throbber
The Gut Microbiota
`
`REVIEW
`
`Interactions Between the Microbiota
`and the Immune System
`
`Lora V. Hooper,1* Dan R. Littman,2 Andrew J. Macpherson3
`
`The large numbers of microorganisms that inhabit mammalian body surfaces have a highly coevolved
`relationship with the immune system. Although many of these microbes carry out functions that are
`critical for host physiology, they nevertheless pose the threat of breach with ensuing pathologies.
`The mammalian immune system plays an essential role in maintaining homeostasis with resident
`microbial communities, thus ensuring that the mutualistic nature of the host-microbial relationship
`is maintained. At the same time, resident bacteria profoundly shape mammalian immunity. Here, we
`review advances in our understanding of the interactions between resident microbes and the immune
`system and the implications of these findings for human health.
`
`Complex communities of microorganisms,
`
`termed the “microbiota,” inhabit the body
`surfaces of virtually all vertebrates. In the
`lower intestine, these organisms reach extraordi-
`nary densities and have evolved to degrade a
`variety of plant polysaccharides and other dietary
`substances (1). This simultaneously enhances host
`digestive efficiency and ensures a steady nutrient
`supply for the microbes. Metabolic efficiency
`was likely a potent selective force that shaped the
`evolution of both sides of the host-microbiota
`relationship. Millions of years of coevolution,
`however, have forged pervasive interconnections
`between the physiologies of microbial commu-
`nities and their hosts that extend beyond metabolic
`functions. These interconnections are particularly
`apparent in the relationship between the microbiota
`and the immune system.
`Despite the symbiotic nature of the intestinal
`host-microbial relationship, the close association
`of an abundant bacterial community with intesti-
`nal tissues poses immense health challenges. The
`dense communities of bacteria in the lower intes-
`tine (≥1012/cm3 intestinal contents) are separated
`from body tissues by the epithelial layer (10 mm)
`over a large intestinal surface area (~200 m2 in
`humans). Opportunistic invasion of host tissue by
`resident bacteria has serious health consequences,
`including inflammation and sepsis. The immune
`system has thus evolved adaptations that work to-
`gether to contain the microbiota and preserve the
`symbiotic relationship between host and microbiota.
`The evolution of the vertebrate immune system has
`therefore been driven by the need to protect the
`
`1The Howard Hughes Medical Institute and Department of Im-
`munology, The University of Texas Southwestern Medical Center
`at Dallas, Dallas, TX 75390, USA. 2Howard Hughes Medical
`Institute and Molecular Pathogenesis Program, The Kimmel
`Center for Biology and Medicine of the Skirball Institute, New
`York University School of Medicine, New York, NY 10016, USA.
`3Maurice Müller Laboratories, University Clinic for Visceral Sur-
`gery and Medicine, University of Bern, Bern, Switzerland.
`*To whom correspondence should be addressed. E-mail:
`lora.hooper@utsouthwestern.edu
`
`host from pathogens and to foster complex micro-
`bial communities for their metabolic benefits (2).
`In this Review, we survey the state of our
`understanding of microbiota-immune system in-
`teractions. We also highlight key experimental
`challenges that must be confronted to advance
`our understanding in this area and consider how
`our knowledge of these interactions might be
`harnessed to improve public health.
`
`Tools for Analyzing the Microbiota–Immune
`System Relationship
`Much of our current understanding of microbiota–
`immune system interactions has been acquired
`from studies of germ-free animals. Such animals
`are reared in sterile isolators to control their
`exposure to microorganisms, including viruses,
`bacteria, and eukaryotic parasites. Germ-free
`animals can be studied in their microbiologically
`sterile state or can serve as living test tubes for the
`establishment of simplified microbial ecosystems
`composed of a single microbial species or defined
`species mixtures. The technology has thus come
`to be known as “gnotobiotics,” a term derived from
`Greek meaning “known life.” Gnotobiotic ani-
`mals, particularly rodents, have become critical
`experimental tools for determining which host
`immune functions are genetically encoded and
`which require interactions with microbes.
