Conlan S, Spencer
against intestinal pathogens through ID2-dependent regulation S (2012) Compartmentalized control of skin immunity by resi-
of the microbiota. Immunity 42(4):731–743 dent commensals. Science 337(6098):1115–1119
135. Rankin LC, Girard-Madoux MJ, Seillet C, Mielke LA, 151. Dutzan N, Abusleme L, Bridgeman H, Greenwell-Wild T,
Kerdiles Y, Fenis A, Wieduwild E, Putoczki T, Mondot S, Zangerle-Murray T, Fife ME, Bouladoux N, Linley H, Brenchley
Lantz O, Demon D (2016) Complementarity and redundancy L, Wemyss K, Calderon G (2017) On-going mechanical damage
of IL-22-producing innate lymphoid cells. Nat Immunol from mastication drives homeostatic Th17 cell responses at the
17(2):179–186 oral barrier. Immunity 46(1):133–147
136. Chua HH, Chou HC, Tung YL, Chiang BL, Liao CC, Liu HH, Ni 152. Bedoui S, Heath WR, Mueller SN (2016) CD 4+ T-cell help
YH (2018) Intestinal dysbiosis featuring abundance of Rumino- amplifies innate signals for primary CD 8+ T-cell immunity.
coccus gnavus associates with allergic diseases in infants. Gas- Immunol Rev 272(1):52–64
troenterology 154(1):154–167 153. Bachem A, Makhlouf C, Binger KJ, de Souza DP, Tull D, Hoch-
137. Sterlin D, Fadlallah J, Adams O, Fieschi C, Parizot C, Dorgham heiser K, Whitney PG, Fernandez-Ruiz D, Dähling S, Kasten-
K, Rajkumar A, Autaa G, El-Kafsi H, Charuel JL, Juste C (2020) müller W, Jönsson J (2019) Microbiota-derived short-chain fatty
Human IgA binds a diverse array of commensal bacteria. J Exper acids promote the memory potential of antigen-activated CD8+
Med. 217(3):223 T cells. Immunity 51(2):285–297
138. Sutherland DB, Suzuki K, Fagarasan S (2016) Fostering of 154. Song X, Sun X, Oh SF, Wu M, Zhang Y, Zheng W, Geva-Zator-
advanced mutualism with gut microbiota by Immunoglobulin sky N, Jupp R, Mathis D, Benoist C, Kasper DL (2020) Micro-
A. Immunol Rev 270(1):20–31 bial bile acid metabolites modulate gut RORγ+ regulatory T cell
139. Kawamoto S, Maruya M, Kato LM, Suda W, Atarashi K, Doi Y, homeostasis. Nature 577(7790):410–415
Tsutsui Y, Qin H, Honda K, Okada T, Hattori M (2014) Foxp3+ 155. Crotty S (2014) T follicular helper cell differentiation, function,
T cells regulate immunoglobulin a selection and facilitate diver- and roles in disease. Immunity 41(4):529–542
sification of bacterial species responsible for immune homeosta- 156. Kawamoto S, Tran TH, Maruya M, Suzuki K, Doi Y, Tsutsui
sis. Immunity 41(1):152–165 Y, Kato LM, Fagarasan S (2012) The inhibitory receptor PD-1
140. Palm NW, De Zoete MR, Cullen TW, Barry NA, Stefanowski J, regulates IgA selection and bacterial composition in the gut. Sci-
Hao L, Degnan PH, Hu J, Peter I, Zhang W, Ruggiero E (2014) ence 336(6080):485–489
Immunoglobulin A coating identifies colitogenic bacteria in 157. Proietti M, Cornacchione V, Jost TR, Romagnani A, Faliti CE,
inflammatory bowel disease. Cell 158(5):1000–1010 Perruzza L, Rigoni R, Radaelli E, Caprioli F, Preziuso S, Bran-
141. Shulzhenko N, Morgun A, Hsiao W, Battle M, Yao M, Gavrilova netti B (2014) ATP-gated ionotropic P2X7 receptor controls
O, Orandle M, Mayer L, Macpherson AJ, McCoy KD, Fraser- follicular T helper cell numbers in Peyer’s patches to promote
Liggett C (2011) Crosstalk between B lymphocytes, microbiota host-microbiota mutualism. Immunity 41(5):789–801
and the intestinal epithelium governs immunity versus metabo- 158. Kubinak JL, Petersen C, Stephens WZ, Soto R, Bake E,
lism in the gut. Nat Med 17(12):1585–1593 O’Connell RM, Round JL (2015) MyD88 signaling in T cells
142. Nagashima K, Sawa S, Nitta T, Tsutsumi M, Okamura T, Pen- directs IgA-mediated control of the microbiota to promote health.
