throbber
REVIEWS
`
`The light and dark sides of intestinal
`intraepithelial lymphocytes
`
`Hilde Cheroutre*, Florence Lambolez*and Daniel Mucida‡
`
`Abstract | The intraepithelial lymphocytes (IELs) that reside within the epithelium of the
`intestine form one of the main branches of the immune system. As IELs are located at this
`critical interface between the core of the body and the outside environment, they must balance
`protective immunity with an ability to safeguard the integrity of the epithelial barrier: failure to
`do so would compromise homeostasis of the organism. In this Review, we address how the
`unique development and functions of intestinal IELs allow them to achieve this balance.
`
`Pathogens
`Opportunistic organisms that
`cause acute or chronic disease
`following host infection.
`Derived from the Greek word
`‘pathos’, which means
`‘suffering’.
`
`Intraepithelial lymphocytes
`(IELs). These lymphocyte
`populations consist mostly of
`T cells and are found within the
`epithelial layer of mammalian
`mucosal linings, such as the
`gastrointestinal tract and
`reproductive tract. However,
`unlike conventional naive
`T cells, IELs are antigen-
`experienced T cells and, on
`encountering antigens, they
`immediately release cytokines
`or mediate killing of infected
`target cells.
`
`*Laboratory of
`Developmental Immunology,
`La Jolla Institute for Allergy
`and Immunology, La Jolla,
`California 92037, USA.
`‡Laboratory of Mucosal
`Immunology, The Rockefeller
`University, New York,
`New York 10065, USA.
`Correspondence to H.C. and
`D.M.
`e-mails: hilde@liai.org;
`mucida@rockefeller.edu
`doi:10.1038/nri3007
`Published online 17 June 2011
`
`The epithelium of the intestine digests and absorbs
`nutrients and fluids, and in adult humans it spans
`an area of about 200–400 m2 (REF. 1). This huge sur-
`face is made up of a single cell layer of epithelial
`cells, which lines the lumen of the intestine to form
`a physical barrier between the core of the body and
`the environment and forms the largest entry port for
`pathogens. Probably as a consequence of this, sophis-
`ticated and complex innate and adaptive immune
`networks intensely communicate and synergize to
`tightly control the integrity of this critical interface.
`Although a major task of the mucosal immune system
`is to provide protection against intestinal pathogens,
`it is important that excessive or unnecessary immune
`responses are avoided.
`In this Review, we focus on the intraepithelial
`lymphocytes (IELs), which, by their direct contact with
`the enterocytes and by their immediate proximity to
`antigens in the gut lumen, form the front line of immune
`defence against invading pathogens. IELs essentially
`comprise antigen-experienced T cells belonging to
`both the T cell receptor-γδ (TCRγδ)+ and TCRαβ+ lin-
`eages. We discuss the thymic (natural) and peripheral
`(induced) differentiation of various IEL subpopulations,
`as well as their beneficial roles (that is, their ‘light side’)
`in preserving the integrity of the mucosal barrier and in
`preventing pathogen entry and spreading. Conversely,
`we also expose the ‘dark side’ of IELs and discuss how
`these cells can contribute to immune pathology and
`inflammatory diseases.
`
`Mucosal IELs are unique and heterogeneous
`IELs are extremely heterogeneous, and the various IEL
`subsets are distributed differently in the epithelium
`of the small and large intestine (TABLE 1). This pattern of
`distribution is probably influenced by the distinct
`
`digestive functions and the physiological conditions
`that allow these two compartments to cope with infec-
`tions while simultaneously maintaining tolerance to
`innocuous antigens from the diet or from resident
`non-invasive commensals. Nevertheless, these IELs
`also share characteristics that distinguish them from
`the conventional T cell pools in the periphery.
`Gut IELs are almost exclusively T cells, and esti-
`mates based on histological sections indicate that there
`are more T cells in the intestinal epithelium than in the
`spleen2. IELs include a significant proportion of TCRγδ+
`cells, which can constitute up to 60% of small intestinal
`IELs3–5. These IELs are antigen-experienced cells that
`typically express activation markers, such as CD44 and
`CD69 (REF. 6). Furthermore, studies using parabiotic
`mice and intestinal grafting indicate that these antigen-
`experienced T cells do not recirculate7,8. The majority of
`IELs contain abundant cytoplasmic granules for cyto-
`toxic activity, and they can express effector cytokines,
`such as interferon-γ (IFNγ), interleukin-2 (IL-2), IL-4
`or IL-17 (REFS 9–16). Furthermore, they characteristi-
`cally express both activating and inhibitory types of
`innate natural killer (NK) cell receptors, which typify
`them as stress-sensing (activated) yet highly regulated
`(resting) immune cells9,11,17–20.
`IELs constitutively express CD103 (also known as the
`αE integrin), which interacts with E-cadherin on intes-
`tinal epithelial cells21,22, and most of them, especially in
`the small intestine, express CD8αα homodimers, which
`is a hallmark of their activated phenotype18,23–25 (BOX 1).
