Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

REvIEWS

Immunity, microbiota and kidney


disease
Felix Knauf1,2, J. Richard Brewer3 and Richard A. Flavell   3,4*
Abstract | The recognition that intestinal microbiota exert profound effects on human health has
led to major advances in our understanding of disease processes. Studies over the past 20 years
have shown that host components, including components of the host immune system, shape
the microbial community. Pathogenic alterations in commensal microorganisms contribute
to disease manifestations that are generally considered to be noncommunicable, such as
inflammatory bowel disease, diabetes mellitus and liver disease, through a variety of mechanisms,
including effects on host immunity. More recent studies have shed new light on how the immune
system and microbiota might also drive the pathogenesis of renal disorders. In this Review , we
discuss the latest insights into the mechanisms regulating the microbiome composition, with a
focus both on genetics and environmental factors, and describe how commensal microorganisms
calibrate innate and adaptive immune responses to affect the activation threshold for pathogenic
stimulations. We discuss the mechanisms that lead to intestinal epithelial barrier inflammation
and the relevance of certain bacteria to the pathogenesis of two common kidney-​based
disorders: hypertension and renal stone disease. Limitations of current approaches to microbiota
research are also highlighted, emphasizing the need to move beyond studies of correlation
to causation.

Renal disease is a leading contributor to mortality world- areas of medical research, including the field of neph-
wide and is increasingly recognized as a global health rology, are now directing part of their research efforts
problem1–3. The disease pathways that lead to compro- towards understanding the role of microbiota in health
mised renal function are diverse and poorly under- and disease.
stood. Consequently, few strategies are available to slow The interaction of the microbiota with the host
the progression of renal disease, with most treatment immune system is a major focal point of interest12–14.
options centred around lowering blood pressure and As the gastrointestinal tract must tolerate the presence of
reducing proteinuria4. Clearly, additional therapeutic luminal bacteria and protect resident cells from invading
1
Department of Nephrology
and Medical Intensive Care,
avenues are needed. Research over the past two decades pathogens, a sophisticated orchestra of various instru-
Charité–Universitätsmedizin has linked imbalances in the composition and function ments has evolved with immunity being the conductor.
Berlin, Berlin, Germany. of gut microbiota, a condition termed dysbiosis, with Gnotobiotic (that is, germ-​free) mice are an essential
2
Section of Nephrology, noncommunicable illnesses such as diabetes mellitus5, tool for deciphering the underlying score. The intestine
Department of Internal obesity6, non-​alcoholic fatty-​liver disease7, malignan- of germ-​free mice contains a severely underdeveloped
Medicine, Yale University
cies8 and major depressive disorders9. Advances in immune system that is restored after colonization with
School of Medicine, New
Haven, CT, USA.
the accuracy of microbial genomic DNA analysis and the bacteria15–17. High-​resolution single-​cell surveys of the
3
Department of
development of novel computational tools, combined gastrointestinal epithelium have demonstrated that
Immunobiology, Yale with mechanistic studies in model systems, are acceler- microorganisms, including pathogenic microorgan-
University School of Medicine, ating our understanding of the importance of the micro- isms such as Salmonella spp., can increase the abundance
New Haven, CT, USA. biota in health and disease10. These advances have led of Paneth cells and enterocytes and alter the composition of
4
Howard Hughes Medical to the identification of new diagnostic biomarkers and absorptive or secretory cell types within the epithelial
Institute, Yale University a surge in translational research in a number of fields. barrier, which has been speculated to have a critical role
School of Medicine, New
Haven, CT, USA.
In fact, enthusiasts have suggested that the field of ‘pre- in the defence against pathogens18. Follow-​up studies will
cision medicine’, which is currently dominated by stud- be required to examine this hypothesis18. Microbial dys-
*e-​mail: Richard.Flavell@
yale.edu ies of the genetic contribution to disease, will be greatly biosis and dysregulation of gut epithelial barrier func-
https://doi.org/10.1038/ enhanced by the ability to tailor therapeutic regimens to tion can also lead to the generation of toxic compounds
s41581-019-0118-7 an individual’s microbiota profile11. As a consequence, all that leak into the circulation. Endotoxin is a component

Nature Reviews | Nephrology


Reviews

Key points leads to microbial uniformity among animals of differ-


ent genotypes or that have been exposed to different
• The gut microbiota influences the host through reciprocal interactions with the host treatments and is used to control for biological effects
immune system. caused by commensal microorganisms26. In humans,
• Imbalances in the microbiota (dysbiosis) lead to epithelial barrier dysfunction and horizontal transfer of microorganisms also seems to
translocation of toxic compounds to the systemic circulation. occur through cohabitation. Of note, infants with older
• Hypertension and kidney stone disease might be linked to compositional or functional siblings have differences in their commensal microbial
changes in the gut microbiome. composition that are believed to provide protection
• Novel research methods are being established to identify causal microorganisms that from atopic diseases27,28. Interspecies interactions can
drive disease progression. also shape human commensal microorganisms in a
• Understanding the crosstalk between immunity, the microbiota and the kidney has beneficial manner; multiple studies have documented
potential for the development of innovative, paradigm-​shifting treatment strategies. increased microbial richness and diversity in human gut
flora of pet owners and dairy farmers27,29. This enhanced
of the outer membranes of Gram-​negative bacteria and exposure to animals and their microorganisms is
is found at elevated levels in the plasma of patients with believed to promote the development of the immune
chronic kidney disease19,20. Plasma endotoxin levels are system and to protect from conditions such as allergies,
also associated with systemic inflammation, accelerated asthma and eczema30,31.
Paneth cells atherosclerosis and cardiovascular disease21. Levels of tri- Emerging research also indicates that the species
Epithelial cells localized in the
methylamine N-​oxide (TMAO) — a gut-​derived amine composition and diversity of the microbiota are influ-
glands of the small intestine.
Microscopically, they are oxide — are also elevated in patients with chronic kid- enced by several noncommunicable environmental fac-
characterized by large ney disease, in whom they might contribute to athero­ tors experienced by the host. Of note, diet serves as a
eosinophilic granules that genesis and the increased risk of cardiovascular disease major determinant of the commensal flora. Acute altera-
occupy the cytoplasm and through dysregulation of lipid handling and the forma- tions in dietary macronutrients induce transient changes
contain antimicrobial
compounds critical for
tion of macrophage foam cells22. Greater understanding in the species composition of human and mouse gut flora
host defence. of the crosstalk between immunity and gut micro­ within days32,33. However, persistent dietary factors also
biota has potential for the development of innovative, cause durable effects on gut microbial diversity, which
Enterocytes paradigm-​shifting strategies for the treatment of several in turn influences host physiology. For example, artifi-
Epithelial cells found in the
noncommunicable diseases, including kidney disease. cial food additives, such as zero-​calorie sweeteners or
small intestine. Microscopically,
they are characterized by Here, we summarize the current understanding of the emulsifying agents, can induce metabolic syndrome and
microvilli found on the apical reciprocal interactions between the immune system and inflammatory disease in a microbiota-​dependent man-
membrane. Enterocytes are intestinal microbiota, focusing on select components of ner34,35. Non-​calorie artificial sweeteners alter microbial
responsible for the absorption the immune system that have been shown to drive the metabolic pathways that influence host energy homeo-
or secretion of molecules
from or into the intestine.
pathogenesis of chronic diseases, not only in the intes- stasis whereas emulsifying agents expand the population
tine but also within the kidney. We also highlight studies of mucolytic bacteria, leading to impairment of mucus
Macrophage foam cells that demonstrate how our understanding of the asso­ barrier function34,35.
Monocyte-​derived ciation of the microbiome with disease can be moved to The host also regulates gut microbial composition
macrophages that clear
understanding the causal role of specific microorganism through a variety of genetically determined mechanisms
oxidized lipids and in
this process develop into populations in disease and how this knowledge might (Fig. 1). Mucosal epithelia secrete a protective layer of
lipid-laden foam cells. These advance the field of renal research. mucus to establish a physical barrier between the host
cells are components of and microorganisms. This viscous gel is produced by
atherosclerotic plaques and The composition of the gut microbiota goblet cells and consists of highly glycosylated mucin pro-
have been implicated in the
pathogenesis of atherosclerotic
Humans are deluged by a broad spectrum of micro- teins, such as MUC2, arranged in a net-​like polymer36.
disease. organisms, which reside at all barrier surfaces. These The structure of the mucus layer differs along the gastro-
microbial communities are collectively referred to as the intestinal tract and is thickest in the large intestine, where
Coprophagic animals ‘microbiota’ and consist of bacteria, viruses, archaea and the mucus barrier consists of two distinct compartments
Animals that eat faeces, which
fungi. Within the gastrointestinal tract alone, the num- described as the ‘inner’ and ‘outer’ layers37. The inner
facilitates the transmission of
microbiomes between ber of bacterial cells is estimated to be comparable to that layer prevents bacterial adherence to the epithelium, is
individual animals. of host-​derived cells23. Furthermore, commensal micro- more dense and contains fewer microbial species than
organisms are believed to encode 100 times the genetic the outer layer38. Mucolytic degradation of the inner layer
Goblet cells information contained within the human genome24. by host or microbially derived degrading enzymes loos-
Epithelial cells found in
various organs. Goblet cells
Given the magnitude of the interactions between host ens the structure of the outer mucus layer, facilitating
secrete mucus, a viscous fluid and microbiota components, it is no surprise these bacterial invasion. Mice that are deficient in MUC2 fail
composed primarily of proteins microorganisms exert profound effects on virtually all to establish this inner mucus layer, which enables bacte-
called mucins. aspects of host physiology. ria to invade the underlying colonic epithelium and leads
At birth, maternal commensal microorganisms are to inflammation of the colon38. Although the production
T helper 2 (TH2) cells
Specialized population of transmitted vertically (that is, from mother to child) to of mucus by goblet cells is constitutive, this process is
T cells that orchestrate offspring to seed an organism’s initial flora25. Following enhanced as part of the response of T helper 2 (TH2) cells
protective type 2 immune birth, commensal and pathogenic microorganisms to extracellular pathogens39. Innate immune factors
responses to pathogens such are exchanged horizontally through interspecies and can also regulate mucus production. NOD-, LRR- and
as parasites as well as tissue
repair. TH2 cells have also been
intraspecies interactions. In the laboratory, co-​housing pyrin domain-​containing 6 (NLRP6), which, similar
implicated in diseases such of coprophagic animals facilitates the horizontal trans- to other NOD-​like receptors, is an intracellular sen-
as asthma. fer of microorganisms between animals. This practice sor of pathogen-​associated molecular patterns (PAMPs),

www.nature.com/nrneph
Reviews

IgA Intestinal
Outer
mucus layer bacteria

AMP
Inner
mucus layer

Secretion
of mucus

MyD88 NLRP6

Intestinal Goblet TLR Paneth


epithelium cell IL-18 cell
Plasma iNKT
TH2 cell
cytokines cell

Fig. 1 | regulation of gut microbial composition by host mechanisms. Goblet cells within the intestine produce a
protective mucus layer. The structure of this mucus layer differs along the gastrointestinal tract. In the large intestine,
the mucus barrier consists of two compartments called the inner and outer layer. The inner layer is more dense than the
outer layer and contains fewer microbial species. The mucus layers also contain antimicrobial proteins (AMPs) and
immunoglobulin A (IgA), produced by Paneth cells and plasma cells, respectively , which limit adherence of bacteria to
the epithelium. The production of mucus is regulated by innate factors such as NOD-, LRR- and pyrin domain-​containing
6 (NLRP6), whereas numbers of goblet cells are enhanced by T helper 2 (TH2) cytokines, such as IL-4 and IL-13, but limited
by the inflammasome cytokine IL-18. The production of AMPs by Paneth cells requires Toll-​like receptor (TLR)–MyD88
signalling and is further promoted by signals from intestinal natural killer T (iNKT) cells.