`The current impetus for gnotobiotic exper-
`imentation has been driven by several impor-
`tant technical advances. First, because any mouse
`strain can be derived to germ-free status (3), large
`numbers of genetically targeted and wild-type
`inbred isogenic mouse strains have become avail-
`able in the germ-free state. The contribution of
`different immune system constituents to host-
`microbial mutualism can thus be determined by
`comparing the effects of microbial colonization
`in genetically altered and wild-type mice (4, 5).
`Second, next-generation sequencing tech-
`nologies have opened the black box of micro-
`biota complexity. Although advances in ex vivo
`culturability are still needed, the composition of
`
`human and animal microbiotas can be opera-
`tionally defined from polymorphisms of bacterial
`genes, especially those encoding the 16S ribo-
`somal RNA sequences. Such analyses have made
`possible the construction of defined microbiotas,
`whose distinct effects on host immunity can now
`be examined (6). Moreover, these advances allow
`the study of experimental animals that are both
`isobiotic and, in a defined inbred host, isogenic.
`A dominant goal of these efforts is to benefit hu-
`man health [see Blumberg and Powie (7)]. With
`the developing technology, the species differ-
`ences can be closed using mice with a defined
`humanized microbiota (8). On the horizon, there
`is even the prospect of humanized isobiotic mice
`that also have a humanized immune system (9).
`A third advance has been the development of
`experimental systems that allow the uncoupling
`of commensal effects on the immune system from
`microbial colonization. This cannot be achieved
`by antibiotic treatment alone because a small pro-
`portion of the targeted microbes will persist.
`Deletion strains of bacteria lacking the ability to
`synthesize prokaryotic-specific amino acids have
`been developed that can be grown in culture but do
`not persist in vivo, so the animals become germ-
`free again. This allows issues of mucosal immune
`induction, memory, and functional protection to
`be explored without permanent colonization (10).
`Finally, important insights about the impact of
`resident microbial communities on mammalian
`host biology have been acquired by using high-
`throughput transcriptomic and metabolomic tools
`to compare germ-free and colonized mice (11, 12).
`These tools include DNA microarrays, which have
`led to a detailed understanding of how microbiota
`shape many aspects of host physiology, includ-
`ing immunity (13, 14) and development (15), as
`well as mass spectrometry and nuclear magnetic
`resonance spectroscopy, which have provided im-
`portant insights into how microbiota influence
`metabolic signaling in mammalian hosts (12). The
`application of these new approaches to the older
`technology of gnotobiotics has revolutionized
`the study of interactions between the microbiota
`and the immune system.
`
`Looking Inside-Out: Immune System
`Control of the Microbiota
`A major driving force in the evolution of the
`mammalian immune system has been the need
`to maintain homeostatic relationships with the
`microbiota. This encompasses control of micro-
`bial interactions with host tissues as well as the
`composition of microbial consortia. Here, we dis-
`cuss recent insights into how the immune system
`exerts “inside-out” control over microbiota local-
`ization and community composition (see Fig. 1).
`Stratification and compartmentalization of the
`microbiota. The intestinal immune system faces
`unique challenges relative to other organs, as it
`must continuously confront an enormous micro-
`bial load. At the same time, it is necessary to avoid
`
`1268
`
`8 JUNE 2012 VOL 336 SCIENCE www.sciencemag.org
`
`Genome Ex. 1035
`Page 1 of 6
`
`

`

`pathologies arising from innate immune signaling
`or from microbiota alterations that disturb essential
`metabolic functions. An important function of the
`intestinal immune system is to control the expo-
`sure of bacteria to host tissues, thereby lessening
`the potential for pathologic outcomes. This oc-
`curs at two distinct levels: first, by minimizing
`direct contact between intestinal bacteria and the
`epithelial cell surface (stratification) and, second,
`by confining penetrant bacteria to intestinal sites
`and limiting their exposure to the systemic im-
`mune compartment (compartmentalization).