`A ligand for CD8αα, the thymus leukaemia antigen (TLA),
`which is a non-classical MHC class I molecule, is abun-
`dantly expressed on mouse small intestinal epithelial
`cells26,27. Many TCRαβ IELs express CD8αα together
`with CD4 or CD8αβ; however, a large fraction expresses
`CD8αα alone16. Finally, under normal conditions, and
`
`NATURE REVIEWS | IMMUNOLOGY
`
` VOLUME 11 | JULY 2011 | 445
`
`© 2011 Macmillan Publishers Limited. All rights reserved
`
`Genome Ex. 1058
`Page 1 of 12
`
`

`

`R E V I E W S
`
`Table 1 | Characteristics of the different intestinal T cells subsets in mice
`Cell
`Characteristics
`type
`Antigen
`IEL phenotype
`dependency
`
`Kinetics of
`accumulation
`
`Frequency of
`TCRγδ and
`TCRαβ IELs
`
`Development and
`TCR repertoire
`
`Potential
`beneficial
`functions
`
`Potential
`pathogenic
`functions
`
`Small intestinal epithelium
`Natural
`Self and
`CD2–CD5–CD28–;
`IELs
`non-self
`CTLA4–THY1–/low;
`B220+/– CD69hi;
`NK receptors
`(+++++)
`
`TCRγδ+ (+++):
`– CD4–CD8– (++)
`– CD8αα+ (+++)
`TCRαβ+ (+++):
`– CD4–CD8– (+)
`– CD8αα+ (+++)
`
`Present
`at birth;
`decrease with
`age
`
`Induced
`IELs
`
`Non-self
`
`CD2+CD5+CD28+/–;
`CTLA4+/–THY1hi;
`B220–CD69hi;
`NK receptors
`(++++)
`
`TCRαβ+ (+++):
`– CD8αβ+ (+)
`– CD8αβ+CD8αα+
` (+++)
`– CD4+ (+)
`– CD4+CD8αα+ (++)
`
`Absent at
`birth; increase
`with age
`
`Anti-inflammatory
`responses;
`antimicrobial
`responses;
`tolerance to
`intestinal antigens;
`immune regulation;
`homeostasis of the
`epithelium
`Oral tolerance;
`protective
`antimicrobial
`responses
`
`‘Alternative’
`positive selection
`for CD8αα+TCRαβ+
`IELs in the thymus;
`oligoclonal TCR
`repertoire; unknown
`MHC restriction
`
`‘Conventional’
`positive selection
`in the thymus;
`oligoclonal TCR
`repertoire; restricted
`on MHC class I or
`MHC class II
`
`Small intestinal lamina propria
`T cells
`Non-self
`Not applicable
`
`TCRαβ (+++):
`– CD8αβ+ (++)
`– CD8αβ+CD8αα+ (+)
`– CD4+ (+++)
`– CD4+CD8αα+ (+)
`
`Absent at
`birth; increase
`with age
`
`Oral tolerance;
`protective
`antimicrobial
`responses
`
`‘Conventional’
`positive selection
`in the thymus;
`polyclonal TCR
`repertoire; restricted
`on MHC class I or
`MHC class II
`
`Promote
`epithelial
`damage during
`intestinal
`inflammation
`
`Excessive
`inflammation
`and cytotoxicity
`in response
`to luminal
`antigens;
`exacerbate
`coeliac disease
`
`Ileitis;
`inflammation
`
`Large intestinal epithelium
`Natural
`Self and
`CD2–CD5–CD28–;
`IELs
`non-self
`CTLA4–THY1–/low;
`B220+/–CD69hi;
`NK receptors (+++)
`
`Induced
`IELs
`
`Non-self
`
`CD2+CD5+CD28+;
`CTLA4+THY1hi;
`B220–CD69hi;
`NK receptors (++)
`
`TCRγδ+ (+):
`– CD4–CD8– (++)
`– CD8αα+ (+)
`TCRαβ (++):
`– CD4–CD8– (++)
`– CD8αα+ (+)
`TCRαβ+ (++):
`– CD8αβ+ (+)
`– CD8αβ+CD8αα+ (+)
`– CD4+ (++)
`– CD4+CD8αα+ (+)
`
`Present
`at birth;
`decrease with
`age
`
`‘Alternative’ positive
`selection for
`CD8αα+TCRαβ+ IELs
`in the thymus; TCR
`repertoire and MHC
`restriction unknown
`
`Anti-inflammatory
`responses;
`antimicrobial
`responses;
`tolerance to
`intestinal antigens
`
`Promote
`epithelial
`damage during
`intestinal
`inflammation
`
`Absent at
`birth; increase
`with age
`
`‘Conventional’
`positive selection
`in the thymus; TCR
`repertoire not
`defined; restricted
`on MHC class I or
`MHC class II
`
`Protective
`antimicrobial
`responses
`
`Drive excessive
`inflammation
`and
`cytotoxicity
`in response to
`microbiota-
`derived
`antigens;
`exacerbate IBD
`
`Large intestinal lamina propria
`T cells
`Non-self
`Not applicable
`
`TCRαβ+ (+++):
`– CD8αβ+ (++)
`– CD8αβ+CD8αα+ (+)
`– CD4+ (+++)
`– CD4+CD8αα+ (+)
`
`Absent at
`birth; increase
`with age
`
`Drive excessive
`inflammation
`and cytotoxicity
`in response to
`microbiota-
`derived
`antigens;
`exacerbate IBD
`Plus symbols represent frequency of expression, from low (+) to high (+++++). CTLA4, cytotoxic T lymphocyte A4; IBD, inflammatory bowel disease;
`IEL,  intraepithelial lymphocyte; NK, natural killer; TCR, T cell receptor.