promotes exocytosis of mucin granules by inducing auto- Paneth cell function. Invariant natural killer T (NKT) cells
phagy in goblet cells40,41. In addition, elevated levels of respond to lipid antigens presented by the major histo-
the pro-​inflammatory cytokine IL-18 promote a devel- compatibility complex (MHC)-like protein CD1d and
opmental programme within the intestinal crypt that promote degranulation of Paneth cells54. Disruption of
results in fewer goblet cells and ultimately compromises this axis by genetic ablation of CD1d enabled ectopic
mucosal integrity42. colonization of the small intestine by multiple bacterial
Specialized epithelial and immune cells within the species and increased epithelial adherence54.
gastrointestinal tract further control the species com- Beneath the intestinal epithelium, plasma cells that
position and localization of commensal microorgan- reside in the lamina propria secrete immunoglobulin
isms by establishing chemical barricades within the A (IgA), which serves as an additional chemical con-
mucus layer. In the small intestine, Paneth cells secrete trol of intestinal microorganisms. Pathogens are coated
multiple proteins into the lumen with mechanistically with high-​affinity IgA molecules; however, the affinity
disparate antimicrobial properties. For example, Paneth of microbial symbionts for IgA are often much lower55,56.
cells secrete proteins of the defensin and REG3 (known This coating can neutralize microbial virulence factors,
Pathogen-​associated
molecular patterns
as HIP or PAP in humans) families of antimicrobial motility or invasiveness, leading to immune exclusion57–60,
(PAMPs). Highly conserved peptides, which are small, pore-​forming proteins that although the absence of B cells (which give rise to IgA-​
groups of molecular motifs can lyse bacterial cell walls43,44. Paneth cells also secrete secreting plasma cells) or IgA induces only modest
detected by pattern hydrolases, such as lysozymes, secretory phospholipase alterations in the gut flora54.
recognition receptor
A2 and angiogenin 4 (ANG4), which exhibit bacteri- Pathogenic alterations in commensal microorgan-
(PRR)-bearing cells of the
innate immune system. cidal activity by cleaving peptidoglycans, phospholipids isms can also arise as a consequence of genetic defi-
and RNA, respectively43,45–48. The bactericidal activity of ciencies, with failure to control enteric flora resulting in
Natural killer T (NKT) cells ANG4, for example, seems to preferentially target the systemic effects on a broad range of host physiological
Subset of T cells that express pathogen Listeria monocytogenes; it is less effective at processes. For example, Tlr5 deficiency leads to micro­
both a T cell receptor, a
classical component of
killing commensals such as Bacteroides thetaiotaomi- bially dependent alterations in host metabolism marked
adaptive immunity, and surface cron48. The production of antimicrobial peptides by by hyperphagia, increased adiposity, hypertension and
receptors for natural killer cells, Paneth cells is constitutive and depends on the innate insulin resistance61,62. Microbially dependent altera-
which are characteristic of recognition of PAMPs. The activation of various Toll-​ tions in the composition of caecal short-​chain fatty
innate immunity.
like receptors (TLRs) increases the expression of defen- acids (SCFAs) of Tlr5-deficient mice increase hepatic
Immune exclusion sins and increases release of antimicrobial proteins via lipogenesis, contributing in part to the development of
Process of clearing pathogenic degranulation of Paneth cells49–52. Further, expression of these metabolic derrangements63. As indicated above, the
microorganisms. Secretory TLRs, IL-1R, IL-18R and the adaptor protein MyD88 inflammasome has also emerged as a critical regulator
immunoglobulin A (IgA) has in Paneth cells is both necessary and sufficient for of the microbiome. Mice that lack the inflammasome
a central role in this process
by blocking antigens and
the production of REG3γ and the establishment of a component Nlrp6 exhibit an over-​representation in the
pathogenic microorganisms bacteria-​depleted mucus barrier adjacent to the intesti- phylum Bacteroidetes and increased susceptibility to
from the intestinal lumen. nal epithelium53. Specialized lymphocytes also regulate chemically induced colitis — a phenotype that can be

Nature Reviews | Nephrology


Reviews

Vivaria transferred horizontally to wild-​type mice64. This pheno- with resident microbial communities72. This observation
Enclosed areas for live animals type was also observed in mice lacking other inflammas- is partially explained by the complex microbial ecology
in a semi-​natural environment ome signalling components, including ASC, caspase 1 in the gut, whereby host niches that are saturated by
for observation or studies. and IL-18 (ref.64). NLRP6 and NLRP3 inflammasome commensals leave few resources for invading patho-
sensors also regulate the ability of enteric microbiota to gens. It is also clear that commensal flora are required
restrict leakage of TLR agonists through the portal vein for proper postnatal development of the immune system.
to the liver, thereby protecting against non-​alcoholic Germ-​free mice exhibit morphological differences in the
steatohepatitis7. Similar to findings in mice with chem- intestinal epithelium, altered expression of antimicro-
ically induced colitis, liver disease could be transferred bial proteins, immature gut-​associated lymphoid tissues,
horizontally by co-​housing inflammasome-​deficient dampened IgA production, fewer intestinal lymphocytes
mice with wild-​type animals7. The link between TLR and an imbalance in T effector cell subsets with a prefer­
and inflammasome signalling with microbial dysbiosis ence towards TH2 cells44,73,74. Most of these differences
has been reproduced by some groups65–67 but not by oth- are reversed by recolonization of germ-​free animals
ers68–70. Differences in environmental factors, experimen- with species-​matched flora, indicating these processes
tal design and microbial communities that exist between are regulated by continuous interaction between the host
vivaria have hampered the reproducibility of microbiome and commensal microorganisms13,73. Although many of
research and led to several controversies in the field. the mechanisms by which microorganisms exert their
Although some of these variables might prove difficult effects on host development and physiology remain
to normalize across global research institutions, stand- enigmatic, considerable progress has been made in
ardized practices have been proposed, and their adoption studying these interactions.
is likely to enhance reproducibility within the field26. In some cases, structural elements of microorgan-
isms are responsible for their immunomodulatory
Influence of microbiota on immunity effects. Polysaccharide A (PSA) and glycosphingolipids
Analyses of germ-​free animals have helped to identify derived from Bacteroides fragilis are one such example75.
some of the means by which microorganisms influence Administration of PSA to germ-​free mice facilitates the
host physiology. Germ-​free rats require increased food proliferation of T cells, restores the balance of TH2 to TH1
consumption to maintain their body weight, and they effector cells and promotes the normal development of
expel more faecal calories than conventionally raised gut-​associated lymphoid tissues76. In addition, B. fragilis
animals, suggesting a key role for microorganisms in induces the production of immunosuppressive regula-
energy extraction and metabolic processes71. Indeed, tory T (Treg) cells and ameliorates disease progression
microorganisms have a crucial role in catabolizing in mouse models of colitis and multiple sclerosis in a
complexed carbohydrates and dietary fibre. However, in PSA-​dependent manner77–79. Mechanistically, PSA asso-
addition to enhancing energy extraction, many micro- ciates with outer membrane vesicles to engage TLR2 on
organisms and their diet-​derived microbial products dendritic cells, which in turn induces IL-10 expression
are immunomodulatory and regulate host physiology in CD4+CD25+ Treg cells80–82 (Fig. 2a). Glycosphingolipids
through immune-​mediated mechanisms. Early studies derived from B. fragilis can also reduce the severity of
of germ-​free mice have demonstrated that these animals colitis in animal models by inhibiting the proliferation
are more readily colonized by pathogens than animals of intestinal NKT cells in the lamina propria83.

a Bacteroides b Dietary fibre


fragilis Gut
microbiota
PSA
Short-chain fatty acids
TLR2 Acetate
Butyrate

DC Propionate

MHC GPR43
class II TCR
HDAC
NLRP3
Treg IL-10
cell Haematopoietic Intestinal T cell
or stromal cell epithelial cell
Tight junctions
Treg cell Activation HDAC inhibition and barrier
induction of GPCRs and T cell fate function

Fig. 2 | Microorganism-​derived factors influence immunity. a | The bacterial structural product polysaccharide A (PSA),
derived from microorganisms such as Bacteroides fragilis, activates Toll-​like receptor 2 (TLR2) on dendritic cells (DCs) to
promote the induction of regulatory T (Treg) cells, which produce IL-10. b | Intestinal bacteria also break down dietary fibre
into short-​chain fatty acids, which influence host physiology by acting as ligands for G protein-​coupled receptors (GPCRs),
such as GPCR 43 (GPR43) expressed on haematopoietic stromal cells and intestinal epithelial cells (in which it regulates
the NOD-, LRR- and pyrin domain-​containing 3 (NLRP3) inflammasome), by inhibiting histone deacetylases (HDACs),
leading to the induction of Treg cells, and by reinforcing tight junctions to promote barrier function. MHC, major
histocompatibility complex; TCR , T cell receptor.