`Several immune effectors function together to
`stratify luminal microbes and to minimize bacterial-
`epithelial contact. Intestinal goblet cells secrete
`mucin glycoproteins that assemble into a ~150-mm-
`thick viscous coating at the intestinal epithelial
`cell surface. In the colon, there are two structurally
`distinct mucus layers. Although the outer mucus
`layer contains large numbers of bacteria, the inner
`mucus layer is resistant to bacterial penetration
`(16). In contrast, the small intestine lacks clearly
`distinct inner and outer mucus layers (17). Here,
`compartmentalization depends in part on antibac-
`terial proteins that are secreted by the intestinal
`epithelium. RegIIIg is an antibacterial lectin that
`is expressed in epithelial cells under the control of
`Toll-like receptors (TLRs) (18–20). RegIIIg limits
`bacterial penetration of the small intestinal mucus
`layer, thus restricting the number of bacteria that
`contact the epithelial surface (5).
`Stratification of intestinal bacteria on the
`luminal side of the epithelial barrier also depends
`on secreted immunoglobulin A (IgA). IgA spe-
`cific for intestinal bacteria is produced with the
`help of intestinal dendritic cells that sample the
`small numbers of bacteria that penetrate the over-
`lying epithelium. These bacteria-laden dendritic
`cells interact with B and T cells in the Peyer’s
`patches, inducing B cells to produce IgA directed
`against intestinal bacteria (21). IgA+ B cells home
`to the intestinal lamina propria and secrete IgA
`that is transcytosed across the epithelium and
`deposited on the apical surface. The transcytosed
`IgAs bind to luminal bacteria, preventing micro-
`bial translocation across the epithelial barrier (22).
`Mucosal compartmentalization functions to
`minimize exposure of resident bacteria to the sys-
`temic immune system (Fig. 1B). Although bacteria
`are largely confined to the luminal side of the
`epithelial barrier, the sheer number of intestinal
`bacteria makes an occasional breach inevita-
`ble. Typically, commensal microorganisms that
`penetrate the intestinal epithelial cell barrier are
`phagocytosed and eliminated by lamina propria
`macrophages (23). However, the intestinal im-
`mune system samples some of the penetrant bac-
`teria, engendering specific immune responses
`that are distributed along the length of the intes-
`tine (21). Bacteria that penetrate the intestinal
`barrier are engulfed by dendritic cells (DCs) re-
`siding in the lamina propria and are carried alive
`to the mesenteric lymph nodes. However, these
`
`SPECIALSECTION
`
`bacteria do not penetrate to systemic secondary
`lymphoid tissues. Rather, the commensal-bearing
`DCs induce protective secretory IgAs (21), which
`are distributed throughout all mucosal surfaces
`by recirculation of activated B and T cells. Thus,
`distinctive anatomical adaptations in the mucosal
`immune system allow immune responses directed
`against commensals to be distributed widely while
`still being confined to mucosal tissues.
`Other immune cell populations also promote
`the containment of commensal bacteria to in-
`testinal sites. Innate lymphoid cells reside in the
`lamina propria and have effector cytokine pro-
`
`files resembling those of T helper (TH) cells (24).
`Innate lymphoid cells that produce interleukin
`(IL)–22 are essential for containment of lymphoid-
`resident bacteria to the intestine, thus preventing
`their spread to systemic sites (25).
`The compartmentalization of mucosal and
`systemic immune priming can be severely per-
`turbed in immune-deficient mice. For example,
`mice engineered to lack IgA show priming of
`serum IgG responses against commensals, indi-
`cating that these bacteria have been exposed to
`the systemic immune system (22). A similar out-
`come is observed when innate immune sensing is
`
`Fig. 1. Looking inside-out: immune system control of the microbiota. Several immune effectors function
`together to stratify luminal microbes and to minimize bacterial-epithelial contact. This includes the mucus
`layer, epithelial antibacterial proteins, and IgA secreted by lamina propria plasma cells. Compartmen-
`talization is accomplished by unique anatomic adaptations that limit commensal bacterial exposure to the
`immune system. Some microbes are sampled by intestinal DCs. The loaded DCs traffic to the mesenteric
`lymph nodes through the intestinal lymphatics but do not penetrate further into the body. This
`compartmentalizes live bacteria and induction of immune responses to the mucosal immune system.