`
`Protective
`antimicrobial
`responses
`
`‘Conventional’
`positive selection
`in the thymus;
`polyclonal TCR
`repertoire; restricted
`on MHC class I or
`MHC class II
`
`in contrast to systemic and lamina propria lymphocytes
`(LPLs), CD4+ cells are greatly under-represented in the
`IEL compartment, especially in the small intestine28,29.
`Although all IELs have an antigen-experienced phe-
`notype, they can be divided into two major subsets based
`on the mechanisms by which they become activated
`and on the cognate antigens that they recognize. The
`
`‘natural’ IELs (which were previously known as ‘type b’
`IELs)5 acquire their activated phenotype during develop-
`ment in the thymus in the presence of self antigens, whereas
`the ‘induced’ IELs (which were previously known as ‘type a’
`IELs)5 are the progeny of conventional T cells that are
`activated post-thymically in response to peripheral
`antigens (FIG. 1; TABLE 1).
`
`446 | JULY 2011 | VOLUME 11
`
` www.nature.com/reviews/immunol
`
`© 2011 Macmillan Publishers Limited. All rights reserved
`
`Genome Ex. 1058
`Page 2 of 12
`
`

`

`Box 1 | CD8αα as a repressor
`CD8αα, which is composed of two CD8α subunits, can be induced on activation
`through the T cell receptor (TCR)–CD3 complex, with the level of its expression
`being proportional with the signal strength. Therefore, CD8αα can be used as an
`activation marker for T cells43. CD8αα has the ability to recruit TCR–CD3 signalling
`components, including LCK (also known as p56LCK) and linker for activation of
`T cells (LAT)165. However, owing to its physical separation from the TCR-activation
`complex166, CD8αα neither functions as a TCR co-receptor nor can it substitute for
`the CD4 or CD8αβ co-receptors (see the figure, part a). Consistent with this, CD8αα
`is unable to support positive selection of MHC class I-restricted thymocytes or
`promote activation of mature MHC class I-restricted T cells167. Instead, co-expression
`of CD8αα on CD8αβ+ (or CD4+) T cells downmodulates, rather than enhances, the
`functional avidity of the MHC–TCR activation complex113. The ability of CD8αα to
`function as a negative regulator is partly due to its exclusion from the TCR activation
`complex (see the figure, part b). Consequently, CD8αα can sequester signalling
`components outside the immunological synapse, thereby interfering with their
`recruitment to the CD8αβ or the CD4 co-receptor–TCR–CD3 activation complex.
`Thus, CD8αα serves as a TCR repressor (see the figure, part b) rather than as a
`TCR co-receptor (see the figure, part a). See REF. 43 for further details.
`
`Ag, antigen; APC, antigen-presenting cell; ITAM, immunoreceptor tyrosine-based
`activation motif.
