throbber
PRESS
`
`Cell
`
`Cell Host & Microbe
`
`Intestinal Commensal Microbes as Immune Modulators
`
`lvaylo l. lvanov1 -* and Kenya Honda2-3-*
`1Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032, USA
`2Department of Immunology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
`3RIKEN Research Center for Allergy and Immunology, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
`*Correspondence: ii2137@columbia.edu (l.l.l.), kenya@m.u-tokyo.ac.jp (K.H.)
`http://dx.doi.org/10.1016/j.chom.2012.09.009
`
`Commensal bacteria are necessary for the development and maintenance of a healthy immune system.
`Harnessing the ability of microbiota to affect host immunity is considered an important therapeutic strategy
`for many mucosal and nonmucosal immune-related conditions, such as inflammatory bowel diseases (IBDs),
`celiac disease, metabolic syndrome, diabetes, and microbial infections. In addition to well-established
`immunostimulatory effects of the microbiota, the presence of individual mutualistic commensal bacteria
`with immunomodulatory effects has been described. These organisms are permanent members of the
`commensal microbiota and affect host immune homeostasis in specific ways. Identification of individual
`examples of such immunomodulatory commensals and understanding their mechanisms of interaction
`with the host will be invaluable in designing therapeutic strategies to reverse intestinal dysbiosis and recover
`immunological homeostasis.
`
`Introduction
`
`Mucosal surfaces are colonized by a complex and dynamic
`microbial ecosystem termed “microbiota." An astounding
`number and diversity of microbes,
`including fungi, bacteria,
`archaea, and viruses, are present
`in any given moment
`throughout our bodies. The vast majority of these organisms
`are not disease-causing invaders, but have made the host their
`one-and-only home. Through coevolution,
`these “commen-
`sals” have established one of the most impressive examples
`of mutualistic relationship in the natural world,
`in which both
`microbes and their animal host depend on each other for
`optimal survival. Indeed, we rely on our microbiota for many
`basic physiologic and metabolic functions, as well as proper
`immune functions. Commensals provide immune protection in
`several ways. They can defend their mucosal home by directly
`combating invading pathogens or by mobilizing host antimicro-
`bial immune defenses. They can also affect host immunity in a
`more inconspicuous, but equally important, way by directing
`the development of host immune cell subsets at steady state
`and therefore affecting mucosal and systemic innate or
`adaptive immune responses. Recent studies have identified
`examples of commensal bacterial species with such immuno—
`modulatory roles. The existence of multiple members of the
`microbiota that affect host immune homeostasis in different
`
`ways means that differences in the composition of this com-
`munity may contribute to individual differences in immune
`responses during infection, autoimmunity, cancer, or other
`immunological conditions. Unveiling the underlying cellular
`and molecular mechanisms of each of these examples holds
`the promise to lead to exciting new ways for regulating mucosal
`immunity.
`
`Microbial-Host Interactions in the Intestine
`
`Decades of studies in germ-free (GF) animals have established
`the importance of microbiota for proper host immune function
`(Macpherson and Harris, 2004). GF animals were first created
`
`496 Cell Host & Microbe 12, October 18, 2012 @2012 Elsevier Inc.
`
`more than a century ago (Nuttal and Thierfeledr, 1895—1896),
`and long-term husbandry of GF rats has been possible since
`the 19403 (Reyniers et al., 1946). However, until recently, the
`composition of gut microbial communities remained largely
`unknown. Advances in high-throughput sequencing in the last
`few years have led to extensive cataloguing of the human
`microbiota (Human Microbiome Project Consortium, 2012).
`In
`addition to correlating changes in microbiota composition with
`disease, this has allowed for the identification of commensal
`species with specific immune effects. The vast majority of
`these studies have focused on the bacterial component of the
`microbiota; however, characterization of the fungal and viral
`components and their function is under way (lliev et al., 2012;
`Reyes et al., 2012).
`It
`The individual's microbiota composition is dynamic.