`There is recirculation of induced B cells and some T cell subsets through the lymphatics and the
`bloodstream to home back to mucosal sites, where B cells differentiate into IgA-secreting plasma cells.
`
`www.sciencemag.org SCIENCE VOL 336
`
`8 JUNE 2012
`
`1269
`
`Genome Ex. 1035
`Page 2 of 6
`
`

`

`The Gut Microbiota
`
`defective. Mice lacking MyD88 or TRIF signal-
`ing adaptors for TLR-mediated sensing of bacteria
`also produce serum IgG responses against com-
`mensals (26). This probably results from the fact
`that in these settings, large numbers of commensals
`cross the epithelial barrier and phagocytic cells
`are less able to eliminate the penetrant organisms.
`Immune system control of microbiota com-
`position. The development of high-throughput
`sequencing technologies for microbiota analysis
`has provided insight into the many factors that
`determine microbiota composition. For example
`nutrients, whether derived from the host diet
`(27) or from endogenous host sources (28), are
`critically important in shaping the structure of
`host-associated microbial communities. Recent
`evidence suggests that the immune system is also
`likely to be an important contributor to “inside-
`out” host control over microbiota composition.
`Certain secreted antibacterial proteins produced
`by epithelial cells can shape the composition of in-
`testinal microbial communities. a-defensins are
`small (2 to 3 kD) antibacterial peptides secreted by
`Paneth cells of the small intestinal epithelium. Anal-
`ysis of the microbiota in mice that were either de-
`ficient in functional a-defensins or that overexpressed
`human a-defensin-5 showed that although there
`was no impact on total numbers of colonizing bacte-
`ria, there were substantial a-defensin–dependent
`changes in community composition, with reciprocal
`differences observed in the two mouse strains (29).
`An interesting question is how far secreted in-
`nate immune effectors “reach” into the luminal
`microbial consortia. For example, the impact of hu-
`man a-defensin-5 on luminal community composi-
`tion contrasts with the antibacterial lectin RegIIIg,
`which limits penetration of bacteria to the epithelial
`surface but does not alter luminal communities (5).
`This suggests that some antimicrobial proteins, such
`as a-defensins, reach into the lumen to shape overall
`community composition, whereas others, such as
`RegIIIg, have restricted effects on surface-associated
`bacteria and thus control microbiota location relative
`to host surface tissues. Questions remain as to ex-
`actly how a-defensin-5 controls luminal community
`composition, however. In one scenario, these small
`antimicrobial peptides diffuse through the mucus
`layer and directly act on bacteria that inhabit the lu-
`men. Another possibility is that a-defensin-5 exerts
`its antibacterial activity on bacteria that are trapped in
`the outer reaches of the mucus layer, with those bac-
`teria acting as reservoirs that seed luminal commu-
`nities and thus dictate their composition. Answering
`these questions will require improved tools for fine-
`mapping microbiota composition and consortia from
`the surface of the intestine to the interior of the lumen.