`
`(cid:67)
`
`(cid:35)(cid:50)(cid:37)
`
`(cid:54)(cid:2)(cid:69)(cid:71)(cid:78)(cid:78)
`
`(cid:68)
`
`(cid:37)(cid:38)(cid:26)β
`(cid:37)(cid:38)(cid:26)α
`
`(cid:47)(cid:42)(cid:37)(cid:2)(cid:69)(cid:78)(cid:67)(cid:85)(cid:85)(cid:2)(cid:43)
`(cid:35)(cid:73)
`(cid:54)(cid:37)(cid:52)αβ
`
`(cid:37)(cid:38)(cid:26)α
`(cid:37)(cid:38)(cid:26)α
`
`(cid:37)(cid:38)(cid:26)β
`(cid:37)(cid:38)(cid:26)α
`
`(cid:46)(cid:35)(cid:54)
`
`ε γ ζζ δδ
`
`(cid:46)(cid:35)(cid:54)
`
`(cid:46)(cid:35)(cid:54)
`
`(cid:46)(cid:35)(cid:54)
`
`ε γ ζζ δδ
`
`(cid:46)(cid:37)(cid:45)
`
`(cid:46)(cid:75)(cid:82)(cid:75)(cid:70)(cid:2)(cid:84)(cid:67)(cid:526)
`
`(cid:43)(cid:54)(cid:35)(cid:47)
`
`(cid:46)(cid:37)(cid:45)
`
`(cid:46)(cid:37)(cid:45)
`
`(cid:46)(cid:37)(cid:45)
`
`(cid:54)(cid:37)(cid:52)(cid:2)(cid:85)(cid:75)(cid:73)(cid:80)(cid:67)(cid:78)
`
`(cid:37)(cid:38)(cid:26)αα(cid:2)(cid:85)(cid:87)(cid:82)(cid:82)(cid:84)(cid:71)(cid:85)(cid:85)(cid:71)(cid:85)
`(cid:54)(cid:37)(cid:52)(cid:2)(cid:85)(cid:75)(cid:73)(cid:80)(cid:67)(cid:78)(cid:78)(cid:75)(cid:80)(cid:73)
`
`(cid:54)(cid:37)(cid:52)(cid:2)(cid:85)(cid:75)(cid:73)(cid:80)(cid:67)(cid:78)(cid:78)(cid:75)(cid:80)(cid:73)
`
`Thymus leukaemia antigen
`(TLA). A non-polymorphic,
`non-classical MHC class I
`molecule (MHC class I-b family)
`with a restricted expression
`pattern. It is constitutively
`expressed on intestinal epithelial
`cells and can be induced on
`antigen-presenting cells. TLA is
`structurally incapable of binding
`or presenting peptide antigens
`and it does not engage with
`T cell receptors. However, the
`α3 extracellular domain of
`TLA interacts with CD8α. TLA
`displays stronger affinity for
`CD8αα homodimers compared
`with CD8αβ heterodimers, and
`CD8αα expression can be
`detected with TLA-specific
`tetramers.
`
`(cid:48)(cid:67)(cid:86)(cid:87)(cid:84)(cid:71)(cid:2)(cid:52)(cid:71)(cid:88)(cid:75)(cid:71)(cid:89)(cid:85)(cid:2)(cid:94)(cid:2)(cid:43)(cid:79)(cid:79)(cid:87)(cid:80)(cid:81)(cid:78)(cid:81)(cid:73)(cid:91)
`
`Natural IELs. Natural IELs are either CD8αα+ or
`CD8αα– T cells that express TCRγδ or TCRαβ but
`do not express either CD4 or CD8αβ. Typically, they
`are also negative for expression of CD2, CD5, CD28,
`lymphocyte function-associated antigen 1 (LFA1; also
`known as αLβ2 integrin) and THY1 (REFS 9,28,30,31).
`In addition, the majority of TCRαβ+ natural IELs
`are B220+CD44low/midCD69+, a phenotype that is
`rarely found among peripheral T cells32,33. Many of
`the natural IELs also share molecules expressed by
`NK cells, such as CD16, CD122, DNAX-activation
`protein 12 (DAP12; also known as TYROBP), lympho-
`cyte antigen 49A (Ly49A), Ly49E, Ly49G2, and NK1.1
`(REFS 9,18,34,35). Lastly, they frequently express a CD3
`complex that is composed of either CD3ζ–FcεRIγ
`heterodimers or FcεRIγ–FcεRIγ homodimers instead
`of the CD3ζ–CD3ζ homodimers that are expressed by
`conventional T cells9,36–38.
`
`R E V I E W S
`
`Induced IELs. Induced IELs arise from conventional
`CD4+ or CD8αβ+ TCRαβ+ T cells, which are MHC
`class II-restricted and MHC class I-restricted, respec-
`tively. In contrast to the natural IELs, induced IELs
`acquire an activated phenotype in response to cog-
`nate antigens encountered in the periphery 33,39–42.
`Consistent with this, they typically express a ‘memory-
`like phenotype’ (CD2+CD5+CD44+LFA1+THY1+)9,28,30,
`together with the activation marker CD8αα43 (BOX 1).
`Contrary to the polyclonal nature of the conventional
`T cells in the periphery, the TCR repertoire of induced
`IELs is oligoclonal and does not significantly overlap
`with the limited TCR repertoire of the natural TCRαβ+
`IEL compartment44.
`
`Thymic development of IEL precursor cells
`Similarly to peripheral T cells, all IEL subsets are prog-
`eny of bone marrow precursor cells that initially develop
`in the thymus45. However, thymocyte maturation is not
`uniform, and multiple pathways exist that ultimately
`determine the diversity of mature T cells, including IELs.
`
`Development of natural IELs. The origin and develop-
`ment of TCRγδ and TCRαβ natural IELs are the subjects
`of long-standing debates24,46–49 (BOX 2). For an overview
`of recent advances on this topic, we refer the reader to
`previously published reviews6,45,50,51. Briefly, the term
`‘natural’ refers to the ontogeny of the precursor cells.
`For the natural IELs, these precursors go through an
`‘alternative’ self-antigen-based thymic maturation
`process that results in the functional differentiation
`of mature CD4 and CD8αβ double-negative, TCRγδ-
`expressing or TCRαβ-expressing T cells that directly
`migrate to the intestinal epithelium23,24,52 (FIG. 1).