`changes with age and fluctuates with environmental changes,
`such as geographical location, diet, antibiotic use, or influx and
`efflux of external microbes (Clemente et al., 2012). In addition,
`vastly different microbial communities reside in different parts
`of the body (Costello et al., 2009). Based on their colonization
`ability, bacteria in the gut can be transient or permanent. Tran-
`sient bacteria represent microbes that are introduced during
`adult life from the external environment and do not permanently
`colonize the intestinal tract for various reasons, such as lack of
`appropriate adaptations for colonization or inability to compete
`with the resident microbiota. Many food-associated microbes,
`including pathogens and conventional commercial probiotics,
`are part of this category. Transient organisms can affect the
`immune system in different ways and be innocuous, pathogenic,
`or even beneficial, e.g., ingestion of probiotic-containing foods.
`These organisms have not coevolved and therefore do not
`establish a mutualistic relationship with the host, but rather
`try to survive in the gut environment despite established host
`defenses. Indeed, many acute intestinal pathogens have devel—
`oped strategies to forcefully colonize the intestine, which in
`most cases induces a strong immune response aimed at clearing
`Genome Ex. 1034
`
`Page 1 of 13
`
`Genome Ex. 1034
`Page 1 of 13
`
`

`

`Cell Host & Microbe
`
`
`
`Table 1 . Nonpathogenic Bacterial Members of the Intestinal Microbiota with Immune Effects
`Association
`
`Probiotics
`
`Autobionts
`
`Concept
`o Confer health benefit to
`the host when administered
`in adequate amount
`0 Not necessarily part of
`the “normal microbiota”
`0 May affect beneficial
`microbiota (indirect effects)
`0 Direct influence on host
`immune cell homeostasis
`or function
`0 Part of the ”normal
`microbiota"
`
`Examplesa
`Bifidobacterium spp;
`Lactobacillus spp
`
`with Host
`Transient
`
`Immune Effects
`Innocuous,
`immunostimulatory
`
`Mechanismsa
`Cytokine induction,
`TLR activation, pathobiont
`and pathogen suppression,
`lactic acid, short-chain
`fatty acids
`
`Bacteroides fragilis,
`Clostridia XIVa and IV,
`SFB, Faecalibacterium
`prauznitsii
`
`Permanent, host
`dependent,
`symbiotic
`
`Immunomodulatory
`
`Largely unknown (TLR2,
`metabolites [?], antigens [?],
`effects on IEC function [?])
`
`Pathobionts
`
`Helicobacter hepaticus,
`Clostridium difficile,
`Prevotela spp.,
`KIebsie/Ia spp.,
`Bilophila wadsworthia
`
`Permanent,
`parasitic/
`infectious
`
`0 Do not cause disease
`in the presence of normal
`microbiota in healthy host
`0 Cause disease when
`microbiota or host
`immunity is perturbed
`(?), speculative mechanisms.
`aExamples and mechanistic studies have been performed in mice (with the exception of F. prauznitsii and C. difficile). See text for details and
`references.
`
`Innocuous,
`detrimental
`
`Invasive mechanisms,
`spore formation, toxins
`
`the pathogen. The pathologic immunological consequences of
`this inflammatory response may persist for years after the clear-
`ance of the transient organisms, as in postinfectious irritable
`bowel syndrome (Spiller and Garsed, 2009).
`In contrast to transient bacteria, permanent bacteria are long-
`term members of the microbial community. Their colonization
`occurs in successive waves during ontogeny, and they have
`developed evolutionary adaptations to establish a permanent
`relationship with the host. In most cases they have coevolved
`with the host and are not normally found as free-living organisms.