www.nature.com/nrneph
Reviews

As mentioned earlier, metabolites derived from the effect and enhanced NLRP6-dependent IL-18 production
breakdown of dietary components by microorganisms to stimulate the production of antimicrobial proteins.
can also influence the host’s immune system (Fig. 2b). The Of note, histamine and spermine are more abundant
SCFAs acetate, propionate and butyrate, for example, are under conditions of dysbiosis than under normal
produced by enteric bacterially -mediated catabolism of physio­logical conditions and might therefore contrib-
dietary fibre and cannot be produced by host digestive ute to inflammation by restricting the inflammasome66.
enzymes. Consequently, germ-​free mice contain lower Collectively, these studies demonstrate that host and com-
concentrations of SCFAs throughout their intestinal mensal microorganisms interact through many intricate
tract84. Studies from the past decade indicate that some mechanisms with profound influences on host physiology.
of the beneficial effects of dietary fibre are mediated
by G protein-​coupled receptor (GPCR) 43 (GPR43; The epithelial barrier in inflammation
also known as free fatty acid receptor 2), a GPCR that Although serving as an immune barrier, the intestinal
is expressed on haematopoietic and stromal cells and is epithelium must maintain some degree of permeability
activated by acetate, propionate and butyrate85–87. This to facilitate nutrient uptake. To balance these seemingly
proposal is supported by the finding that administration contradictory roles, specialized secretory cells, called gob-
of a high-​fibre diet reduced severity of colitis in conven- let cells, produce a mucus layer containing anti­microbial
tional but not germ-​free mice, suggesting that a microbi- proteins. As described above, this layer establishes a pro-
ally derived factor is capable of suppressing inflammation tective boundary that prevents pathogens and commen-
in this model85. Dietary acetate did, however, protect sals from invading the host. Beneath this mucus barrier,
germ-​free animals from colitis in a GPR43-dependent epithelial cells further regulate gut permeability through
manner. Similar effects were also observed in mouse paracellular transport and transcellular transport pathways.
models of arthritis and lung inflammation, indicating Paracellular transport is limited by tight junctions,
that the immunosuppressive effects of acetate mediated which attach the apical–lateral surfaces of epithelial cells
by GPR43 may be systemic85. In the intestine, GPR43 together and restrict the ability of large molecules to trav-
functions in intestinal epithelial cells by regulating erse the epithelium. Although tight junctions allow water,
the NLRP3 inflammasome and IL-18 secretion88. ions and other small molecules to pass between epithelial
SCFAs produced by commensal microorganisms also cells, tissue permeability varies along the gastrointestinal
influence lymphocytes by modulating their epigenetic tract depending on the composition of the tight junctions
status. The induction of peripheral Treg cells is dependent (that is, the presence of various transmembrane proteins
on commensal flora and is increased in mice placed on a such as claudins, occludins, junctional adhesion mole-
high-​fibre diet89–91. This increase in peripheral Treg cells cules and tricellulin, as well as the zonula occludens intra-
depends on bacterially produced butyrate, which cellular scaffold proteins) and their abundance. Although
enhances the frequency of Treg cells in vitro and increases considerable genetic redundancy exists within this sys-
the number of extra-​thymically derived Treg cells in vivo. tem, mice that lack tight junction proteins claudin 7,
Butyrate inhibits histone deacetylases (HDACs), leading JAMA or EpCAM exhibit increased epithelial permea-
to an accumulation of histone acetylation at the Foxp3 bility and spontaneously develop intestinal inflamma-
locus and increased abundance of FOXP3 protein — an tion98–100, highlighting the importance of tight junctions
Histone deacetylases important regulator of Treg cells92. Dietary butyrate also in establishing the gut barrier and demonstrating that
(HDACs). Class of enzymes that reduces disease severity in a model of T cell-​dependent disruption of this process leads to autoinflammation.
remove acetyl groups. HDACs colitis, further supporting a role for this SCFA in regulat- Increased intestinal permeability is also thought
regulate access to DNA by ing the activity of lymphocytes89,90. More recent studies to contribute to several autoinflammatory disorders
modulating chromatin and
thereby cellular processes.
have shown that SCFAs promote the differentiation of and could serve as a tool to aid diagnosis as well as to
HDACs are also involved in TH17 and TH1 cells by inhibiting HDACs91, indicating evaluate the risk of disease progression and relapse101.
immunity by, for example, that microbially derived SCFAs influence multiple T cell For example, impairment of barrier function, poten-
regulating CD4+ T cells. subsets. In addition, some bacterial species induce spe- tially caused by altered expression of tight junction
cific lymphocyte subsets by adhering to the intestinal proteins, has been reported in patients with inflamma-
Paracellular transport
Transfer of molecules or epithelium to influence the expression of host genes93. tory bowel disease (IBD)102. In vitro, pro-​inflammatory
solutes across the epithelium For example, adherence of segmented filamentous bac- and anti-​inflammatory cytokines repress and promote
by passing through the teria (SFB) to the intestinal epithelium of the ileum is the expression and function of tight junction proteins,
intercellular space between required for the induction of TH17 cells94. Adherence of respectively103–106, suggesting that immunomodulatory
cells. This route of transport
can be enhanced if junctional
SFB enables epithelial cells to secrete serum amyloid A, strategies for autoinflammatory disorders might function
proteins are displaced or have which acts on CD11c+ cells (most likely dendritic cells) in part by modulating intestinal permeability107,108.
altered expression. to promote polarization of TH17 cells94–97. Several dietary and microbial factors can modulate
Histamine, spermine and taurine represent a group intestinal barrier function through effects on tight junc-
Transcellular transport
of commensal-​derived metabolites that influence the tion proteins. Of note, consumption of a Western diet
Transfer of molecules or solutes
across the epithelium by innate immune system by activating the inflammas- has been shown to influence the gut microbial compo-
passing through the cell. This ome66. Histamine and spermine, for example, suppressed sition and lead to increased intestinal permeability109.
route of transport is mediated NLRP6, ASC and caspase 1 and/or caspase 11-dependent A 2018 study showed that hyperglycaemia causes
by specialized proteins that production of IL-18 in the intestinal epithelium. Secreted increased intestinal permeability associated with
transport from the lumen
to the blood (absorption) or
IL-18 induces the expression of resistin-​like-β (RETNLB) decreased expression of the cell adhesion protein
from the blood to the lumen and ANG4 antimicrobial proteins to maintain a sym- E-​cadherin and tight junction protein ZO1 (ref.110).
(secretion). biotic gut flora. Interestingly, taurine had the opposite Conversely, dietary zinc enhances barrier function and

Nature Reviews | Nephrology


Reviews

increases the number of tight junctions in animal models alterations in the microbiota can change the metabolic
of colitis111. Moreover, zinc has also proved beneficial in and immunological profile of the host. These alter-
promoting barrier function in patients with diarrhoea ations can have consequences for several organ sys-
or Crohn’s disease112–114. Commensal microorganisms tems. Below, we discuss how the microbiota influences
and probiotics can also promote gut barrier function. kidney function and disease, focusing on two examples:
Microorganism-​derived SCFAs, particularly butyrate, hypertension and kidney stone disease.
promote barrier function and provide protection in
experimental and human colitis115–118; however, these The microbiota and hypertension
effects are likely mediated through multiple mecha- Microcirculatory observations in the 1970s demon-
nisms because (as mentioned earlier) SCFAs promote strated that the rate of vasomotor activity of precapil-
mucus secretion, regulate T cell development and induce lary arterioles was markedly decreased in germ-​free rats
inflammasome activation88–91,119,120. Finally, there is con- compared with that of conventional laboratory rats125.
siderable interest in developing probiotics to restore a Moreover, blood vessels in germ-​f ree rats were less
symbiotic enteric flora and treat autoinflammatory dis- responsive to the catecholamines adrenaline and
orders (reviewed elsewhere101,121). The administration noradrenaline than precapillary vessels of conventional
of immunomodulatory probiotics is likely to influence rats and responded poorly to vasopressin. A much more
host physiology pleiotropically; however, several stud- recent study further explored the relationship between
ies have demonstrated that the beneficial effects of dysbiosis and hypertension by analysing bacterial
probiotics might occur, at least in part, by promoting DNA isolated from faecal samples of two rat models of
barrier function122–124. Together, these studies show that hypertension and a small cohort of patients126, reveal-
ing that microbial richness, diversity and evenness were
a Dietary fibre Commensal Epithelial barrier
decreased in hypertensive rats and humans. A further
bacteria study127 showed that transplantation of faecal micro-
biota from stroke-​prone hypertensive rats to control
Wistar-​Kyoto rats was sufficient to induce increased
blood pressure in the normotensive rat. Targeted meta­
SCFAs GPCR bolomics profiling of SCFAs established the presence
activation
of elevated concentrations of acetate and heptanoate in
the plasma of normotensive rats transplanted with the
• Renin release caecal content of hypertensive rats, indicating that these
• Vasodilation or SCFAs might underlie the changes in blood pressure.
vasoconstriction These observations have been validated and extended
in an additional animal model of hypertension, which
demonstrated that a high-​fibre diet and acetate supple-
b Dietary oxalate Epithelial barrier mentation change the gut microbiota composition and
Cl– promote the development of hypertension128.
The mechanism by which SCFAs affect blood pressure
SLC26A6 Transcellular
transport
Crystal- is an area of active investigation (Fig. 3a). SCFAs that trav-
induced erse the intestinal epithelial barrier enter the bloodstream,
Oxalate NLRP3
activation where they activate GPCRs. Some of the effects of SCFAs
Paracellular might be mediated by effects on the kidney. Olfactory
transport
• Degrade receptor 78 (OLFR78; also known as OR51E2), for exam-
oxalate
• Regulate
• Cytokine ple, responds to acetate, propionate and partially to lac-
release
oxalate • Renal tate129,130 and is expressed in renal afferent arterioles and
transport failure on the smooth muscle cells of peripheral blood vessels9.
SCFAs activate OLFR78 in the afferent arteriole to release
Antibiotics renin; mice deficient in OLFR78 are hypotensive130.
By contrast, GPR41 (also known as free fatty acid receptor 3)
Fig. 3 | The gut–kidney axis in blood pressure regulation and kidney stone disease. and GPR43, which respond to propionate, acetate and
a | Dietary fibre that is digested in the colon generates short-​chain fatty acids (SCFAs), similar compounds87, are expressed in major blood ves-
which cross the epithelial barrier to activate G protein-​coupled receptors (GPCRs), in sels including renal arteries130 and lower blood pressure
the kidney and vasculature. Activation of olfactory receptor 78 (OLFR78) in the afferent upon binding of SCFAs, presumably via vasodilation131.
arteriole by SCFA binding induces the release of renin. Binding of SCFAs to other It is currently unclear why different GPCRs demonstrate
GPCRs, such as GPR41 and GPR43, in kidney and smooth muscle cells leads to either different responses to SCFAs with regard to blood pres-
vasoconstriction or vasodilation. b | Oxalate is also absorbed as a dietary component. sure control. It is also unclear why the same SCFA can
Some bacteria can degrade oxalate and/or increase the secretion of oxalate from exert different responses in different models. As an exam-
intestinal epithelia. Oxalate can also be back-​secreted by a transcellular route mediated
ple, elevated acetate levels were associated with increased
by SLC26A6, the activity of which can be regulated by bacteria such as Oxalobacter
formigenes. Antibiotic exposure could heighten the risk of kidney stones by inducing blood pressure in one animal model132, whereas acetate-​
perturbations in the microbiota that impair oxalate metabolism and transport. Excess producing bacteria tended to decrease blood pressure
absorption of dietary oxalate can lead to crystal formation in the kidney , NOD-, LRR- in a hypertension model126. One possible explanation
and pyrin domain-​containing 3 (NLRP3) inflammasome activation, inflammation and for these differences is that the receptors have different
kidney failure. affinities for SCFAs: GPR41 is activated in the presence