`
`Development of induced IELs. Induced IELs are the prog-
`eny of conventional CD4+ or CD8αβ+ TCRαβ+ T cells
`that are selected in the thymus. The development of
`conventional TCRαβ+ thymocytes, including selection
`and lineage commitment has been extensively reviewed
`elsewhere53–55. Following positive selection, mature
`thymocytes exit the thymus and reach the periphery as
`conventional naive CD4+ or CD8αβ+ TCRαβ+ T cells. In
`response to cognate antigens, they can further mature
`into antigen-experienced cells, including induced
`IELs (FIG. 1).
`
`Local differentiation of IELs
`Because of the antigen-experienced phenotype that
`they acquire in the thymus, one can assume that the
`repertoire of the natural IELs is predominantly tuned to
`self antigens, whereas induced IELs are mainly shaped
`by non-self antigens encountered in the periphery.
`Consequently, induced IELs are sparse early in life, but
`the population steadily increases with age in response
`to exposure to exogenous antigens56–59 (FIG. 2; TABLE 1).
`The gradual accumulation of induced IELs allows the
`mucosal immune system to adapt and develop an almost
`‘personalized’ mucosal immune repertoire that is directed
`against those environmental antigens that are most
`likely to be re-encountered by a particular individual.
`
`NATURE REVIEWS | IMMUNOLOGY
`
` VOLUME 11 | JULY 2011 | 447
`
`© 2011 Macmillan Publishers Limited. All rights reserved
`
`Genome Ex. 1058
`Page 3 of 12
`
`

`

`R E V I E W S
`
`(cid:54)(cid:74)(cid:91)(cid:79)(cid:87)(cid:85)
`
`(cid:54)(cid:84)(cid:75)(cid:82)(cid:78)(cid:71)(cid:15)
`(cid:80)(cid:71)(cid:73)(cid:67)(cid:86)(cid:75)(cid:88)(cid:71)
`(cid:86)(cid:74)(cid:91)(cid:79)(cid:81)(cid:69)(cid:91)(cid:86)(cid:71)
`
`(cid:38)(cid:81)(cid:87)(cid:68)(cid:78)(cid:71)(cid:15)
`(cid:82)(cid:81)(cid:85)(cid:75)(cid:86)(cid:75)(cid:88)(cid:71)
`(cid:86)(cid:74)(cid:91)(cid:79)(cid:81)(cid:69)(cid:91)(cid:86)(cid:71)
`
`(cid:47)(cid:46)(cid:48)(cid:85)(cid:2)(cid:81)(cid:84)(cid:2)(cid:50)(cid:71)(cid:91)(cid:71)(cid:84)(cid:111)(cid:85)(cid:2)(cid:82)(cid:67)(cid:86)(cid:69)(cid:74)(cid:71)(cid:85)
`(cid:50)(cid:71)(cid:82)(cid:86)(cid:75)(cid:70)(cid:71)(cid:115)(cid:47)(cid:42)(cid:37)
`(cid:54)(cid:37)(cid:52)
`
`(cid:43)(cid:80)(cid:86)(cid:71)(cid:85)(cid:86)(cid:75)(cid:80)(cid:71)
`
`(cid:110)(cid:37)(cid:81)(cid:80)(cid:88)(cid:71)(cid:80)(cid:86)(cid:75)(cid:81)(cid:80)(cid:67)(cid:78)(cid:111)
`(cid:82)(cid:81)(cid:85)(cid:75)(cid:86)(cid:75)(cid:88)(cid:71)
`(cid:85)(cid:71)(cid:78)(cid:71)(cid:69)(cid:86)(cid:75)(cid:81)(cid:80)
`
`(cid:37)(cid:38)(cid:22)(cid:13)(cid:2)
`(cid:54)(cid:37)(cid:52)αβ(cid:13)
`
`(cid:35)(cid:50)(cid:37)
`
`(cid:48)(cid:67)(cid:75)(cid:88)(cid:71)(cid:2)(cid:37)(cid:38)(cid:22)(cid:13)(cid:2)
`(cid:54)(cid:37)(cid:52)αβ(cid:13)(cid:2)(cid:54)(cid:2)(cid:69)(cid:71)(cid:78)(cid:78)
`
`(cid:37)(cid:38)(cid:26)αβ(cid:13)
`(cid:54)(cid:37)(cid:52)αβ(cid:13)
`
`(cid:35)(cid:50)(cid:37)
`
`(cid:48)(cid:67)(cid:75)(cid:88)(cid:71)(cid:2)(cid:37)(cid:38)(cid:26)αβ(cid:13)
`(cid:54)(cid:37)(cid:52)αβ(cid:13)(cid:2)(cid:54)(cid:2)(cid:69)(cid:71)(cid:78)(cid:78)