`These are the true commensal bacteria. Commensals have mul-
`
`tiple effects on the immune system of the host. On one hand,
`the presence of a large number of “innocuous” bacteria has
`immunostimulatory effects. Commensals stimulate general
`recruitment of immune cells to the mucosa, as well as generation
`and maturation of organized gut-associated lymphoid tissues
`(Macpherson and Harris, 2004). They also stimulate protective
`epithelial cell
`functions, such as mucus and antimicrobial
`peptide secretion (Hooper and Macpherson, 2010). On the other
`hand, recent studies have identified the presence of commensal
`species with immunomodulatory effects. These effects are
`specific for individual bacteria or groups of bacteria,
`i.e., for
`specific components of the microbiota. They involve reversible
`changes in differentiation or effector function of host immune
`cell subsets. In this way, microbiota composition can influence
`the type and robustness of host immune responses. Here, we
`refer to such permanent microbiota members with immunomod-
`ulatory effects as autobionts. In contrast to transient pathogens
`or pathobionts (see below), the immune effects of autobionts are
`more subtle because they do not cause any overt change in the
`health state of the host. Rather, they help maintain and regulate
`the host’s healthy immune steady state. Permanent microbiota
`members that can demonstrate detrimental effects under
`
`special conditions are called pathobionts. Pathobionts colonize
`
`the host, but do not cause disease with a full complement of
`normal microbiota. However,
`they can expand and cause
`disease if microbiota or host immune homeostasis is perturbed
`(for example, after antibiotic treatment or under conditions of
`intestinal inflammation). The immune effects and characteristics
`of different intestinal bacteria are summarized in Table 1.
`
`In contrast to pathogens, pathobionts, and even probiotics,
`very little is known about the mechanisms by which autobionts
`exert their immunomodulatory effects. Until recently, one reason
`for this was the lack of specific examples of immunomodulatory
`commensals. Another reason is the difficulty in culturing these
`organisms ex vivo and the relative lack of genetic tools to study
`their genome function.
`In this review we focus on recently
`described examples of immunomodulatory commensals and
`speculate on potential cellular and molecular mechanisms
`involved in their interaction with the host and the establishment
`
`of a healthy steady immune state. Knowledge of these mecha-
`nisms can benefit the development of future therapies for intes-
`tinal diseases (Clemente et al., 2012).
`
`The Intestinal Microbial Community Influences
`Immunity
`During steady-state conditions, the microbiota affects the devel-
`opment and function of various immune cell populations,
`including lgA—secreting plasma cells, Th17 cells, regulatory T
`(Treg) cells, invariant natural killer T (iNKT) cells, yET cells, NK
`cells, macrophages, dendritic cells (DOS), and innate lymphoid
`cells (lLCs) (Honda and Littman, 2012). For example,
`lgA+
`plasma cells in gut lymphoid tissues and lamina propria (LP)
`are greatly reduced in GF or antibiotics-treated conventional
`animals (Macpherson and Harris, 2004).
`In another example,
`the abundance and function of CD4+ T cells expressing the
`FoxP3 transcription factor (Tregs) or interleukin (lL)-17 (Th17
`cells) in the intestinal mucosa at steady state are affected by
`Cell Host & Microbe 12, October 18, 2012 @2012 Elsevier Inc.Goggglne Ex. 1034
`Page 2 of 13
`
`Genome Ex. 1034
`Page 2 of 13
`
`

`

`
`
`the microbiota. In GF or antibiotic-treated mice, the percentages
`of both Tregs and Th17 cells are markedly reduced, and expres-
`sion of the immune-suppressive cytokine lL-10 in Treg cells is
`severely reduced (Atarashi et al., 2008, 2011;
`lvanov et al.,
`2008). These reductions are quickly restored by transplantation
`of intestinal or fecal microbiota from conventionally raised/
`specific pathogen-free (SPF) mice. As described below, the
`development of these CD4+ T cell subsets is differentially regu-
`lated by certain components of
`the intestinal microbiota
`including Bacteroides fragilis, Clostridia species, and segmented
`filamentous bacteria (SFB) (Atarashi et al., 2011; lvanov et al.,
`2009; Round and Mazmanian, 2010) (Table 1). Th17 cells
`contribute to host defense against infection with pathogenic
`microbes but can also augment harmful autoinflammatory func—
`tions, whereas Tregs play critical roles in immune suppression.
`Therefore, under healthy conditions Th17 cells and Tregs should
`coexist in a well-regulated balance. The particular combinations
`and relative abundances of the corresponding commensal
`species would generate distinct
`immune environments and
`immune responses in the host.
`In addition to inducing development or recruitment of host
`immune cell subsets, microbiota may also affect the function
`of these subsets. For example, gut microbiota provides an envi-
`ronment not only for the accumulation of lgA+ cells but also for
`the functional maturation of lgA+ plasma cells by inducing the
`generation of iNOS+ lgA+ plasma cells (Fritz et al., 2012).