www.nature.com/nrneph
Reviews

of lower levels of SCFAs and drives vasodilation, whereas of available data. Studies to date have identified single
OLFR78, which has a higher effective concentration for instruments (that is, individual microorganisms) that
half-​maximum response (EC50) than GPR41, is activated produce a noise, yet we are far from understanding how
by higher levels of SCFAs to prevent overt hypotension133. those instruments interact with the orchestra as a whole.
The important interactions between diet, microbial In order to move the field forward, a number of ques-
composition and blood pressure were further demon- tions need to be answered, such as whether large-​scale
strated in a 2017 study that investigated the mechanisms metagenomics studies in humans can identify differ-
of salt-​induced hypertension. Administration of a high-​ ences in the composition of microbiota between control
salt diet to mice for 14 days induced hypertension, asso- individuals, patients with hypertension and those with
ciated with reduced levels of Lactobacillus murinus and resistant hypertension and whether novel therapeutic
increased numbers of pro-​inflammatory TH17 cells134. strategies, such as prebiotics and probiotics, have the
Similar findings were observed in healthy humans, potential to lower blood pressure in humans.
in whom a 14-day salt challenge led to an increase in
blood pressure and numbers of TH17 lymphocytes The microbiota and kidney stones
in peripheral blood and reduced intestinal survival Kidney stones are one of the most common conditions
of Lactobacillus spp. in individuals with Lactobacillus affecting the kidney, with a lifetime risk of nearly 1 in
strains at baseline. The inhibitory effects of salt on 5 men and 1 in 10 in women in the United States139.
Lactobacillus spp. are supported by in vitro data show- The prevalence has increased by 70% over the past
ing that increasing concentrations of sodium chloride three decades, with the greatest increase in incidence
specifically inhibit the growth of Lactobacillus isolates. observed in women and children for reasons that are
In addition to imbalances in microbiota composition poorly defined140–142. Kidney stones are not simply an iso-
and the production of microbial metabolites, hyper- lated nephrologic condition but rather a disorder with
tensive mice also exhibit epithelial barrier dysfunc- systemic complications, including an increased risk of
tion, as assessed by dextran permeability and reduced progressive loss of kidney function143 and cardiovascular
expression of occludins and claudins135. These changes disease144. During the past six decades, research into the
were also present in pre-​hypertensive mice, suggest- pathophysiology of kidney stones has focused mainly on
ing that decreases in gut tight junction proteins, and the physicochemistry of urinary crystal formation145,146
subsequently increased gut permeability, might cause and the biology of crystal retention147,148. Similar to hyper-
vascular stiffness and high blood pressure. Additional tension, no new drugs to treat or prevent the disease
structural changes that are evident in the epithelial bar- have entered our formulary for decades.
rier of hypertensive mice include reduced villi length Studies over the past few years, however, suggest
and reduced numbers of goblet cells135. Furthermore, an the existence of differences in the composition of the
increase in the number of vascular smooth muscle cells gut microbiota between stone-​formers and stone-​free
and in collagen deposition was noted in the intestinal controls149,150. A pilot study of 23 patients with neph-
wall of hypertensive rats136. These studies support the rolithiasis and 6 non-​stone-forming controls identified
concept that intestinal epithelial barrier dysfunction 178 bacteria at the genus level, with particular abundance
might contribute to the pathogenesis of hypertension. of Prevotella and Bacteroides150. In fact, Bacteroides spp.
In addition to contributing to the pathogenesis of were 3.4 times more abundant in stone-​formers than in
hypertension, gut microbial enzymes can also modify healthy controls, whereas Prevotella spp. were 2.8 times
the biotransformation of orally administered antihy- more abundant in non-​stone-formers. However, this
pertensives and thereby influence drug absorption study did not control for diet or the production of gut
and blood pressure control. For example, rats treated metabolites, two important factors that influence micro-
with amlodipine and antibiotics had higher systemic biome composition151. Moreover, only half of the patients
concentrations of amlodipine than rats treated with returned a 24 h urine collection, allowing limited corre-
amlodipine alone, suggesting an involvement of gut lation of urinary electrolytes and metabolites with bacte-
microbiota in the metabolism of this antihypertensive rial genera. Furthermore, the accuracy of metagenomic
agent137. Probiotics that contain Lactobacillus acidophi- analyses was confined to high taxonomic levels, and the
lus increase the expression of drug transporters, such differences observed in bacterial abundance did not
as multidrug resistance protein 1 (MDR1; also known allow any causative conclusion to be made regarding
as P-​g lycoprotein and ABCB1)138. MDR1 mediates the pathways of stone formation.
the efflux of xenobiotics from the intestinal mucosa A more extensive investigation attempted to link
into the lumen; its substrates include antihypertensive specific solutes that affect crystallization and renal
drugs such as diltiazem, verapamil and losartan. Hence, stone formation to gut microbiota composition and
increased expression of the MDR1 transporter as a con- functionality in 52 stone-​formers and 48 controls149.
sequence of L. acidophilus administration might affect Compared with healthy controls, patients with nephro-
the bioavailability of several antihypertensive agents. lithiasis had reduced diversity of faecal microbiota with
Thus, an accumulating body of evidence now sug- under-​representation of some taxa (Faecalibacterium,
gests that the microbiota have an important role in Enterobacter and Dorea). In addition, the microbiota of
Metagenomics regulating blood pressure. The microbial genome is stone-​formers had significantly lower levels of taxa that
Direct genetic analysis of entire
microbial communities to
modifiable and therefore offers a unique opportunity are predicted to be involved in dietary oxalate degrada-
provide information on for the development of new antihypertensive drugs. tion, and levels of these taxa correlated with 24 h uri-
microbial diversity. However, it is important to recognize the limitations nary oxalate excretion. Of note, dietary habits such as

Nature Reviews | Nephrology


Reviews

macronutrient and salt intake did not differ between the by promoting its elimination by the faeces), leading
two groups. Similar to the pilot study described above, to hyperoxalaemia, hyperoxaluria and chronic oxalate
this study is limited by its observational nature, which nephropathy170. As described earlier, NLRP3 inflammas-
does not enable causality to be established. Moreover, ome sensors control the ability of enteric microbiota to
the lifetime cumulative exposure of the study partic- restrict leakage of TLR agonists to the liver through the
ipants to antibiotics was not assessed — an important portal vein, thereby protecting against non-​alcoholic ste-
omission given that studies have described associations atohepatitis7. The same sensor is involved in recognizing
between antibiotic usage and diseases such as IBD152 and crystals, such as oxalate, uric acid and cysteine, in the
asthma153 that might be mediated by alterations of the kidney171–174. Mice with genetic171 or pharmacological175
human microbiome152. The association between antibi- inhibition of NLRP3 are protected from oxalate-​induced
otic use and nephrolithiasis risk was assessed in a 2018 nephropathy. These studies provide proof of principle
study that used conditional logistic regression models to that gut dietary solutes such as oxalate are relevant to
compare antibiotic use in 26,981 stone-​formers with that kidney stone formation via the activation of innate
of 259,797 matched non-​stone-forming controls154. The immune receptors. Moreover, these studies highlight the
investigators found that exposure to any of five different complex role of innate immune sensors such as NLRP3:
antibiotic classes was associated with an increased risk of in rodent models, their presence in the gastrointestinal
renal stone disease. The magnitude was greatest for indi- tract protects from liver disease, whereas in a model of
viduals who had been exposed to antibiotics at a young human disease their absence prevents crystal-​induced
age and for those who had received broad-​spectrum pen- kidney disease.
icillin (β-​lactam-containing antibiotics); the increased
risk of nephrolithiasis remained statistically significant Advancing from association to causality
3–5 years after exposure. The association between oral The above-​described studies indicate that the microbi-
antibiotic use and nephrolithiasis might explain the ome contributes to the pathogenesis of two of the most
increasing incidence of nephrolithiasis, particularly common renal disorders — a concept that is extremely
among children, and suggests that health-​care providers enticing. However, our understanding of these associa-
may be unintentionally contributing to the rising prev- tions is very preliminary, and several limitations of the
alence of stone disease by affecting the composition of methodological approaches used to date must be consid-
the microbiome beyond that needed to treat an infection. ered. Most studies have been performed by comparing
Again, the design of that study precluded any conclu- microbial populations in two groups of interest using
sions regarding the causality of the relationship; thus, microbiome-​wide association studies (MWAS); differ-
further studies are needed to examine whether reduced ences in the abundance of bacteria are then correlated
antibiotic treatment or the administration of specific with the phenotype of interest176, such as hypertension
probiotics after antibiotic use reduces kidney stone risk. or stone disease. A major challenge of this approach
In parallel to the above-​described clinical studies, is discerning whether the association of specific
progress has been made in our understanding of the microorganisms with a disease represents causation.
mechanisms leading to stone disease. Oxalate is the pre- A new triangulation-​based approach to assess micro-
dominant constituent of stones in 75% of patients with organism–phenotype relationships has been presented as
nephrolithiasis155 (Fig. 3b). Several studies have shown a way of identifying microorganisms that cause or con-
that the gut microbiota can affect oxalate homeosta- tribute pathologically to models of disease (Fig. 4a). This
sis156–160. Certain bacterial strains, including Oxalobacter approach has been used to identify a bacterial species
spp., Bifidobacterium spp. and Lactobacillus spp., can that protects mice from dextran-​sulfate-sodium-​induced
degrade oxalate159,161. Moreover, intestinal bacteria might colitis. The researchers who developed this approach
not only alter oxalate metabolism but also increase oxa- first used an MWAS-​based approach to compare bac-
late secretion from intestinal epithelia162–165. Oxalic acid, terial strains in gnotobiotic mice with colitis177 that had
which is absorbed from dietary sources, is absorbed pas- been colonized either with mouse microbiota (MMb) or
sively in the intestine via paracellular transport across with microbiota from healthy human donors (HMb)13,
tight junctions owing to its low molecular mass and identifying hundreds of differentially expressed bacte-
size166. Transcellular oxalate secretion via the oxalate rial strains. Co-​housing of these mice led to the gen-
transporter SLC26A6 mediates back-​flux of oxalate into eration of mice with a hybrid microbiota composition
the intestine to limit its net absorption166. Mice that are and intermediate susceptibility to colitis. By compar-
deficient in SLC26A6 develop hyperoxalaemia, hyper- ing the microbial communities of these three groups
oxaluria and calcium–oxalate stones owing to a defect of mice (MMb, HMb and hybrid) and screening for
in this intestinal back-​flux of ingested oxalate167,168. The MWAS-​identified taxa that correlated with the disease
importance of the microbiome to this process is demon- phenotype, the researchers discovered a single taxon,
strated by studies showing that conditioned media from Lachnospiraceae, that modified the disease of interest.
cultured Oxalobacter formigenes stimulates enteric Future studies will need to confirm the value of this new
oxalate secretion via SLC26A6 (refs162,169), providing method, particularly as it is based on the assumption
evidence that bacteria or their metabolites modulate that co-​housing will lead to an intermediate phenotype.
epithelial oxalate transport. Similarly, oxalate can pass However, as in the case of NLRP6 described earlier, one
the intestinal epithelial barrier in the absence of dietary microbiome may completely dictate the phenotype of
calcium (which normally complexes with oxalic acid in a disease, prevent an intermediate phenotype and not
the intestinal lumen and thereby limits its absorption allow microorganism–phenotype triangulation64,178.