`
`(cid:39)(cid:82)(cid:75)(cid:86)(cid:74)(cid:71)(cid:78)(cid:75)(cid:67)(cid:78)
`(cid:69)(cid:71)(cid:78)(cid:78)
`
`(cid:54)(cid:37)(cid:52)γδ(cid:13)(cid:2)
`(cid:80)(cid:67)(cid:86)(cid:87)(cid:84)(cid:67)(cid:78)(cid:2)(cid:43)(cid:39)(cid:46)(cid:2)
`(cid:10)(cid:37)(cid:38)(cid:26)αα(cid:13)(cid:17)(cid:115)(cid:11)(cid:2)
`
`(cid:37)(cid:38)(cid:22)(cid:13)
`(cid:75)(cid:80)(cid:70)(cid:87)(cid:69)(cid:71)(cid:70)(cid:2)(cid:43)(cid:39)(cid:46)
`(cid:10)(cid:37)(cid:38)(cid:26)αα(cid:13)(cid:17)(cid:115)(cid:11)
`
`(cid:37)(cid:38)(cid:26)αβ(cid:13)
`(cid:75)(cid:80)(cid:70)(cid:87)(cid:69)(cid:71)(cid:70)(cid:2)(cid:43)(cid:39)(cid:46)
`(cid:10)(cid:37)(cid:38)(cid:26)αα(cid:13)(cid:17)(cid:115)(cid:11)
`
`(cid:54)(cid:84)(cid:75)(cid:82)(cid:78)(cid:71)(cid:15)
`(cid:82)(cid:81)(cid:85)(cid:75)(cid:86)(cid:75)(cid:88)(cid:71)
`(cid:86)(cid:74)(cid:91)(cid:79)(cid:81)(cid:69)(cid:91)(cid:86)(cid:71)
`
`(cid:110)(cid:35)(cid:78)(cid:86)(cid:71)(cid:84)(cid:80)(cid:67)(cid:86)(cid:75)(cid:88)(cid:71)(cid:111)
`(cid:82)(cid:81)(cid:85)(cid:75)(cid:86)(cid:75)(cid:88)(cid:71)
`(cid:85)(cid:71)(cid:78)(cid:71)(cid:69)(cid:86)(cid:75)(cid:81)(cid:80)
`
`(cid:38)(cid:81)(cid:87)(cid:68)(cid:78)(cid:71)(cid:15)
`(cid:80)(cid:71)(cid:73)(cid:67)(cid:86)(cid:75)(cid:88)(cid:71)(cid:2)
`(cid:54)(cid:37)(cid:52)αβ(cid:13)
`
`(cid:54)(cid:37)(cid:52)αβ(cid:13)(cid:2)(cid:80)(cid:67)(cid:86)(cid:87)(cid:84)(cid:67)(cid:78)(cid:2)(cid:43)(cid:39)(cid:46)
`(cid:10)(cid:37)(cid:38)(cid:26)αα(cid:13)(cid:17)(cid:115)(cid:11)(cid:2)
`
`(cid:38)(cid:81)(cid:87)(cid:68)(cid:78)(cid:71)(cid:15)
`(cid:80)(cid:71)(cid:73)(cid:67)(cid:86)(cid:75)(cid:88)(cid:71)(cid:2)
`(cid:54)(cid:37)(cid:52)γδ(cid:13)
`
`(cid:48)(cid:67)(cid:86)(cid:87)(cid:84)(cid:71)(cid:2)(cid:52)(cid:71)(cid:88)(cid:75)(cid:71)(cid:89)(cid:85)(cid:2)(cid:94)(cid:2)(cid:43)(cid:79)(cid:79)(cid:87)(cid:80)(cid:81)(cid:78)(cid:81)(cid:73)(cid:91)
`Figure 1 | Thymic and peripheral differentiation of natural and induced IELs. In the thymus, immature
`CD4+CD8αβ+CD8αα+ (triple positive) thymocytes undergo agonist (‘alternative’) selection and differentiate into
`double-negative T cell receptor-αβ (TCRαβ)+ cells that are the precursors of natural TCRαβ+ intraepithelial
`lymphocytes (IELs). The TCRαβ+ natural IEL precursor cells partly acquire their antigen-experienced phenotype
`‘naturally’ during selection with self antigens. In addition, the precursor cells for TCRαβ+ and TCRγδ+ natural IELs
`upregulate intestinal homing receptors during their maturation in the thymus, which allows these cells to directly
`seed the intestinal epithelium after they leave the thymus. CD4+CD8αβ+ (double positive) thymocytes undergo
`‘conventional’ thymic selection and differentiate into naive CD4+ and CD8αβ+ TCRαβ+ T cells that migrate to the
`periphery. These naive T cells can differentiate into effector T cells in response to peripheral antigens and subsequently
`migrate to the gut and become incorporated into the induced IEL compartment. APC, antigen-presenting cell;
`MLNs, mesenteric lymph nodes.