`iNOS+ lgA+ plasma cells are absent in GF mice but present in
`conventionally raised mice. This subset plays critical roles in
`the enhancement of lgA+ plasma cell development and the
`host defense against enteric pathogens, such as Citrobacter
`rodentium (Fritz et al., 2012). Commensal bacteria also affect
`NK cell function. Even though NK cell numbers are normal in
`GF mice, NK cell priming and antiviral activity is deficient in the
`absence of microbiota (Ganal et al., 2012). This effect is due to
`microbiota-directed introduction of epigenetic changes and
`induction of type | interferons from monocytic macrophages,
`which are required for proper NK cell priming (Ganal et al., 2012).
`Microbiota-controlled immune effects may also play role in
`regulation of the microbiota itself. Commensal-induced lgA+
`plasma cells contribute to controlling microbiota abundance
`and composition. For instance, mice carrying a knockin mutation
`of activation-induced cytidine deaminase (AID) (AlDGzas), which
`can mediate normal
`lgA class switching but cannot induce
`somatic hypermutation and high-affinity lgA responses, exhibit
`excessive proliferation of anaerobic bacteria in the small intes-
`tine (Wei et al., 2011). Similar overgrowth of microbiota was
`also reported in programmed cell death-1 (PD-1)—deficient
`mice, which have increased follicular helperT (TFH) cell develop-
`ment, and therefore low-affinity lgA—producing plasma cells are
`aberrantly selected in germinal centers (Kawamoto et al.,
`2012). Thus, microbiota is required for the development of fully
`functional lgA+ cells, which in turn function to maintain microbial
`homeostasis in the gut.
`The microbiota also plays suppressive roles in immune cell
`function and accumulation in the gut. For example, it represses
`constitutive production of lL-22 in lymphoid tissue inducer (LTi)
`cells and NKp46+ cells (both of which are FiOFtyt+ lLCs; see
`detailed review by Tait Wojno and Artis, 2012 in this issue of
`Cell Host & Microbe) through epithelial expression of lL-25
`
`498 Cell Host & Microbe 12, October 18, 2012 @2012 Elsevier Inc.
`
`Cell Host & Microbe
`
`(Sawa et al., 2011). Another example is that early exposure to
`gut microbiota provides an epigenetic suppressive marking in
`the regulatory element of CXCL16 gene in the host, resulting in
`lifelong suppression of CXCL16 expression in gut and lung (Ols-
`zak et al., 2012). This suppression is accompanied by reduced
`abundance of iNKT cells in the colon and lung at steady state
`and affects host resistance to colitis and asthma (Olszak et al.,
`2012). Taken together,
`intestinal microbiota provide diverse
`signals for activation and suppression of the immune system,
`thereby having the ability to skew host immune status toward
`either effector or regulator dominance.
`Commensals have immunomodultory functions not only in the
`intestine. For example, resident commensals in the skin induce
`local Th17 and Th1 responses that are crucial
`in protection
`from bacterial infections (Naik et al., 2012). More importantly,
`these responses were compartmentalized to the skin and were
`independent of gut commensals (Naik et al., 2012).
`Commensals influence pathogenesis of many diseases.
`Because this community is evolutionarily established to sustain
`healthy immune steady state, any major perturbations in its
`composition may have negative effects and perpetuate the cycle
`of chronic inflammation, allergy, or metabolic syndrome. Such
`”dysbiosis” can be induced by diet, pharmacological agents,
`infection, inflammation, and host genetics (Honda and Littman,
`2012). Once established, the dysbiotic microbiota may become
`stable and transplantable to GF or even conventionally raised
`animals. Indeed, cohousing of wild-type mice with disease-
`prone mutant mice, such as Tbet‘l‘Fi’ag‘l‘, erp6‘l‘, or Ase"—
`mice, results in transfer of dysbiotic microbiota and predisposi-
`tion of the wild-type mice to disease,
`including colitis and
`metabolic syndrome (Elinav et al., 2011; Garrett et al., 2007;
`Henao-Mejia et al., 2012).