www.nature.com/nrneph
Reviews

a b to the identification of bacteria that contribute to the


Gut pathogenesis of IgA nephropathy.
microbiota Translational studies in humans are, however, criti-
Intermediate cal for advancing our understanding of the microbiome
disease IgA nephropathy in disease. Findings obtained in isogenic rodents under
highly controlled conditions might not translate to simi-
Cell sorting
lar findings in humans. Two meta-​analyses of controlled
clinical trials examining the effect of probiotic fermented
Causal IgA negative IgA positive
microbes
milk or probiotics on hypertension showed only a very
modest decrease or no change in diastolic blood pressure
16S rRNA gene sequencing in response to treatment186,187. In the above-​described
study in which dietary supplementation of sodium led to
an increase in blood pressure and reduced levels of faecal
Identification of specific bacteria
for functional classification L. murinus in 14 healthy male volunteers134, it is question-
Healthy Severe disease
able whether the increased amount of ingested salt would
Fig. 4 | Approaches for assessing the causality of microbiome constituents in kidney markedly affect the amount of NaCl presented to the
disease. a | Microbiome–phenotype triangulation identifies causal bacteria not only by colon owing to NaCl secretion occurring under physi­
comparing differences in microbial species between two mouse populations of interest
ological conditions in this segment (which is required
but also by including a co-​housing group of mice with an intermediate phenotype. By
performing three-​way comparisons, the number of bacterial species of interest can be
for the absorption of digested amino acids and sugars);
refined and specific species can be tested for disease causality. b | Identification of larger studies are needed to assess whether increased die-
pathogenic microorganisms could be achieved for diseases such as immunoglobulin A tary salt intake can affect levels of faecal L. murinus or
(IgA) nephropathy by combining bacterial cell sorting (to identify bacteria coated with whether its supplementation can reduce blood pressure.
IgA) and 16S ribosomal RNA (rRNA) sequencing to identify specific microbiota that are With regard to nephrolithiasis, preclinical studies in
coated with IgA for functional classification. Both of the above approaches enable the mouse models of hyperoxaluria have shown that coloni-
identification of specific bacteria that may be involved in disease processes. Part a is zation with oxalate-​metabolizing bacteria, such as O. for-
adapted with permission from ref.178, Elsevier. migenes, increases epithelial oxalate secretion and reduces
urinary oxalate excretion162,188, suggesting that these bac-
Identification of causative bacteria in the context of teria may be a powerful tool to treat affected patients.
kidney disease — particularly in terms of defining ‘good’ Despite the promising preliminary data, however, results
versus ‘bad’ bacteria — remains a technical challenge. from clinical trials have failed to support the use of oral
Kidney disease itself may affect the colonic microenvi- O. formigenes therapy in patients with primary hyperox-
ronment and composition, making it difficult to assess aluria189. A formulation containing a variety of bacteria
cause–effect relationships179. Omics approaches, such as (L. acidophilus, Lactobacillus brevis, Streptococcus ther-
shotgun metagenomics and metaproteomics176,180, might mophiles and Bifidobacterium longum subsp. infantis) had
aid in the identification of causative bacteria. A new no effect on urinary oxalate excretion in patients with
method involving ‘targeted’ functional classification can hyperoxaluric kidney stones190,191. Therefore, although the
be used to assign functions to taxa without relying on gut microbiome probably affects blood pressure control
prior functional predictions55,181 (Fig. 4b). The approach and the risk of kidney stones, no microbiome-​modifying
uses a combination of fluorescence-​activated cell sort- interventions have yet been identified that substantially
ing (FACS) and 16S ribosomal RNA (rRNA) gene modify these diseases in humans.
sequencing to identify bacteria with antigen-​specific
immune responses and was successfully used to iden- Conclusions
tify disease-​causing microbiota in humans with IBD55. A body of basic research supports a role for the micro-
Such an approach could theoretically be used to iden- biota in a variety of renal disorders. In addition, clinical
tify pathogenic microorganisms that contribute to IgA studies have identified an association between vari-
nephropathy — one of the most common causes of glo- ous commensals and common renal diseases, includ-
merulonephritis in the developed world182. Although the ing hypertension and nephrolithiasis. Key remaining
aetiology of IgA nephropathy remains unclear, it seems to challenges include the identification of causal patho-
have an autoimmune component that involves dysregula- gens, our ability to translate findings obtained in ani-
tion of mucosal-​type IgA immune responses183. Findings mal models successfully to patients and our ability to
from genome-​wide association studies (GWAS) suggest develop microbiome-​modifying treatment approaches
a role for host–intestinal pathogen interactions in the to treat renal disorders. New methodologies are likely to
development and progression of the disease184, a concept help in addressing these challenges and advancing the
that is supported by the finding that a targeted release field of microbiome research. Importantly, our ability to
formulation of budesonide, designed to target intestinal decipher the diversity of commensals living in the gas-
mucosal immunity, successfully reduced proteinuria in trointestinal tract and understand their role in diseases
patients with IgA nephropathy185. As described earlier, requires a multidisciplinary approach; therefore, one of
IgA is secreted into the intestinal lumen, where it binds our goals must be to integrate the expertise of special-
to and coats bacteria with higher affinity for pathogenic ists, including immunologists, experts in sequencing
Metaproteomics
Characterization of all the
microorganisms than for commensal microorganisms. technology and nephrologists, to advance the field.
protein samples recovered Thus, use of methods such as targeted functional clas-
from environmental sources. sification to identify IgA-​associated bacteria might lead Published online xx xx xxxx