`
`The development of these antigen-specific immune cells
`not only provides focused protective immunity at this
`mucosal interface but, at the same time, also reduces the
`risk of unwanted immune responses directed against
`innocuous antigens. By contrast, natural IELs do not
`depend on exogenous antigen-driven differentiation and
`so they are the first type of antigen-experienced T cells
`to populate the gut, even before birth60 (FIG. 2). Although
`direct evidence is still lacking, it is likely that the early
`accumulation of natural IELs provides a self-antigen-
`based, stress-sensing surveillance mechanism that is
`tolerant to dietary antigens and colonizing microbiota
`but provides protective immunity against stress-induc-
`ing invasive pathogens. With time, the population of
`induced IELs gradually becomes larger than the natural
`IEL population, which remains steady in actual numbers
`but represents a minor IEL population at later stages in
`life (FIG. 2).
`Nevertheless, regardless of the nature of the cog-
`nate antigens or the location of the initial differentia-
`tion, all IELs are directly influenced by the intestinal
`environment61. This was demonstrated with elegant
`studies in ‘germ-free’ and ‘antigen-free’ mice (the germ-
`free mice were fed an elementary diet), which showed
`that both the microbiota and dietary proteins have
`a crucial role in the establishment of a normal IEL
`
`repertoire, as virtually all IEL populations, with the
`exception of TCRγδ-expressing T cells, were mark-
`edly reduced in such an antigen-deprived environ-
`ment62–64. Interestingly, although more than 95% of
`the commensal bacteria in the body normally reside in the
`large intestine, the small intestine contains at least ten
`times more IELs than the colon6. Furthermore, mice
`fed an amino-acid-based, protein-free diet, displayed
`a poorly developed intestinal immune system, simi-
`lar to that of germ-free mice, with a strong decrease
`in most IEL populations65. These effects could be due
`to direct effects of the diet on the immune system or
`could result from diet-induced shaping of the intesti-
`nal microbiota. This highlights the importance of the
`diet, in addition to the microbiota, as a major driv-
`ing factor in establishing and shaping this mucosal
`immune branch.
`
`Migration of natural IELs. In addition to their anti-
`gen-experienced phenotype, natural IEL precursors
`may also acquire expression of gut-homing receptors
`(including αEβ7 integrin and CC-chemokine recep-
`tor 9 (CCR9)) in the thymus21,22,66–68. The expression of
`the ligands for αEβ7 integrin and CCR9, E-cadherin
`and CC-chemokine ligand 25 (CCL25), respectively,
`by small intestinal epithelial cells results in the direct
`
`Lamina propria
`Connective tissue that
`underlies the epithelium of the
`mucosa and contains various
`myeloid and lymphoid cells,
`including macrophages,
`dendritic cells, T cells and
`B cells.
`
`Microbiota
`The microorganisms present
`in normal, healthy individuals.
`These microorganisms live
`mostly in the digestive tract
`but are also found in some
`other tissues.
`
`Germ-free mice
`Mice born and raised in sterile
`isolators. They are devoid of
`colonizing microorganisms,
`but after they have been
`experimentally colonized by
`known bacteria, they are said
`to be gnotobiotic.
`
`448 | JULY 2011 | VOLUME 11
`
` www.nature.com/reviews/immunol
`
`© 2011 Macmillan Publishers Limited. All rights reserved
`
`Genome Ex. 1058
`Page 4 of 12
`
`

`

`Box 2 | The distinct pathways of CD8αα+TCRαβ+ IEL differentiation
`This Review does not aim to discuss the debates surrounding the importance of the
`thymic versus the extrathymic differentiation pathway for CD8αα+ T cell receptor-αβ
`(TCRαβ+) intestinal epithelial lymphocytes (IELs), as this has been extensively reviewed
`elsewhere47,48,168. Here, we give a brief overview of the data that have challenged or
`supported the thymic differentiation pathway. CD8αα+ IELs were originally thought to
`differentiate locally in the gut. This attractive idea was fuelled, in part, by the presence
`of self-reactive TCRs in their TCR repertoire, by supporting data derived from studies
`using athymic mouse models, and by the presence of haematopoietic immature cells
`in the small intestine that display characteristics of T cell precursor cells169–171.
`Nevertheless, ample data indicate that the absolute numbers of CD8αα+TCRαβ+ IELs
`in athymic mice are extremely reduced compared with euthymic mice16,172–174. These
`data therefore support the notion that the vast majority of CD8αα+TCRαβ+ IELs are
`the progeny of cells with a thymic origin. Other data suggest that, under certain
`experimental conditions, immature T cell-committed precursors may leave the
`thymus prematurely, before TCR rearrangements have occurred, and complete their
`maturation locally in the gut175. In conclusion, if indeed the gut contains precursors of
`bone marrow and thymic origin that have the potential under physiological conditions
`to further differentiate locally, multiple crucial questions remain unanswered. For
`example, under what circumstances is this local pathway functional? And how can
`the gut environment support selection? However, there is currently no evidence
`available to answer these questions.