`The impact of the immune system on micro-
`biota composition is also suggested by several im-
`mune deficiencies that alter microbial communities
`in ways that predispose to disease. For example,
`Garrett et al. studied mice that lack the transcription
`factor T-bet (encoded by Tbx21), which governs
`inflammatory responses in cells of both the innate
`
`and the adaptive immune system (30). When
`Tbx21–/– mice were crossed onto Rag2–/– mice,
`which lack adaptive immunity, the Tbx21–/–/Rag2–/–
`progeny developed ulcerative colitis in a microbiota-
`dependent manner (30). Remarkably, this colitis
`phenotype was transmissible to wild-type mice by
`adoptive transfer of the Tbx21–/–/Rag2–/– micro-
`biota. This demonstrated that altered microbiota
`were sufficient to induce disease and could thus be
`considered “dysbiotic.” Similarly, mice lacking the
`bacterial flagellin receptor TLR5 exhibit a syn-
`drome encompassing insulin resistance, hyper-
`lipidemia, and increased fat deposition associated
`with alterations in microbiota composition (31).
`These metabolic changes are transferable to wild-
`type mice that acquire the Tlr5–/– gut microbiota.
`A third example of immune-driven dysbiosis is
`seen in mice deficient for epithelial cell expres-
`sion of the inflammasome component NLRP6.
`These mice develop an altered microbiota with
`increased abundance of members of the Bacte-
`roidetes phylum associated with increased intes-
`tinal inflammatory cell recruitment and susceptibility
`to chemically induced colitis. Again, there is evi-
`dence that dysbiosis alone is sufficient to drive the
`intestinal inflammation, because conventionally
`raised wild-type mice that acquire the dysbiotic
`microbiota show similar immunopathology (32).
`Together, these findings suggest that the im-
`mune system affords mammalian hosts some con-
`trol over the composition of their resident microbial
`communities. It is also clear that these commu-
`nities can be perturbed by defects in the host im-
`mune system. This leads to the idea of the immune
`system as a form of ecosystem management that
`exerts critical control over microbiota compo-
`sition, diversity, and location [see Costello et al.
`(33)]. However, a number of questions remain.
`First, although it is apparent that the immune sys-
`tem shapes community composition at the species
`level, it is not yet clear whether the immune sys-
`tem shapes the genetics and physiology of indi-
`vidual microbial species. Second, how much does
`the immune system combine with gastric acid and
`intestinal motility to control the longitudinal dis-
`tribution of microbial species in the gastrointes-
`tinal tract? Finally, it will be important to determine
`the extent to which the immune system also con-
`trols microbial community composition and loca-
`tion in other organ systems, such as the respiratory
`tract, urogenital tract, and skin.
`
`Looking Outside-In: How Microbiota
`Shape Immunity
`The earliest comparisons of germ-free and colonized
`mice revealed a profound effect of microbial colo-
`nization on the formation of lymphoid tissues and
`subsequent immune system development. It was
`thus quickly apparent that the microbiota influ-
`ence the immune system from “outside-in.” Recent
`studies have greatly amplified this understanding
`and have revealed some of the cellular and mo-
`lecular mediators of these interactions (see Fig. 2).
`
`The impact of the microbiota on lymphoid
`structure development and epithelial function.
`The tissues of the gastrointestinal tract are rich in
`myeloid and lymphoid cells, many of which
`reside in organized lymphoid tissues. It has long
`been appreciated that the gut microbiota have a
`critical role in the development of organized lym-
`phoid structures and in the function of immune
`system cells. For example, isolated lymphoid fol-
`licles in the small intestine do not develop in
`germ-free mice, and such mice are also deficient
`in secretory IgA and CD8ab intraepithelial lym-
`phocytes. The specific microbial molecules en-
`dowed with this inductive function have not yet
`been described, however.
`Sensing of commensal microbiota through the
`TLR-MyD88 signaling pathway triggers several
`responses that are critical for maintaining host-
`microbial homeostasis. The microbiota induce
`repair of damaged intestinal epithelium through a
`MyD88-dependent process that can be rescued in
`microbe-depleted animals by gavage with bacterial
`lipopolysaccharide (LPS). The innate signals, con-
`veyed largely through myeloid cells, are required to
`enhance epithelial cell proliferation (34, 35). As
`discussed above, MyD88-dependent bacterial sig-
`nals are also required for the induction of epithelial
`antimicrobial proteins such as RegIIIg (5, 19). This
`expression can be induced by LPS (19, 20 ) or flagel-
`lin (36). The flagellin signals are relayed through
`TLR5 expressed by CD103+CD11b+ dendritic cells
`in the lamina propria, stimulating production of IL-
`23 that, in turn, promotes the expression of IL-22
`by innate lymphoid cells (37). IL-22 then stimu-
`lates production of RegIIIg, which is also secreted
`upon direct activation of MyD88 in epithelial
`cells (5, 20). This is one clear example of the
`importance of commensals in the induction of host
`innate responses, but it likely represents a tiny
`fraction of the multitude of effects of microbiota on
`the host immune system.