`Dysbiosis may lead to elimination of beneficial bacteria or
`outgrowth of pathobionts. Pathobionts are permanent members
`of the microbiota, present at low levels and innocuous under
`normal conditions (Table 1). They can become pathogenic if al-
`lowed sufficient expansion due to loss of microbiota or immune
`homeostasis. For instance, overgrowth of members of Prevotel-
`laceae and TM7 has been implicated in host susceptibility
`to DSS colitis in an6-/- mice (Elinav et al., 2011). In Their-’-
`Rag”’ mice, Proteus mirabilis and Klebsiella pneumoniae
`were identified to, at least in part, be responsible for the pheno-
`type of spontaneous colitis (Garrett et al., 2010). Most impor-
`tantly, disease susceptibility was transferable to wild-type mice.
`The composition of the gut microbiota is also altered by
`diet. Milk fat- and taurocholic acid-rich diets induced marked
`
`increase in Bilophila wadsworthia colonization, which is associ-
`ated with enhancement of Th1 responses and acceleration
`of colitis development in II10"‘ mice (Devkota et al., 2012).
`It should be noted that, depending on the host genotype, other-
`wise innocuous symbionts may become pathogenic. Indeed,
`in an inflammatory bowel disease (IBD) mouse model with
`deficiencies in lL-10 and TGF-B signaling, Bacteroides thetaio-
`taomicron, a well-characterized symbiotic species, potently
`induces colitis (Bloom et al., 2011).
`
`Autobionts—Mutualistic lmmunomodulatory Microbes
`As discussed above, there is abundant evidence that microbiota
`directs host
`immunity. Regulation of immune responses by
`Genome Ex. 1034
`
`Page 3 of 13
`
`Genome Ex. 1034
`Page 3 of 13
`
`

`

`Cell Host & Microbe
`
`influencing development, differentiation, or effector function of
`different cells of the immune system is especially interesting,
`because it results not simply from the presence of innocuous
`bacteria, but from the biological activity of commensals.
`In
`many cases these effects are functionally distinct, e.g., induction
`of Th1 7 cells versus Tregs, and depend on the activity of different
`members of the commensal community. The relative abundance
`of these “autobionts” can direct the general type of immunity
`in the host mucosa of an individual at a given time. Currently
`there are relatively few specific examples of commensals with
`immunomodulatory effects as discussed below. More are surely
`going to be identified in the future.
`Despite considerable species diversity, the intestinal micro-
`biota in most mammals consists of bacteria belonging to two
`major phyla—Gram-negative Bacteroidetes and Gram-positive
`Firmicutes. This probably reflects evolutionary adaptations of
`these phyla to survive in the gut environment, and both of
`them contain important immunomodulatory commensals.
`
`Bacteorides fragilis Promotes Treg Function
`B. fragilis was the first commensal to be implicated in affecting
`T helper cell balance by promoting Th1 development systemi-
`cally (Mazmanian et al., 2005). However, further studies showed
`that B.
`fragilis also affects mucosal T cell homeostasis by
`promoting regulatory T cell function (Round and Mazmanian,
`2010). B. fragilis is a Gram-negative member of the phylum
`Bacteroidetes.
`It
`is not a very abundant member of the gut
`microbiota; however, the genus Bacteroides is well represented
`in human gut and has a superior ability to utilize the nutrients in
`the gut microenvironment (Flint et al., 2008). For example, the
`genome of the prototypical dominant commensal member
`of the class, B.
`thetaiotaomicron, contains several hundred
`proteins involved in harvesting and metabolizing of dietary poly-
`saccharides (Sonnenburg et al., 2005). These are probably
`adaptations that have allowed Bacteroides to establish mutual-
`istic relationship with the host, by (1) being able to flourish in
`the plant polysaccharide-enriched gut environment and (2) being
`able to provide biological “byproducts" necessary for the well-
`being of the host. Different Bacteroidetes species produce
`different beneficial biological byproducts, which may have
`helped establish them as permanent symbionts. For example,
`B.