ninger JM, Nakashima T, Takayanagi H (2017) Identification of Cell Host Microbe 17(2):153–163
subepithelial mesenchymal cells that induce IgA and diversify 159. Teng F, Klinger CN, Felix KM, Bradley CP, Wu E, Tran NL,
gut microbiota. Nat Immunol 18(6):675–682 Umesaki Y, Wu HJ (2016) Gut microbiota drive autoimmune
143. Arpaia N, Campbell C, Fan X, Dikiy S, Van Der Veeken J, Deroos arthritis by promoting differentiation and migration of Peyer’s
P, Liu H, Cross JR, Pfeffer K, Coffer PJ, Rudensky AY (2013) patch T follicular helper cells. Immunity 44(4):875–888
Metabolites produced by commensal bacteria promote peripheral 160. Rescigno M, Rotta G, Valzasina B, Ricciardi-Castagnoli P (2001)
regulatory T-cell generation. Nature 504(7480):451–455 Dendritic cells shuttle microbes across gut epithelial monolayers.
144. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Immunobiology 204(5):572–581
Y, Cheng G, Yamasaki S, Saito T, Ohba Y, Taniguchi T (2011) 161. Martínez-López M, Iborra S, Conde-Garrosa R, Mastrangelo
Induction of colonic regulatory T cells by indigenous Clostrid- A, Danne C, Mann ER, Reid DM, Gaboriau-Routhiau V, Chap-
ium species. Science 331(6015):337–341 arro M, Lorenzo MP, Minnerup L (2019) Microbiota sensing by

13
1358 International Urology and Nephrology (2024) 56:1345–1358

Mincle-Syk axis in dendritic cells regulates interleukin-17 and- 176. Caruso R, Lo BC, Núñez G (2020) Host–microbiota interactions
22 production and promotes intestinal barrier integrity. Immunity in inflammatory bowel disease. Nat Rev Immunol 20(7):411–426
50(2):446–461 177. Ronco C, Bellomo R, Kellum JA (2019) Acute kidney injury.
162. Jie Z, Yang JY, Gu M, Wang H, Xie X, Li Y, Liu T, Zhu L, Shi J, Lancet 394(10212):1949–1964
Zhang L, Zhou X (2018) NIK signaling axis regulates dendritic 178. Khwaja A (2012) KDIGO clinical practice guidelines for acute
cell function in intestinal immunity and homeostasis. Nat Immu- kidney injury. Nephron Clin Pract 120(4):c179–c184
nol 19(11):1224–1235 179. Li X, Yuan F, Zhou L (2022) Organ crosstalk in acute kidney
163. Wingender G, Hiss M, Engel I, Peukert K, Ley K, Haller H, Kro- injury: evidence and mechanisms. J Clin Med 11(22):6637
nenberg M, von Vietinghoff S (2012) Neutrophilic granulocytes 180. Radi ZA (2018) Immunopathogenesis of acute kidney injury.