`
`Gut-associated lymphoid
`tissues
`Lymphoid structures and
`aggregates associated with the
`intestinal mucosa, specifically
`the tonsils, Peyer’s patches,
`lymphoid follicles, appendix
`and caecal patch. Enriched in
`lymphocytes and specialized
`dendritic cell and macrophage
`subsets.
`
`Peyer’s patches
`Groups of lymphoid nodules
`present in the small intestine
`(usually the ileum). They occur
`in the intestinal wall, opposite
`the line of attachment of the
`mesentery. They consist of a
`dome area, B cell follicles and
`interfollicular T cell areas. High
`endothelial venules are present
`mainly in the interfollicular
`areas.
`
`Mesenteric lymph nodes
`Lymph nodes, located at the
`base of the mesentery, that
`collect lymph (including cells
`and antigens) draining from the
`intestinal mucosa.
`
`Microfold cells
`(M cells). Specialized
`antigen-sampling cells that are
`located in the follicle-associated
`epithelium of the organized
`mucosa-associated lymphoid
`tissues. M cells deliver antigens
`by transepithelial vesicular
`transport from the
`aero-digestive lumen directly to
`subepithelial lymphoid tissues
`of nasopharynx-associated
`lymphoid tissue and Peyer’s
`patches.
`
`recruitment of mature natural IEL precursors to the
`small intestinal epithelium24,66. Consistent with this,
`the seeding of mucosal tissues with these cells is inde-
`pendent of sphingosine-1-phosphate receptor 1 (S1P1,
`also known as S1PR1), which is required for the migra-
`tion of conventionally-selected T cells69. The direct
`and early population of the mucosal barrier with these
`stress-sensing self-reactive effector cells provides a
`layer of pre-existing immunity before the development
`of antigen-specific immune cells in response to exog-
`enous antigens (FIG. 2). Additional local endogenous
`and exogenous factors, such as the cytokine IL-15 and
`vitamins A and D, further expand and adapt the natu-
`ral IEL compartment. In light of this, a recent study
`showed that vitamin D receptor (VDR)-deficient mice
`had reduced numbers of CD8αα+ IELs and that this
`co incided with low levels of IL-10 in the small intestine
`and increased inflammation in the steady state70.
`
`Migration of induced IELs. One study showed that,
`similarly to natural IELs, CD8αβ+ recent thymic emi-
`grants (RTEs) can directly migrate to the small intesti-
`nal compartment66. However, in general, conventionally
`selected T cells do not express mucosal homing recep-
`tors and naive T cells are normally not detected within
`the intestinal epithelium. Instead, naive T cells acquire
`gut-homing capacity following priming in gut-associated
`lymphoid tissues (GALTs), such as Peyer’s patches, and in
`mesenteric lymph nodes (MLNs)71. Specialized intesti-
`nal epithelial cells or microfold cells (M cells), resident
`CX3C-chemokine receptor 1 (CX3CR1)+ macrophages
`and migratory CD103+ dendritic cells (DCs) all have
`the capacity to sample antigens in the gut and pro-
`mote appropriate T cell responses42,72–75. On priming in
`mucosal sites, naive T cells upregulate homing receptors
`that allow them to enter intestinal tissues in response
`to specific ligands expressed by the tissues76. These
`receptor–ligand pairs include: LFA1 and intercellular
`
`R E V I E W S
`
`adhesion molecule 1 (ICAM1); very late antigen 1 (VLA1;
`also known as α1β1 integrin) and collagen; and α4β7
`integrin and mucosal vascular addressin cell adhe-
`sion molecule 1 (MADCAM1). α4β7 integrin can also
`be induced on T cells primed in other sites, allowing
`peripherally activated cells to migrate to the intestine71.
`Upregulation of CCR9 expression further directs
`T cells to the small intestine compartment in response
`to its ligand, CCL25, which is constitutively expressed
`by intestinal epithelial cells.
`Induction of the receptors that promote migration
`of antigen-experienced T cells to the intestine can be
`promoted by environmental factors that are unique
`to the gut. It was shown that a diet-derived factor,
`the vitamin A metabolite retinoic acid, is an impor-
`tant inducer of gut-homing molecules. Retinoic acid
`promotes upregulation of α4β7 integrin and CCR9,
`thereby directing activated T cells mainly to the small
`intestine77. Although migration to the colon is also
`dependent on α4β7 integrin78, retinoic acid seems to
`be neither necessary nor sufficient to induce migra-
`tion to this site78,79. The ability to produce retinoic acid
`and to imprint gut-homing receptors during T cell
`priming is characteristic of the CCR7+CD103+ migra-
`tory DC subset77, which have been programmed previ-
`ously by retinoic acid from intestinal epithelial cells and
`stromal cells80,81. It is important to note that mucosal
`imprinting is not absolutely required for entering the
`intestinal tissues, and T cells primed in the periph-
`ery also readily migrate to the gut as effector cells.
`However, in response to gut-derived antigens, the
`imprinting process might greatly focus the immune
`response to the mucosal effector site.
`The main adhesion molecule that is involved in the
`specific localization of IELs in the ep

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