`thetaiotaomicron is one of the major producers of short-
`chain fatty acids (SCFAs), which are necessary for proper host
`metabolic and immune functions. B.
`fragilis seems to have
`pronounced immunomodulatory functions. B.
`fragilis is not
`normally present in conventionally raised SPF mice, and coloni-
`zation with B. fragilis protects mice from colitis in the T cell
`transfer and 2,4,6-trinitrobenzene sulfonic acid (TNBS) models
`(Mazmanian et al., 2008). This protection is due to the expansion
`of immune-suppressive IL-10 producing Tregs by the bacteria
`(Round and Mazmanian, 2010). The introduction of B. fragilis
`as a permanent member of the microbiota also affects T cell
`homeostasis in the absence of inflammation. Colonization of
`
`SPF or GF mice with B. fragilis leads to an induction of lL-10
`production by Foxp3+ Tregs even at steady state (Round and
`Mazmanian, 201 0). Thus, B. fragilis colonization modulates intes-
`tinal T cell homeostasis by boosting Treg function. This anti-
`inflammatory effect of the bacteria likely represents an evolu-
`tionary adaptation for establishing mutualism. Indeed, when
`
`
`
`Tregs were depleted, B. fragilis could not efficiently colonize
`host
`tissues (Round et al., 2011). Most
`importantly,
`the
`identification of B.
`fragilis as a modulator of Treg function
`allowed for investigation of the bacterial and host mechanisms
`involved.
`It was found that the anti-inflammatory effects of
`B. fragilis require the expression of bacterial capsular polysac-
`charide A (PSA). PSA-deficient B. fragilis mutants were incapable
`of inducing IL-10 production by Tregs and did not provide
`protection from colitis (Mazmanian et al., 2008; Round and
`Mazmanian, 2010). Instead, lack of PSA expression led to expan-
`sion of Th1 7 cells and loss of the mutualistic ability of B. fragilis to
`colonize host tissues (Round et al., 201 1). Moreover, treatment of
`mice with purified PSA is sufficient to replicate the effects of the
`bacteria,
`including induction of lL-10 production by Tregs,
`suppression of Th1 7 cell production, protection in colitis models,
`and colonization of the host (Mazmanian et al., 2008; Round
`et al., 2011; Round and Mazmanian, 2010). These studies re-
`present an elegant example of how investigating an immu-
`nomodulatory commensal and its effect on the host immune
`system can lead to elucidation of underlying molecular mecha-
`nisms and identification of clinically relevant immunomodulatory
`molecules.
`
`Cluster IV and XWa Clostridia Induce Treg
`Differentiation
`
`Intestinal Clostridia are a heterogeneous group that forms the
`core of Firmicutes of the normal commensal microbiota. Clostri-
`
`dia are Gram-positive, rod-shaped, endospore-forming bacteria.
`They are a highly heterogeneous class that is composed of at
`least 19 clusters based on genomic similarity (Collins et al.,
`1994). The prototypical Clostridia from cluster I contain frequent
`environmental toxin-producing members, such as Clostridium
`perfringens, C. difficile, and C. tetani. These Clostridia may be
`present in the intestine, but usually as transient pathogens or,
`at best, pathobionts (e.g., C. difficile).
`In contrast, most com-
`mensal intestinal Clostridia are nontoxinogenic members of clus-
`ters XIVa and IV. They are typically described as fusiform-shaped
`bacteria and constitute 10%—40% of the total microbiota (Frank
`et al., 2000. Cluster XIVa includes the genera Clostridium, Eu-
`bacterium, Ruminococcus, Coprococcus, and Roseburia. The
`cluster IV group includes species belonging to the Clostridium,
`Faecalibacten'um, and Ruminococcus genera. Clostridia colo-
`nize the mucus layers in the vicinity of the epithelium, in contrast
`to Bacteroidaceae, Enterococcaceae, and Lactobacillaceae,
`which colonize in regions ofthe central lumen, suggesting unique
`influences of Clostridia on host physiology (Nava and Stappen-
`beck, 2011). Indeed, the cluster XIVa Lachnospiraceae family
`is significantly less abundant
`in IBD patients compared to
`healthy subjects (Frank et al., 2007). Loss of mucosa-associated
`Clostridia and cluster IV Clostridia, particularly Faecalibacterium
`prausnitzii,
`is observed in IBD patients (Sokol et al., 2008).