Nature Reviews | Nephrology


Reviews

1. Levin, A. et al. Global kidney health 2017 and beyond: composition and diversity. Allergy Asthma Clin. 54. Shulzhenko, N. et al. Crosstalk between
a roadmap for closing gaps in care, research, and Immunol. 9, 15 (2013). B lymphocytes, microbiota and the intestinal
policy. Lancet 390, 1888–1917 (2017). 28. Hasegawa, K. et al. Household siblings and nasal and epithelium governs immunity versus metabolism
2. Thompson, S. et al. Cause of death in patients with fecal microbiota in infants. Pediatr. Int. 59, 473–481 in the gut. Nat. Med. 17, 1585–1593 (2011).
reduced kidney function. J. Am. Soc. Nephrol. 26, (2017). 55. Palm, N. W. et al. Immunoglobulin A coating identifies
2504–2511 (2015). 29. Shukla, S. K. et al. The nasal microbiota of dairy colitogenic bacteria in inflammatory bowel disease.
3. Jha, V. et al. Understanding kidney care needs and farmers is more complex than oral microbiota, Cell 158, 1000–1010 (2014).
implementation strategies in low- and middle-​income reflects occupational exposure, and provides 56. Slack, E., Balmer, M. L., Fritz, J. H. & Hapfelmeier, S.
countries: conclusions from a “Kidney Disease: competition for staphylococci. PLOS ONE 12, Functional flexibility of intestinal IgA - broadening the
Improving Global Outcomes” (KDIGO) Controversies e0183898 (2017). fine line. Front. Immunol. 3, 100 (2012).
Conference. Kidney Int. 90, 1164–1174 (2016). 30. Fall, T. et al. Early exposure to dogs and farm animals 57. Pabst, O. New concepts in the generation and
4. Abboud, H. & Henrich, W. L. Clinical practice. Stage IV and the risk of childhood asthma. JAMA Pediatr. 169, functions of IgA. Nat. Rev. Immunol. 12, 821–832
chronic kidney disease. N. Engl. J. Med. 362, 56–65 e153219 (2015). (2012).
(2010). 31. Strachan, D. P. Hay fever, hygiene, and household size. 58. Mantis, N. J. & Forbes, S. J. Secretory IgA: arresting
5. Forslund, K. et al. Disentangling type 2 diabetes and BMJ 299, 1259–1260 (1989). microbial pathogens at epithelial borders. Immunol.
metformin treatment signatures in the human gut 32. David, L. A. et al. Diet rapidly and reproducibly alters Invest. 39, 383–406 (2010).
microbiota. Nature 528, 262–266 (2015). the human gut microbiome. Nature 505, 559–563 59. Lycke, N., Erlandsson, L., Ekman, L., Schon, K. &
6. Turnbaugh, P. J. et al. An obesity-​associated gut (2014). Leanderson, T. Lack of J chain inhibits the transport
microbiome with increased capacity for energy 33. Sonnenburg, E. D. et al. Diet-​induced extinctions of gut IgA and abrogates the development of intestinal
harvest. Nature 444, 1027–1031 (2006). in the gut microbiota compound over generations. antitoxic protection. J. Immunol. 163, 913–919
7. Henao-​Mejia, J. et al. Inflammasome-​mediated Nature 529, 212–215 (2016). (1999).
dysbiosis regulates progression of NAFLD and obesity. 34. Suez, J. et al. Artificial sweeteners induce glucose 60. Forbes, S. J., Eschmann, M. & Mantis, N. J.
Nature 482, 179–185 (2012). intolerance by altering the gut microbiota. Nature Inhibition of Salmonella enterica serovar
8. Dejea, C. M. et al. Patients with familial adenomatous 514, 181–186 (2014). typhimurium motility and entry into epithelial cells
polyposis harbor colonic biofilms containing 35. Chassaing, B. et al. Dietary emulsifiers impact the by a protective antilipopolysaccharide monoclonal
tumorigenic bacteria. Science 359, 592–597 (2018). mouse gut microbiota promoting colitis and metabolic immunoglobulin A antibody. Infect. Immun. 76,
9. Zheng, P. et al. Gut microbiome remodeling induces syndrome. Nature 519, 92–96 (2015). 4137–4144 (2008).
depressive-​like behaviors through a pathway mediated 36. Gum, J. R. et al. Molecular cloning of human intestinal 61. Vijay-​Kumar, M. et al. Deletion of TLR5 results in
by the host’s metabolism. Mol. Psychiatry 21, mucin (MUC2) cDNA. Identification of the amino spontaneous colitis in mice. J. Clin. Invest. 117,
786–796 (2016). terminus and overall sequence similarity to prepro-​von 3909–3921 (2007).
10. The Integrative HMP (iHMP) Research Network Willebrand factor. J. Biol. Chem. 269, 2440–2446 62. Vijay-​Kumar, M. et al. Metabolic syndrome and altered
Consortium. The Integrative Human Microbiome (1994). gut microbiota in mice lacking Toll-​like receptor 5.
Project: dynamic analysis of microbiome-​host omics 37. Atuma, C., Strugala, V., Allen, A. & Holm, L. The Science 328, 228–231 (2010).
profiles during periods of human health and disease. adherent gastrointestinal mucus gel layer: thickness 63. Singh, V. et al. Microbiota-​dependent hepatic
Cell Host Microbe 16, 276–289 (2014). and physical state in vivo. Am. J. Physiol. Gastrointest. lipogenesis mediated by stearoyl CoA desaturase 1
11. Kuntz, T. M. & Gilbert, J. A. Introducing the Liver Physiol. 280, G922–G929 (2001). (SCD1) promotes metabolic syndrome in TLR5-
microbiome into precision medicine. Trends 38. Johansson, M. E. et al. The inner of the two Muc2 deficient mice. Cell. Metab. 22, 983–996 (2015).
Pharmacol. Sci. 38, 81–91 (2017). mucin-​dependent mucus layers in colon is devoid 64. Elinav, E. et al. NLRP6 inflammasome regulates
12. Zou, J. et al. Fiber-​mediated nourishment of gut of bacteria. Proc. Natl Acad. Sci. USA 105, colonic microbial ecology and risk for colitis. Cell 145,
microbiota protects against diet-​induced obesity by 15064–15069 (2008). 745–757 (2011).
restoring IL-22-mediated colonic health. Cell Host 39. Kim, J. J. & Khan, W. I. Goblet cells and mucins: role 65. Seregin, S. S. et al. NLRP6 protects Il10(−/−) mice
Microbe 23, 41–53 (2018). in innate defense in enteric infections. Pathogens 2, from colitis by limiting colonization of akkermansia
13. Chung, H. et al. Gut immune maturation depends on 55–70 (2013). muciniphila. Cell Rep. 19, 733–745 (2017).
colonization with a host-​specific microbiota. Cell 149, 40. Birchenough, G. M., Nystrom, E. E., Johansson, M. E. 66. Levy, M. et al. Microbiota-​modulated metabolites
1578–1593 (2012). & Hansson, G. C. A sentinel goblet cell guards the shape the intestinal microenvironment by regulating
14. Mao, K. et al. Innate and adaptive lymphocytes colonic crypt by triggering Nlrp6-dependent Muc2 NLRP6 inflammasome signaling. Cell 163,
sequentially shape the gut microbiota and lipid secretion. Science 352, 1535–1542 (2016). 1428–1443 (2015).
metabolism. Nature 554, 255–259 (2018). 41. Wlodarska, M. et al. NLRP6 inflammasome 67. Galvez, E. J. C., Iljazovic, A., Gronow, A., Flavell, R.
15. Moghadamrad, S. et al. Attenuated portal orchestrates the colonic host-​microbial interface by & Strowig, T. Shaping of intestinal microbiota in Nlrp6-
hypertension in germ-​free mice: Function of bacterial regulating goblet cell mucus secretion. Cell 156, and Rag2-deficient mice depends on community
flora on the development of mesenteric lymphatic 1045–1059 (2014). structure. Cell Rep. 21, 3914–3926 (2017).
and blood vessels. Hepatology 61, 1685–1695 42. Nowarski, R. et al. Epithelial IL-18 equilibrium controls 68. Ubeda, C. et al. Familial transmission rather than
(2015). barrier function in colitis. Cell 163, 1444–1456 defective innate immunity shapes the distinct
16. Geuking, M. B. et al. Intestinal bacterial colonization (2015). intestinal microbiota of TLR-​deficient mice. J. Exp.
induces mutualistic regulatory T cell responses. 43. Ragland, S. A. & Criss, A. K. From bacterial killing to Med. 209, 1445–1456 (2012).
Immunity 34, 794–806 (2011). immune modulation: recent insights into the functions 69. Lemire, P. et al. The NLR protein NLRP6 does not
17. Hapfelmeier, S. et al. Reversible microbial colonization of lysozyme. PLOS Pathog. 13, e1006512 (2017). impact gut microbiota composition. Cell Rep. 21,
of germ-​free mice reveals the dynamics of IgA immune 44. Cash, H. L., Whitham, C. V., Behrendt, C. L. 3653–3661 (2017).
responses. Science 328, 1705–1709 (2010). & Hooper, L. V. Symbiotic bacteria direct expression 70. Mamantopoulos, M. et al. Nlrp6- and ASC-​dependent
18. Haber, A. L. et al. A single-​cell survey of the small of an intestinal bactericidal lectin. Science 313, inflammasomes do not shape the commensal gut
intestinal epithelium. Nature 551, 333–339 (2017). 1126–1130 (2006). microbiota composition. Immunity 47, 339–348
19. Wiedermann, C. J. et al. Association of endotoxemia 45. Qu, X. D. & Lehrer, R. I. Secretory phospholipase A2 (2017).
with carotid atherosclerosis and cardiovascular is the principal bactericide for staphylococci and other 71. Wostmann, B. S., Larkin, C., Moriarty, A. & Bruckner-​
disease: prospective results from the Bruneck study. gram-​positive bacteria in human tears. Infect. Immun. Kardoss, E. Dietary intake, energy metabolism, and
J. Am. Coll. Cardiol. 34, 1975–1981 (1999). 66, 2791–2797 (1998). excretory losses of adult male germfree Wistar rats.
20. McIntyre, C. W. et al. Circulating endotoxemia: a novel 46. Koduri, R. S. et al. Bactericidal properties of human Lab. Anim. Sci. 33, 46–50 (1983).
factor in systemic inflammation and cardiovascular and murine groups I, II, V, X, and XII secreted 72. Stecher, B. et al. Comparison of Salmonella enterica
disease in chronic kidney disease. Clin. J. Am. Soc. phospholipases A2. J. Biol. Chem. 277, 5849–5857 serovar Typhimurium colitis in germfree mice and mice
Nephrol. 6, 133–141 (2011). (2002). pretreated with streptomycin. Infect. Immun. 73,
21. Poesen, R. et al. Associations of soluble CD14 and 47. Holly, M. K. & Smith, J. G. Paneth cells during viral 3228–3241 (2005).
endotoxin with mortality, cardiovascular disease, and infection and pathogenesis. Viruses 10, 225 (2018). 73. Gordon, H. A. Morphological and physiological
progression of kidney disease among patients with 48. Hooper, L. V., Stappenbeck, T. S., Hong, C. V. & characterization of germfree life. Ann. NY Acad. Sci.
CKD. Clin. J. Am. Soc. Nephrol. 10, 1525–1533 Gordon, J. I. Angiogenins: a new class of microbicidal 78, 208–220 (1959).
(2015). proteins involved in innate immunity. Nat. Immunol. 4, 74. Reinhardt, C. et al. Tissue factor and PAR1 promote
22. Wang, Z. et al. Gut flora metabolism of 269–273 (2003). microbiota-​induced intestinal vascular remodelling.
phosphatidylcholine promotes cardiovascular disease. 49. Rumio, C. et al. Degranulation of paneth cells via toll-​ Nature 483, 627–631 (2012).
Nature 472, 57–63 (2011). like receptor 9. Am. J. Pathol. 165, 373–381 (2004). 75. Surana, N. K. & Kasper, D. L. Deciphering the tete-​
23. Sender, R., Fuchs, S. & Milo, R. Revised estimates for 50. Foureau, D. M. et al. TLR9-dependent induction of a-tete between the microbiota and the immune
the number of human and bacteria cells in the body. intestinal alpha-​defensins by Toxoplasma gondii. system. J. Clin. Invest. 124, 4197–4203 (2014).
PLOS Biol. 14, e1002533 (2016). J. Immunol. 184, 7022–7029 (2010). 76. Mazmanian, S. K., Liu, C. H., Tzianabos, A. O.
24. Gill, S. R. et al. Metagenomic analysis of the human 51. Selsted, M. E. & Ouellette, A. J. Mammalian defensins & Kasper, D. L. An immunomodulatory molecule of
distal gut microbiome. Science 312, 1355–1359 in the antimicrobial immune response. Nat. Immunol. symbiotic bacteria directs maturation of the host
(2006). 6, 551–557 (2005). immune system. Cell 122, 107–118 (2005).
25. Koenig, J. E. et al. Succession of microbial consortia 52. Carvalho, F. A., Aitken, J. D., Vijay-​Kumar, M. & 77. Ochoa-​Reparaz, J. et al. Central nervous system
in the developing infant gut microbiome. Proc. Natl Gewirtz, A. T. Toll-​like receptor-​gut microbiota demyelinating disease protection by the human
Acad. Sci. USA 108 (Suppl. 1), 4578–4585 (2011). interactions: perturb at your own risk! Annu. Rev. commensal Bacteroides fragilis depends on
26. Stappenbeck, T. S. & Virgin, H. W. Accounting for Physiol. 74, 177–198 (2012). polysaccharide A expression. J. Immunol. 185,
reciprocal host-​microbiome interactions in 53. Vaishnava, S., Behrendt, C. L., Ismail, A. S., Eckmann, L. 4101–4108 (2010).
experimental science. Nature 534, 191–199 (2016). & Hooper, L. V. Paneth cells directly sense gut 78. Ochoa-​Reparaz, J. et al. A polysaccharide from the
27. Azad, M. B. et al. Infant gut microbiota and the commensals and maintain homeostasis at the human commensal Bacteroides fragilis protects
hygiene hypothesis of allergic disease: impact intestinal host-​microbial interface. Proc. Natl Acad. against CNS demyelinating disease. Mucosal Immunol.
of household pets and siblings on microbiota Sci. USA 105, 20858–20863 (2008). 3, 487–495 (2010).