`Microbiota shaping of T cell subsets. It has
`recently become evident that individual commensal
`species influence the makeup of lamina propria T
`lymphocyte subsets that have distinct effector func-
`tions. Homeostasis in the gut mucosa is maintained
`by a system of checks and balances between poten-
`tially proinflammatory cells, which include TH1 cells
`that produce interferon-g; TH17 cells that produce
`IL-17a, IL-17f, and IL-22; diverse innate lymphoid
`cells with cytokine effector features resembling
`TH2 and TH17 cells; and anti-inflammatory Foxp3+
`regulatory T cells (Tregs). Colonization of mice with
`segmented filamentous bacteria (SFB) results in
`accumulation of TH17 cells and, to a lesser extent, in
`an increase in TH1 cells (38, 39). SFB appear able to
`penetrate the mucus layer overlying the intestinal
`epithelial cells in the terminal ileum, and they in-
`teract closely with the epithelial cells, inducing host
`cell actin polymerization at the site of interaction
`and, presumably, signaling events that result in a
`TH17 polarizing environment within the lamina
`propria. There is little known about host cell
`
`1270
`
`8 JUNE 2012 VOL 336 SCIENCE www.sciencemag.org
`
`Genome Ex. 1035
`Page 3 of 6
`
`

`

`signaling pathways initiated by SFB. It is possible
`that SFB influence epithelial gene expression, re-
`sulting, for example, in expression of antimicro-
`bial proteins such as RegIIIg and of molecules
`that participate in TH17 cell polarization. SFB
`may also act directly on cells of the immune sys-
`tem, either through interactions with myeloid cells
`that extend processes through the epithelium to
`the mucus layer or by production of metabolites
`that act on various receptors expressed by host cells.
`Other bacteria have been shown to enhance the
`anti-inflammatory branches of the adaptive immune
`system by directing the differentiation of Tregs or by
`inducing IL-10 expression. For example, coloniza-
`tion of gnotobiotic mice with a complex cocktail of
`46 mouse Clostridial strains, originally isolated from
`mouse feces and belonging mainly to cluster IVand
`XIVa of the Clostridium genus, results in the
`expansion of lamina propria and systemic Tregs.
`These have a phenotype characteristic of Tregs in-
`duced in the periphery in response to transforming
`growth factor (TGF)–b and retinoic acid [in contrast
`to thymic-derived natural (n) Tregs (40)], and many
`
`of these inducible Tregs (iTregs) express IL-10. The
`exact Clostridial strains within the complex exper-
`imental mixture that drive this regulatory response
`remain to be defined. Furthermore, polysaccharide
`A (PSA) of Bacteroides fragilis induces an IL-10
`response in intestinal T cells, which prevents the
`expansion of TH17 cells and potential damage to the
`mucosal barrier (41). In contrast, mutant B. fragilis
`lacking PSA has a proinflammatory profile and fails
`to induce IL-10. Production of PSA by B. fragilis
`has been proposed to be instrumental for the bac-
`terium’s success as a commensal.