`Although it remains unclear whether the decrease in Clostridia
`is a cause or effect of chronic inflammation, it is likely that main-
`tenance of the Clostridia community is necessary to prevent IBD.
`In addition to the role in the intestinal (local) immune homeostasis,
`Clostridia also affect systemic immunity. Indeed,
`it has been
`shown that reduction of Clostridia clusters XIVa and IV by neo-
`natal vancomycin treatment promotes airway hypersensitivity in
`a mouse model (Russell et al., 2012). Furthermore, decreased
`Gen
`me Ex. 1034
`Cell Host & Microbe 12, October 18, 2012 @2012 Elsevier Inc. 489
`Page 4 of 13
`
`Genome Ex. 1034
`Page 4 of 13
`
`

`

`
`
`abundances of clusters IV and XIVa Clostridia have been associ-
`
`ated with atopy during childhood (Candela et al., 2012).
`The levels of Clostridia clusters IV and XIVa in adult mouse
`colon microbiota were found to correlate with the numbers of
`
`Tregs (Atarashi et al., 2011; Russell et al., 2012). Consistent
`with a role in colonic Treg induction, GF mice colonized with
`46 strains of Clostridia clusters XIVa and IV showed accumula-
`
`tion of Treg cells in the colon (Atarashi et al., 201 1). The 46 strains
`of Clostridia were originally isolated from the sporulating micro-
`biota fraction of conventional mice based on their capacity to
`normalize the enlarged cecum in GF mice (Itoh and Mitsuoka,
`1985). These Clostridia were found to induce near steady-state
`numbers of colonic Tregs in GF mice, in contrast to other intes-
`tinal bacteria including Treg-associated autobionts such as
`B. fragilis (Atarashi et al., 2011). Whereas most Treg cells in the
`colon of GF mice were Helios”, which has been proposed as
`a marker for thymically derived Treg cells (Thornton et al.,
`2010), Treg cells in mice colonized with 46 strains of Clostridia
`clusters XIVa and IV were mostly Helios'° (Atarashi et al., 2011),
`suggesting that Clostridia robustly trigger peripheral differentia-
`tion of induced Treg (iTreg) cells. Consistent with these findings,
`colonization of CF mice with altered Schaedler flora (ASF),
`a defined bacterial cocktail containing eight enteric species
`that includes Clostridium clostridioforme, induces the accumula-
`tion of Treg cells in the colon (Geuking et al., 2011). Treg cells in
`the intestine exhibit different characteristics from those in
`
`secondary lymphoid organs, and they express CD103, killer
`cell
`Iectin-Iike receptor G1
`(KLRG1), granzyme B (szb),
`IL—10, and IL—35 (Feuerer et al., 2010). In particular, IL—10 plays
`an indispensable role in the suppression of aberrant activation
`of Th1 7 cells, myeloid cells, and y6T cells in the intestine. Indeed,
`Treg-specific disruption of IL-1 0 results in severe colitis (Rubtsov
`et al., 2008), as does a Treg-specific deficiency of STATS, which
`regulates many of the above-mentioned genes (Chaudhry et al.,
`2009). STAT3-deficient Tregs lack IL-10 expression and the
`mice develop spontaneous, Th17-mediated, commensal micro-
`biota-dependent fetal colitis (Chaudhry et al., 2009). Coloniza-
`tion of GF mice with the 46 strains of Clostridia clusters XIVa
`
`and IV induced not only an increase in Treg numbers but also
`high levels of IL-10 production (Atarashi et al., 2011). In humans,
`F. prausnitzii, which belongs to Clostridium cluster IV, increases
`IL-10 expression in peripheral blood mononuclear cells in vitro
`(Sokol et al., 2008). Therefore, autochthonous Clostridia con-
`stitutively induce accumulation and functional activation of
`Tregs in the colon and the relative abundance of Clostridia
`in the microbiota may strongly affect the immune status of the
`host.