www.nature.com/nrneph
Reviews

79. Round, J. L. et al. The Toll-​like receptor 2 pathway Time course, reversibility, and site of cytokine binding. failure in hypertensive mice. Circulation 135,
establishes colonization by a commensal of the human J. Immunol. 150, 2356–2363 (1993). 964–977 (2017).
microbiota. Science 332, 974–977 (2011). 105. Al-​Sadi, R., Ye, D., Dokladny, K. & Ma, T. Y. Mechanism 129. Chang, A. J., Ortega, F. E., Riegler, J., Madison, D. V.
80. Mazmanian, S. K., Round, J. L. & Kasper, D. L. A of IL-1beta-​induced increase in intestinal epithelial & Krasnow, M. A. Oxygen regulation of breathing
microbial symbiosis factor prevents intestinal tight junction permeability. J. Immunol. 180, through an olfactory receptor activated by lactate.
inflammatory disease. Nature 453, 620–625 (2008). 5653–5661 (2008). Nature 527, 240–244 (2015).
81. Shen, Y. et al. Outer membrane vesicles of a human 106. Howe, K. L., Reardon, C., Wang, A., Nazli, A. 130. Pluznick, J. L. et al. Olfactory receptor responding
commensal mediate immune regulation and disease & McKay, D. M. Transforming growth factor-​beta to gut microbiota-​derived signals plays a role in renin
protection. Cell Host Microbe 12, 509–520 (2012). regulation of epithelial tight junction proteins secretion and blood pressure regulation. Proc. Natl
82. Dasgupta, S., Erturk-​Hasdemir, D., Ochoa-​Reparaz, J., enhances barrier function and blocks enterohemorrhagic Acad. Sci. USA 110, 4410–4415 (2013).
Reinecker, H. C. & Kasper, D. L. Plasmacytoid Escherichia coli O157:H7-induced increased 131. Nutting, C. W., Islam, S., Ye, M. H., Batlle, D. C. &
dendritic cells mediate anti-​inflammatory responses permeability. Am. J. Pathol. 167, 1587–1597 Daugirdas, J. T. The vasorelaxant effects of acetate:
to a gut commensal molecule via both innate and (2005). role of adenosine, glycolysis, lyotropism, and pHi and
adaptive mechanisms. Cell Host Microbe 15, 107. Suenaert, P. et al. Anti-​tumor necrosis factor Cai2+. Kidney Int. 41, 166–174 (1992).
413–423 (2014). treatment restores the gut barrier in Crohn’s disease. 132. Mell, B. et al. Evidence for a link between gut
83. An, D. et al. Sphingolipids from a symbiotic microbe Am. J. Gastroenterol. 97, 2000–2004 (2002). microbiota and hypertension in the Dahl rat.
regulate homeostasis of host intestinal natural killer 108. Suenaert, P. et al. Hyperresponsiveness of the mucosal Physiol. Genom. 47, 187–197 (2015).
T cells. Cell 156, 123–133 (2014). barrier in Crohn’s disease is not tumor necrosis factor-​ 133. Pluznick, J. L. Gut microbiota in renal physiology:
84. Hoverstad, T. & Midtvedt, T. Short-​chain fatty acids in dependent. Inflamm. Bowel Dis. 11, 667–673 (2005). focus on short-​chain fatty acids and their receptors.
germfree mice and rats. J. Nutr. 116, 1772–1776 109. Martinez-​Medina, M. et al. Western diet induces Kidney Int. 90, 1191–1198 (2016).
(1986). dysbiosis with increased E coli in CEABAC10 mice, 134. Wilck, N. et al. Salt-​responsive gut commensal
85. Maslowski, K. M. et al. Regulation of inflammatory alters host barrier function favouring AIEC modulates TH17 axis and disease. Nature 551,
responses by gut microbiota and chemoattractant colonisation. Gut 63, 116–124 (2014). 585–589 (2017).
receptor GPR43. Nature 461, 1282–1286 (2009). 110. Thaiss, C. A. et al. Hyperglycemia drives intestinal 135. Santisteban, M. M. et al. Hypertension-​linked
86. Tan, J. K., McKenzie, C., Marino, E., Macia, L. & barrier dysfunction and risk for enteric infection. pathophysiological alterations in the gut. Circ. Res.
Mackay, C. R. Metabolite-​sensing G protein-​coupled Science 359, 1376–1383 (2018). 120, 312–323 (2017).
receptors-​facilitators of diet-​related immune 111. Sturniolo, G. C. et al. Effect of zinc supplementation on 136. Stewart, D. C. et al. Hypertension-​linked mechanical
regulation. Annu. Rev. Immunol. 35, 371–402 (2017). intestinal permeability in experimental colitis. J. Lab. changes of rat gut. Acta Biomater. 45, 296–302
87. Le Poul, E. et al. Functional characterization of human Clin. Med. 139, 311–315 (2002). (2016).
receptors for short chain fatty acids and their role in 112. Roy, S. K. et al. Impact of zinc supplementation on 137. Yoo, H. H., Kim, I. S., Yoo, D. H. & Kim, D. H. Effects of
polymorphonuclear cell activation. J. Biol. Chem. 278, intestinal permeability in Bangladeshi children with orally administered antibiotics on the bioavailability
25481–25489 (2003). acute diarrhoea and persistent diarrhoea syndrome. of amlodipine: gut microbiota-​mediated drug
88. Macia, L. et al. Metabolite-​sensing receptors GPR43 J. Pediatr. Gastroenterol. Nutr. 15, 289–296 (1992). interaction. J. Hypertens. 34, 156–162 (2016).
and GPR109A facilitate dietary fibre-​induced gut 113. Sturniolo, G. C., Di Leo, V., Ferronato, A., D’Odorico, A. 138. Saksena, S. et al. Upregulation of P-​glycoprotein by
homeostasis through regulation of the inflammasome. & D’Inca, R. Zinc supplementation tightens “leaky gut” probiotics in intestinal epithelial cells and in the
Nat. Commun. 6, 6734 (2015). in Crohn’s disease. Inflamm. Bowel Dis. 7, 94–98 dextran sulfate sodium model of colitis in mice.
89. Furusawa, Y. et al. Commensal microbe-​derived (2001). Am. J. Physiol. Gastrointest. Liver Physiol. 300,
butyrate induces the differentiation of colonic 114. Alam, A. N., Sarker, S. A., Wahed, M. A., Khatun, M. G1115–G1123 (2011).
regulatory T cells. Nature 504, 446–450 (2013). & Rahaman, M. M. Enteric protein loss and intestinal 139. Scales, C. D. et al. Prevalence of kidney stones in the
90. Arpaia, N. et al. Metabolites produced by commensal permeability changes in children during acute United States. Eur. Urol. 62, 160–165 (2012).
bacteria promote peripheral regulatory T cell shigellosis and after recovery: effect of zinc 140. Romero, V., Akpinar, H. & Assimos, D. G. Kidney
generation. Nature 504, 451–455 (2013). supplementation. Gut 35, 1707–1711 (1994). stones: a global picture of prevalence, incidence,
91. Park, J. et al. Short-​chain fatty acids induce both 115. Venkatraman, A., Ramakrishna, B. S., Pulimood, A. B., and associated risk factors. Rev. Urol. 12, e86–96
effector and regulatory T cells by suppression of Patra, S. & Murthy, S. Increased permeability in (2010).
histone deacetylases and regulation of the mTOR-​S6K dextran sulphate colitis in rats: time course of 141. Alexander, R. T. et al. Kidney stones and
pathway. Mucosal Immunol. 8, 80–93 (2015). development and effect of butyrate. Scand. J. cardiovascular events: a cohort study. Clin. J. Am.
92. Hori, S., Nomura, T. & Sakaguchi, S. Control of Gastroenterol. 35, 1053–1059 (2000). Soc. Nephrol. 9, 506–512 (2014).
regulatory T cell development by the transcription 116. Mariadason, J. M., Barkla, D. H. & Gibson, P. R. Effect 142. Kittanamongkolchai, W. et al. The changing incidence
factor Foxp3. Science 299, 1057–1061 (2003). of short-​chain fatty acids on paracellular permeability and presentation of urinary stones over 3 decades.
93. Flannigan, K. L. & Denning, T. L. Segmented in Caco-2 intestinal epithelium model. Am. J. Physiol. Mayo Clin. Proc. 93, 291–299 (2018).
filamentous bacteria-​induced immune responses: 272, G705–G712 (1997). 143. Zhe, M. & Hang, Z. Nephrolithiasis as a risk factor
a balancing act between host protection and 117. Peng, L., He, Z., Chen, W., Holzman, I. R. & Lin, J. of chronic kidney disease: a meta-​analysis of cohort
autoimmunity. Immunology 154, 537–546 (2018). Effects of butyrate on intestinal barrier function in a studies with 4,770,691 participants. Urolithiasis 45,
94. Atarashi, K. et al. Th17 cell induction by adhesion Caco-2 cell monolayer model of intestinal barrier. 441–448 (2016).
of microbes to intestinal epithelial cells. Cell 163, Pediatr. Res. 61, 37–41 (2007). 144. Ferraro, P. M. et al. History of kidney stones and the
367–380 (2015). 118. Vernia, P. et al. Topical butyrate improves efficacy of risk of coronary heart disease. JAMA 310, 408–415
95. Gaboriau-​Routhiau, V. et al. The key role of segmented 5-ASA in refractory distal ulcerative colitis: results of a (2013).
filamentous bacteria in the coordinated maturation of multicentre trial. Eur. J. Clin. Invest. 33, 244–248 145. Asplin, J. R., Parks, J. H. & Coe, F. L. Dependence
gut helper T cell responses. Immunity 31, 677–689 (2003). of upper limit of metastability on supersaturation in
(2009). 119. Finnie, I. A., Dwarakanath, A. D., Taylor, B. A. & nephrolithiasis. Kidney Int. 52, 1602–1608 (1997).
96. Ivanov, I. I. et al. Induction of intestinal Th17 cells by Rhodes, J. M. Colonic mucin synthesis is increased 146. Bergsland, K. J., Zisman, A. L., Asplin, J. R.,
segmented filamentous bacteria. Cell 139, 485–498 by sodium butyrate. Gut 36, 93–99 (1995). Worcester, E. M. & Coe, F. L. Evidence for net renal
(2009). 120. Barcelo, A. et al. Mucin secretion is modulated by tubule oxalate secretion in patients with calcium
97. Ivanov, I. I. et al. Specific microbiota direct the luminal factors in the isolated vascularly perfused rat kidney stones. Am. J. Physiol. Renal Physiol. 300,
differentiation of IL-17-producing T-​helper cells in the colon. Gut 46, 218–224 (2000). F311–F318 (2011).
mucosa of the small intestine. Cell Host Microbe 4, 121. Ulluwishewa, D. et al. Regulation of tight junction 147. Coe, F. L., Worcester, E. M. & Evan, A. P. Idiopathic
337–349 (2008). permeability by intestinal bacteria and dietary hypercalciuria and formation of calcium renal stones.
98. Lei, Z. et al. EpCAM contributes to formation of components. J. Nutr. 141, 769–776 (2011). Nat. Rev. Nephrol. 12, 519–533 (2016).
functional tight junction in the intestinal epithelium by 122. Patel, R. M. et al. Probiotic bacteria induce 148. Coe, F. L., Evan, A. P., Lingeman, J. E. & Worcester, E. M.
recruiting claudin proteins. Dev. Biol. 371, 136–145 maturation of intestinal claudin 3 expression and Plaque and deposits in nine human stone diseases.
(2012). barrier function. Am. J. Pathol. 180, 626–635 Urol. Res. 38, 239–247 (2010).
99. Laukoetter, M. G. et al. JAM-​A regulates permeability (2012). 149. Ticinesi, A. et al. Understanding the gut-​kidney axis
and inflammation in the intestine in vivo. J. Exp. Med. 123. Garcia Vilela, E. et al. Influence of Saccharomyces in nephrolithiasis: an analysis of the gut microbiota
204, 3067–3076 (2007). boulardii on the intestinal permeability of patients composition and functionality of stone formers. Gut
100. Tanaka, H. et al. Intestinal deletion of Claudin-7 with Crohn’s disease in remission. Scand. J. 67, 2097–2106 (2018).
enhances paracellular organic solute flux and initiates Gastroenterol. 43, 842–848 (2008). 150. Stern, J. M. et al. Evidence for a distinct gut
colonic inflammation in mice. Gut 64, 1529–1538 124. Zakostelska, Z. et al. Lysate of probiotic Lactobacillus microbiome in kidney stone formers compared to
(2015). casei DN-114 001 ameliorates colitis by non-stone formers. Urolithiasis 44, 399–407 (2016).
101. Michielan, A. & D’Inca, R. Intestinal permeability in strengthening the gut barrier function and changing 151. Wu, G. D. et al. Linking long-​term dietary patterns
inflammatory bowel disease: pathogenesis, clinical the gut microenvironment. PLOS ONE 6, e27961 with gut microbial enterotypes. Science 334,
evaluation, and therapy of leaky gut. Mediators (2011). 105–108 (2011).
Inflamm. 2015, 628157 (2015). 125. Baez, S. & Gordon, H. A. Tone and reactivity of 152. Kronman, M. P., Zaoutis, T. E., Haynes, K., Feng, R. &
102. Zeissig, S. et al. Changes in expression and vascular smooth muscle in germfree rat mesentery. Coffin, S. E. Antibiotic exposure and IBD development
distribution of claudin 2, 5 and 8 lead to J. Exp. Med. 134, 846–856 (1971). among children: a population-​based cohort study.
discontinuous tight junctions and barrier dysfunction 126. Yang, T. et al. Gut dysbiosis is linked to hypertension. Pediatrics 130, e794–e803 (2012).
in active Crohn’s disease. Gut 56, 61–72 (2007). Hypertension 65, 1331–1340 (2015). 153. Mitre, E. et al. Association between use of acid-​
103. Ma, T. Y. et al. TNF-​alpha-induced increase in intestinal 127. Adnan, S. et al. Alterations in the gut microbiota can suppressive medications and antibiotics during infancy
epithelial tight junction permeability requires NF-​ elicit hypertension in rats. Physiol. Genom. 49, and allergic diseases in early childhood. JAMA Pediatr.
kappa B activation. Am. J. Physiol. Gastrointest. Liver 96–104 (2017). 172, e180315 (2018).
Physiol. 286, G367–G376 (2004). 128. Marques, F. Z. et al. High-​fiber diet and acetate 154. Tasian, G. E. et al. Oral antibiotic exposure and kidney
104. Adams, R. B., Planchon, S. M. & Roche, J. K. IFN-​ supplementation change the gut microbiota and stone disease. J. Am. Soc. Nephrol. 29, 1731–1740
gamma modulation of epithelial barrier function. prevent the development of hypertension and heart (2018).