modulate invariant NKT cell function in mice and humans. J Toxicol Pathol 46(8):930–943
Immunol 188(7):3000–3008 181. Lee SA, Noel S, Sadasivam M, Hamad AR, Rabb H (2017)
164. An D, Oh SF, Olszak T, Neves JF, Avci FY, Erturk-Hasdemir D, Role of immune cells in acute kidney injury and repair. Nephron
Lu X, Zeissig S, Blumberg RS, Kasper DL (2014) Sphingolipids 137(4):282–286
from a symbiotic microbe regulate homeostasis of host intestinal 182. Vallés PG, Lorenzo AG, Bocanegra V, Vallés R (2014) Acute
natural killer T cells. Cell 156(1–2):123–133 kidney injury: what part do toll-like receptors play? Int J Nephrol
165. Yoo JY, Groer M, Dutra SV, Sarkar A, McSkimming DI (2020) Renov Dis 19:241–251
Gut microbiota and immune system interactions. Microorgan- 183. Habib R (2021) Multifaceted roles of Toll-like receptors in acute
isms 8(10):1587 kidney injury. Heliyon 7(3):e06441
166. Tlaskalova-Hogenova H, Tuckova L, Mestecky J, Kolinska J, 184. Andrade-Oliveira V, Amano MT, Correa-Costa M, Castoldi A,
Rossmann P, Stepankova R, Kozakova H, Hudcovic T, Hrncir T, Felizardo RJ, de Almeida DC, Bassi EJ, Moraes-Vieira PM,
Frolova L, Kverka M (2005) Interaction of mucosal microbiota Hiyane MI, Rodas AC, Peron JP (2015) Gut bacteria products
with the innate immune system. Scand J Immunol 62:106–113 prevent AKI induced by ischemia-reperfusion. J Am Soc Nephrol
167. Riazi-Rad F, Behrouzi A, Mazaheri H, Katebi A, Ajdary S (2021) 26(8):1877–1888
Impact of gut microbiota on immune system. Acta Microbiol 185. Al-Harbi NO, Nadeem A, Ahmad SF, Alotaibi MR, Alasmari
Immunol Hung 68(3):135–144 AF, Alanazi WA, Al-Harbi MM, El-Sherbeeny AM, Ibrahim KE
168. Maynard CL, Elson CO, Hatton RD, Weaver CT (2012) Recipro- (2018) Short chain fatty acid, acetate ameliorates sepsis-induced
cal interactions of the intestinal microbiota and immune system. acute kidney injury by inhibition of NADPH oxidase signaling
Nature 489(7415):231–241 in T cells. Int Immunopharmacol 1(58):24–31
169. Abraham C, Medzhitov R (2011) Interactions between the host 186. Lobel L, Cao YG, Fenn K, Glickman JN, Garrett WS (2020) Diet
innate immune system and microbes in inflammatory bowel dis- posttranslationally modifies the mouse gut microbial proteome
ease. Gastroenterology 140(6):1729–1737 to modulate renal function. Science 369(6510):1518–1524
170. Sugihara K, Kamada N (2021) Diet–microbiota interactions in
inflammatory bowel disease. Nutrients 13(5):1533 Publisher's Note Springer Nature remains neutral with regard to
171. McCoy KD, Burkhard R, Geuking MB (2019) The micro- jurisdictional claims in published maps and institutional affiliations.
biome and immune memory formation. Immunol Cell Biol
97(7):625–635 Springer Nature or its licensor (e.g. a society or other partner) holds
172. Kogut MH, Lee A, Santin E (2020) Microbiome and pathogen exclusive rights to this article under a publishing agreement with the
interaction with the immune system. Poult Sci 99(4):1906–1913 author(s) or other rightsholder(s); author self-archiving of the accepted
173. Cosola C, Rocchetti MT, Gesualdo L (2021) Gut microbiota, manuscript version of this article is solely governed by the terms of
the immune system, and cytotoxic T lymphocytes. Cytotoxic such publishing agreement and applicable law.
T-Cells: Meth Prot. 1:229–241
174. Wang L, Zhu L, Qin S (2019) Gut microbiota modulation on
intestinal mucosal adaptive immunity. J Immunol Res 1:19
175. Hayase E, Jenq RR (2021) Role of the intestinal microbiome and
microbial-derived metabolites in immune checkpoint blockade
immunotherapy of cancer. Genome Med 13(1):107

13

You might also like