`
`SFB Induce Th17 Cell Differentiation
`T cell homeostasis in the intestine can be defined as the balance
`
`between T cell subsets that promote immune responses and
`T cell subsets that subdue immune responses. IL-17-producing
`Th17 cells are proinflammatory CD4 T cells that contribute to
`disease pathogenesis in a number of chronic autoimmune
`inflammatory conditions, including IBD, multiple sclerosis, rheu-
`matoid arthritis, psoriasis, and certain cancers. At the same time,
`Th17 cells are crucial for efficient immune responses against
`mucosal pathogens,
`including viruses, bacteria, and fungi.
`Th17 cells differentiate from naive T cells under the combined
`
`500 Cell Host & Microbe 12, October 18, 2012 @2012 Elsevier Inc.
`
`Cell Host & Microbe
`
`effects of TGF-B and proinflammatory cytokines, such as IL-6,
`IL-23, and IL-18. The latter group of cytokines is upregulated in
`secondary lymphoid tissues during certain infections, which
`instructs Th17 cell differentiation in this context. However, at
`steady state, in the absence of infection or overt inflammation,
`Th17 cells are highly enriched in the intestinal LP and are not
`present in secondary lymphoid tissues (Ivanov et al., 2006). In
`the gut, Th17 cells coexist with Foxp3+ Tregs. The two subsets
`share common developmental pathway and alternative differen-
`tiation fates. This includes shared dependence on TGF—B and
`direct interaction and mutual functional
`inhibition of the two
`
`(Th17 cells) and
`regulators, RORyt
`master transcriptional
`FoxP3 (Tregs) (Zhou et al., 2008). This overlap of cytokine and
`transcriptional networks results in an elegant balance between
`two functionally opposing T cell subsets.
`Importantly,
`this
`balance is flexible and can be quickly reversed depending on
`the required immune response. One of the most
`important
`factors controlling the homeostasis between these two T cell
`subsets in the gut is the composition of intestinal microbiota.
`Th17 cells are not present in GF mice but are induced upon
`colonization with the full complement of gut bacteria from SPF
`mice (Atarashi et al., 2008; Ivanov et al., 2008).
`In contrast,
`colonization with culturable intestinal isolates, including Treg—
`inducing commensals, such as B. fragilis or the mix of 46 Clostri-
`dia described above, does not lead to Th17 cell induction (Ivanov
`et al., 2009; Round et al., 2011), arguing that commensal micro—
`biota contains unknown Th17 cell-inducing bacteria. This was
`further supported by the discovery that CS7BL/6 mice from
`a colony at the Jackson Laboratory did not contain gut Th1 7 cells
`due to the lack of Th17-celI-inducing bacteria and that total
`microbiota from these mice could not induce Th17 cells in CF
`animals,
`in contrast to microbiota from CS7BL/6 mice from
`a colony at Taconic Farms (Ivanov et al., 2008). Comparison of
`the microbiota between these two colonies revealed that
`
`Taconic B6 mice are highly enriched in SFB, which are absent
`from Jackson B6 mice (Ivanov et al., 2009). Interestingly, mono-
`colonization of GF animals with SFB or introduction of SFB into
`Jackson B6 mice induces Th17 cell differentiation in the LP
`
`(Gaboriau-Routhiau et al., 2009; Ivanov et al., 2009), identifying
`SFB as a Th17-cell-inducing autobiont.
`SFB are Gram-positive anaerobic bacteria that are known to
`permanently colonize the intestinal tract of many animal species.
`SFB or SFB-like bacteria have been described in invertebrates,
`such as termites and cockroaches, and in vertebrate animals
`such as fish, chickens, rabbits, mice, rats, cats, dogs, sheep,
`cows, pigs, zebras, and monkeys (Klaasen et al., 1992). How-
`ever, SFB have not yet been detected in humans (Sczesnak
`et al., 2011). SFB are members of the Firmicutes phylum and,
`based on the 168 rRNA sequence, were assigned to Clostridia
`(Snel et al., 1995). The similarity to Clostridia was later confirmed
`by the full SFB genomic sequence (Sczesnak et al., 2011). In
`fact, more than

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