`Within the intestine, the balance of effector lym-
`phoid cells and Treg cells can have a profound in-
`fluence on how the mucosa responds to stresses that
`elicit damage. The relative roles of commensal-
`regulated T cells differ according to the models used
`to study inflammation. For example, in mice sub-
`jected to chemical or pathogen-induced damage to
`the mucosa, TH17 cells have a beneficial effect that
`promotes healing. In contrast, TH1 and TH17 cells,
`as well as IL-23–dependent innate lymphoid cells,
`promote colitis in models in which Treg cells are
`
`Fig. 2. Looking outside-in: how microbiota shape host immunity. Some of the many ways that intestinal
`microbiota shape host immunity are depicted. These include microbiota effects on mucosal as well as
`systemic immunity. ILFs, isolated lymphoid follicles.
`
`SPECIALSECTION
`
`depleted. It is likely that inflammatory bowel dis-
`eases in humans can be similarly triggered by
`commensal-influenced imbalance of lymphoid cell
`subsets. This is supported by numerous observations,
`including the strong linkage of IL23R polymor-
`phisms with Crohn’s disease, a serious condition
`with relapsing intestinal inflammation and a risk of
`malignancy, and the severe enterocolitis associated
`with IL10 and IL10R mutations (42, 43).
`Microbiota effects on systemic immunity. The
`influence of commensal bacteria on the balance of
`T cell subsets is now known to extend well beyond
`the intestinal lamina propria. Homeostatic T cell
`proliferation itself is driven by the microbiota or
`their penetrant molecules (44). Systemic auto-
`immune diseases have long been suggested to have
`links to infections, but firm evidence for causality
`has been lacking. Recent studies in animal models,
`however, have reinforced the notion that commen-
`sal microbiota contribute to systemic autoimmune
`and allergic diseases at sites distal to the intestinal
`mucosa. Several mouse models for autoimmunity
`are dependent on colonization status. Thus, germ-
`free mice have marked attenuation of disease in
`models of arthritis and experimental autoimmune
`encephalomyelitis (EAE), as well as in various
`colitis models. In models of TH17 cell–dependent
`arthritis and EAE, monoassociation with SFB is
`sufficient to induce disease (42, 45, 46). In all of
`these models, induction of TH17 cells in the in-
`testine has a profound influence on systemic dis-
`ease. Exacerbation of arthritis and EAE is likely the
`consequence of an increase in the number of
`arthritogenic or encephalitogenic TH17 cells that
`traffic out of the lamina propria. The antigen spec-
`ificity of such cells remains to be examined.
`Induction of iTregs by the cluster IV and XIVa
`Clostridia also has a systemic effect on inflamma-
`tory processes. Colonization of germ-free mice with
`these bacteria not only results in attenuated disease
`after chemical damage of the gut epithelium but
`also reduces the serum IgE response after immuni-
`zation with antigen under conditions that favor a
`TH2 response (40). As with pathogenic TH17 cells,
`the antigen specificity of the commensal-induced
`iTregs that execute systemic anti-inflammatory func-
`tions is not yet known, although at least some of the
`Tregs in the gut have T cell receptors with specificity
`for distinct commensal bacteria (47).
`Finally, B. fragilis PSA affects the develop-
`ment of systemic T cell responses. Colonization of
`germ-free mice with PSA-producing B. fragilis
`results in higher numbers of circulating CD4+ T
`cells compared to mice colonized with B. fragilis
`lacking PSA. PSA-producing B.
`fragilis also
`elicits higher TH1 cell frequencies in the circulation
`(48). Together, these findings show that commen-
`sal bacteria have a general impact on immunity
`that reaches well beyond mucosal tissues.
`Microbiota influences on invariant T cells and
`innate lymphoid cells. A recent study extends the
`role of microbiota to the control of the function
`invariant natural killer T cells (iNKT cells), which
`
`www.sciencemag.org SCIENCE VOL 336
`
`8 JUNE 2012
`
`1271
`
`Genome Ex. 1035
`Page 4 of 6
`
`

`

`The Gut Microbiota
`
`bear an invariant T cell receptor specific for lipid
`antigens presented by the atypical class I mol-
`ecule CD1d. Germ-free mice were found to have
`increased susceptibility to iNKT cell–mediated
`oxazolone-induced colitis and ovalbumin-induced
`asthma. Unexpectedly, this effect could be reversed
`only if mice were exposed to microbiota in the
`neonatal period. The regulation of iNKT cell ex-
`pansion was ascribed to reduced expression of the
`chemokine CXCL16 in the presence of microbiota.