Nature Reviews | Nephrology


Reviews

155. Pfau, A. & Knauf, F. Update on nephrolithiasis: intestinal epithelial cells. J. Am. Soc. Nephrol. 28, controlled phase 2b trial. Lancet 389, 2117–2127
core curriculum 2016. Am. J. Kidney Dis. 68, 876–887 (2017). (2017).
973–985 (2016). 170. Mulay, S. R. et al. Oxalate-​induced chronic kidney 186. Dong, J. Y. et al. Effect of probiotic fermented milk on
156. Hatch, M. Gut microbiota and oxalate homeostasis. disease with its uremic and cardiovascular blood pressure: a meta-​analysis of randomised
Ann. Transl Med. 5, 36 (2017). complications in C57BL/6 mice. Am. J. Physiol. Renal controlled trials. Br. J. Nutr. 110, 1188–1194 (2013).
157. Knight, J., Deora, R., Assimos, D. G. & Holmes, R. P. Physiol. 310, F785–F795 (2016). 187. Khalesi, S., Sun, J., Buys, N. & Jayasinghe, R. Effect of
The genetic composition of Oxalobacter formigenes 171. Knauf, F. et al. NALP3-mediated inflammation is probiotics on blood pressure: a systematic review and
and its relationship to colonization and calcium a principal cause of progressive renal failure in meta-​analysis of randomized, controlled trials.
oxalate stone disease. Urolithiasis 41, 187–196 oxalate nephropathy. Kidney Int. 84, 895–901 Hypertension 64, 897–903 (2014).
(2013). (2013). 188. Hatch, M., Gjymishka, A., Salido, E. C., Allison, M. J. &
158. Al-​Wahsh, I., Wu, Y. & Liebman, M. Acute probiotic 172. Mulay, S. R. et al. Calcium oxalate crystals induce Freel, R. W. Enteric oxalate elimination is induced and
ingestion reduces gastrointestinal oxalate absorption renal inflammation by NLRP3-mediated IL-1beta oxalate is normalized in a mouse model of primary
in healthy subjects. Urol. Res. 40, 191–196 (2012). secretion. J. Clin. Invest. 123, 236–246 (2013). hyperoxaluria following intestinal colonization with
159. Turroni, S. et al. Oxalate-​degrading activity in 173. Kim, S. M. et al. Hyperuricemia-​induced NLRP3 Oxalobacter. Am. J. Physiol. Gastrointest. Liver
Bifidobacterium animalis subsp. lactis: impact of activation of macrophages contributes to the Physiol. 300, G461–G469 (2011).
acidic conditions on the transcriptional levels of the progression of diabetic nephropathy. Am. J. Physiol. 189. Milliner, D., Hoppe, B. & Groothoff, J. A randomised
oxalyl coenzyme A (CoA) decarboxylase and formyl-​ Renal Physiol. 308, F993–F1003 (2015). Phase II/III study to evaluate the efficacy and safety of
CoA transferase genes. Appl. Environ. Microbiol. 76, 174. Prencipe, G. et al. Inflammasome activation by cystine orally administered Oxalobacter formigenes to treat
5609–5620 (2010). crystals: implications for the pathogenesis of primary hyperoxaluria. Urolithiasis 46, 313–323
160. Mehta, M., Goldfarb, D. S. & Nazzal, L. The role of the cystinosis. J. Am. Soc. Nephrol. 25, 1163–1169 (2018).
microbiome in kidney stone formation. Int. J. Surg. (2014). 190. Goldfarb, D. S., Modersitzki, F. & Asplin, J. R. A
36, 607–612 (2016). 175. Ludwig-​Portugall, I. et al. An NLRP3-specific randomized, controlled trial of lactic acid bacteria for
161. Klimesova, K., Whittamore, J. M. & Hatch, M. inflammasome inhibitor attenuates crystal-​induced idiopathic hyperoxaluria. Clin. J. Am. Soc. Nephrol. 2,
Bifidobacterium animalis subsp. lactis decreases kidney fibrosis in mice. Kidney Int. 90, 525–539 745–749 (2007).
urinary oxalate excretion in a mouse model of primary (2016). 191. Lieske, J. C. et al. Diet, but not oral probiotics,
hyperoxaluria. Urolithiasis 43, 107–117 (2015). 176. Gilbert, J. A. et al. Microbiome-​wide association effectively reduces urinary oxalate excretion and
162. Hatch, M. et al. Oxalobacter sp. reduces urinary studies link dynamic microbial consortia to disease. calcium oxalate supersaturation. Kidney Int. 78,
oxalate excretion by promoting enteric oxalate Nature 535, 94–103 (2016). 1178–1185 (2010).
secretion. Kidney Int. 69, 691–698 (2006). 177. Surana, N. K. & Kasper, D. L. Moving beyond
163. Sidhu, H. et al. Direct correlation between microbiome-​wide associations to causal microbe Acknowledgements
hyperoxaluria/oxalate stone disease and the absence identification. Nature 552, 244–247 (2017). F.K. is supported by the Deutsche Forschungsgemeinschaft
of the gastrointestinal tract-​dwelling bacterium 178. Rosen, C. E. & Palm, N. W. Navigating the microbiota (DFG; project KN 1148/2-1 and CRC 1365 Renoprotection),
Oxalobacter formigenes: possible prevention by gut seas: triangulation finds a way forward. Cell Host the Oxalosis and Hyperoxaluria Foundation and TRENAL, a
recolonization or enzyme replacement therapy. J. Am. Microbe 23, 1–3 (2018). thematic network grant of the Deutscher Akademischer
Soc. Nephrol. 10 (Suppl. 14), 334–340 (1999). 179. Poesen, R. et al. The influence of CKD on colonic Austauschdienst (DAAD). J.R.B. is supported by an American
164. Hatch, M. & Freel, R. W. A human strain of microbial metabolism. J. Am. Soc. Nephrol. 27, Cancer Society Postdoctoral Fellowship (PF-17-237-01-DMC).
Oxalobacter (HC-1) promotes enteric oxalate 1389–1399 (2016). R.A.F. is supported by the Howard Hughes Medical Institute.
secretion in the small intestine of mice and reduces 180. Mondot, S. & Lepage, P. The human gut microbiome
urinary oxalate excretion. Urolithiasis 41, 379–384 and its dysfunctions through the meta-​omics prism. Author contributions
(2013). Ann. NY Acad. Sci. 1372, 9–19 (2016). F.K. and J.R.B. researched data for the article and wrote the
165. Hatch, M., Freel, R. W. & Vaziri, N. D. Regulatory 181. Rosen, C. E. & Palm, N. W. Functional classification of article. All authors contributed substantially to discussion of
aspects of oxalate secretion in enteric oxalate the gut microbiota: the key to cracking the microbiota the article’s content and reviewed and edited the manuscript
elimination. J. Am. Soc. Nephrol. 10 (Suppl. 14), composition code: functional classifications of the gut before submission.
324–328 (1999). microbiota reveal previously hidden contributions of
166. Knauf, F. et al. Net intestinal transport of oxalate indigenous gut bacteria to human health and disease. Competing interests
reflects passive absorption and SLC26A6-mediated Bioessays 39, 1700032 (2017). R.A.F. is a scientific consultant to GlaxoSmithKline and Zie
secretion. J. Am. Soc. Nephrol. 22, 2247–2255 182. Wyatt, R. J. & Julian, B. A. IgA nephropathy. N. Engl. Labs and a founder, shareholder and advisor to SMOC
(2011). J. Med. 368, 2402–2414 (2013). Therapeutics Inc. The other authors declare no competing
167. Jiang, Z. et al. Calcium oxalate urolithiasis in mice 183. Barratt, J., Smith, A. C., Molyneux, K. & Feehally, J. interests.
lacking anion transporter Slc26a6. Nat. Genet. 38, Immunopathogenesis of IgAN. Semin. Immunopathol.
474–478 (2006). 29, 427–443 (2007). Publisher’s note
168. Freel, R. W., Hatch, M., Green, M. & Soleimani, M. 184. Kiryluk, K. et al. Discovery of new risk loci for IgA Springer Nature remains neutral with regard to jurisdictional
Ileal oxalate absorption and urinary oxalate excretion nephropathy implicates genes involved in immunity claims in published maps and institutional affiliations.
are enhanced in Slc26a6 null mice. Am. J. Physiol. against intestinal pathogens. Nat. Genet. 46,
Gastrointest. Liver Physiol. 290, G719–G728 1187–1196 (2014). Reviewer information
(2006). 185. Fellstrom, B. C. et al. Targeted-​release budesonide Nature Reviews Nephrology thanks C. M. Higueras, C. Kurts,
169. Arvans, D. et al. Oxalobacter formigenes-​derived versus placebo in patients with IgA nephropathy L. Nazzal and other anonymous reviewer(s) for their contribution
bioactive factors stimulate oxalate transport by (NEFIGAN): a double-​blind, randomised, placebo-​ to the peer review of this work.

www.nature.com/nrneph

You might also like