`Thus, signals elicited by commensals may repress
`systemic expression by epithelial cells of a chemo-
`kine that interacts with CCR6 that is selectively
`expressed by iNKT cells (49).
`Innate lymphoid cells that produce either
`IL-17 or IL-22 are protective against damage in
`an innate model of colitis and during Citrobacter
`rodentium enteric infection (50, 51). The extent
`to which innate lymphoid cells are regulated by
`the microbiota is not yet clear (52–54), but cryp-
`topatches, which are formed by a subset of innate
`lymphoid cells in the small intestine, differenti-
`ate into isolated lymphoid follicles only when
`commensals are present (55). Thus, it is likely
`that, even if innate lymphoid cell numbers are
`not influenced by commensals, their function
`may be subject to microbiota signals.
`Microbiota can trigger inflammation in immu-
`nocompromised hosts. The commensal microbiota
`clearly have important effects on the normal
`development of immunity. However, commensal
`bacteria can also trigger inflammatory responses in
`immunodeficient hosts. For example, defective
`signaling through the phosphatase SHP-1 causes a
`microbiota-dependent autoinflammatory syndrome
`with lesions on the feet, salivary glands, and lungs;
`such inflammation also occurs in mice without B or
`T lymphocytes (56, 57). There are a series of mono-
`genic conditions of the nucleotide-binding oligo-
`merization domain (NOD) receptor family (58)
`considered to be autoinflammatory. One of the best
`characterized of these is in the NLRP3 inflamma-
`some protein (59). Depending on the exact acti-
`vating mutation involved, the clinical spectrum in
`humans encompasses urticaria triggered by the cold,
`episodic fevers occurring with unknown triggers,
`and neonatal onset multisystem inflammatory dis-
`ease (60). Although the exact cause of these patholo-
`gies is not yet clear, these outcomes are consistent
`with studies in mice showing that NLRP3-deficiency
`can cause dysbiosis of the intestinal microbiota
`(61), as well as studies showing that TLR ligands
`can trigger proinflammatory IL-1b secretion in the
`presence of activating NLRP3 mutations (62, 63).
`Another NOD family member, NOD2 (CARD15),
`a receptor for the muramyl dipeptide structural unit
`of bacterial peptidoglycan, was the first susceptibil-
`ity gene identified for Crohn’s disease (64, 65). This
`reinforced early clinical observations of the benefits
`of surgically diverting the intestinal stream or treat-
`ing with antibiotics, thus implicating intestinal mi-
`crobes in the etiology (66, 67). More recent genetic
`data from genome-wide association studies (GWAS)
`
`of human inflammatory bowel disease have re-
`vealed a highly polygenic picture, with more than
`70 loci described for Crohn’s disease alone. These
`include modulators of the mucosal immune re-
`sponse, proteins functioning in the epithelial stress
`response, and the IL23R polymorphisms described
`above (68, 69). However, the sum total of the con-
`tributions of these loci to overall disease incidence
`leaves a considerable gap. It is clear that some cases
`can be explained by phenotypes from private mu-
`tations, such as those affecting IL-10 signaling (43),
`that are too infrequent to be detected by GWAS but
`that disrupt host-microbial mutualism in animal
`models (70).
`Microbiota can protect against autoimmune
`disease. Type 1 diabetes (T1D) results from auto-
`immune damage to the insulin-secreting islets of
`Langerhans in the pancreas. This autoimmune con-
`dition is also shaped by the interactions between
`immunity and the microbiota, but unlike EAE and
`arthritis, where SFB drive autoimmunity, the micro-
`biota can protect from T1D. The nonobe

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket