throbber
on September 28, 2015
` on September 28, 2015
` on September 28, 2015
` on September 28, 2015
` on September 28, 2015
`
`www.sciencemag.org
`www.sciencemag.org
`www.sciencemag.org
`www.sciencemag.org
`www.sciencemag.org
`
`Downloaded from
`Downloaded from
`Downloaded from
`Downloaded from
`Downloaded from
`
`R E P O R T S
`
`8. Materials and methods are available as supporting
`material on Science Online.
`9. D. R. Piperno, D. M. Pearsall, The Origins of Agriculture
`in the Lowland Neotropics (Academic Press, San Di-
`ego, CA, 1998).
`10. T. C. Andres, R. W. Robinson, in Cucurbitaceae 2002,
`D. N. Maynard, Ed. (ASHS Press, Alexandria, VA,
`2002), pp. 95–99.
`11. M. Nee, Econ. Bot. 44, 56 (1990).
`12. Despite extensive searches, no other wild Cucurbita
`has been found in Ecuador (10, 11). C. ecuadorensis is
`believed to have been at least semi-domesticated
`and used more commonly in the past than it is at the
`present time (10, 11). Many free-living populations,
`which are common in dry forest and along streams
`and road banks, have small (⬃8 to 10 cm long) and
`bitter fruits. Vines found today in house gardens,
`where they are grown or maintained and consumed
`by humans or more commonly fed to domestic ani-
`
`mals, have fruits with sizes of domesticated species
`(from 12 to 14 cm long and 12 to 18 cm in diameter)
`and other domesticated characteristics, such as non-
`bitter flesh, nonlignified rinds, and considerable vari-
`ability in color and pattern (10).
`13. S. R. Bozarth, Am. Antiq. 52, 607 (1987).
`14. D. R. Piperno, I. Holst, L. Wessel-Beaver, T. C. Andres,
`Proc. Natl. Acad. Sci. U.S.A. 99, 10923 (2002).
`15. Phytoliths are placed in fruit rinds in such a way that
`they straddle the interface between the hypodermis
`and the outermost stone (lignified) cells. Phytolith
`thickness is greatly influenced by how large the stone
`cells are. Domesticated fruits make longer and larger
`stone cells than do wild fruits, hence phytolith thick-
`ness is also a sensitive indicator of domestication
`(14).
`16. O. I. Sanjur, D. R. Piperno, T. C. Andres, L. Wessel-
`Beaver, Proc. Natl. Acad. Sci. U.S.A. 99, 535 (2002).
`17. D. W. Lathrap, D. Collier, H. Chandra, Ancient Ecua-
`
`dor: Culture, Clay, and Creativity, 3000-300 B.C.
`(Field Museum of Natural History, Chicago, IL, 1975).
`18. D. M. Pearsall, in Dumbarton Oaks Conference on the
`Ecuadorian Formative, J. S. Raymond, R. Burger, Eds.
`(Dumbarton Oaks, Washington, DC, 1996), pp. 213–
`257.
`19. J. R. Harlan, Science 174, 468 (1971).
`20. Supported by the Smithsonian Tropical Research In-
`stitute and the Museo Antropolo´gico, Banco Central
`del Ecuador (Guayaquil).
`
`Supporting Online Material
`www.sciencemag.org/cgi/content/full/299/5609/1054/
`DC1
`Materials and Methods
`Fig. S1
`Table S1
`
`11 November 2002; accepted 26 December 2002
`
`Control of Regulatory T Cell
`Development by the
`Transcription Factor Foxp3
`Shohei Hori,1 Takashi Nomura,2 Shimon Sakaguchi1,2*
`
`Regulatory T cells engage in the maintenance of immunological self-tolerance
`by actively suppressing self-reactive lymphocytes. Little is known, however,
`about the molecular mechanism of their development. Here we show that
`Foxp3, which encodes a transcription factor that is genetically defective in an
`autoimmune and inflammatory syndrome in humans and mice, is specifically
`expressed in naturally arising CD4⫹ regulatory T cells. Furthermore, retroviral
`gene transfer of Foxp3 converts naı¨ve T cells toward a regulatory T cell phe-
`notype similar to that of naturally occurring CD4⫹ regulatory T cells. Thus,
`Foxp3 is a key regulatory gene for the development of regulatory T cells.
`
`reduction or functional alteration in rodents
`leads to the spontaneous development of var-
`ious organ-specific autoimmune diseases in-
`cluding autoimmune thyroiditis, gastritis, and
`type 1 diabetes (6–9). TR cells also appear to
`maintain a balanced response to environmen-
`tal antigens, preventing inflammatory bowel
`disease (IBD) and allergy in rodents (10, 11).
`Several studies have provided findings
`that offer clues to the potential pathway by
`which TR cells develop. Similar multiorgan
`autoimmune diseases, allergy, and IBD de-
`velop in the X-linked recessive disease, IPEX
`(immune dysregulation, polyendocrinopathy,
`enteropathy, X-linked syndrome) or XLAAD
`(X-linked autoimmunity-allergic dysregula-
`tion syndrome) (12). A mouse mutant strain,
`
`To maintain immunological unresponsive-
`ness to self-constituents (i.e., self-tolerance),
`potentially hazardous self-reactive lympho-
`cytes are eliminated or inactivated during
`their development (1). Activation and expan-
`sion of self-reactive T lymphocytes that have
`escaped thymic clonal deletion is actively
`
`suppressed in the periphery by naturally oc-
`curring CD4⫹ regulatory T cells (TR), the
`majority of which constitutively express
`CD25 [interleukin (IL)-2 receptor ␣-chain]
`(2–6). CD25⫹CD4⫹ TR cells are at least in
`part produced by the thymus as a functionally
`mature T cell subpopulation (7, 8), and their
`
`1Laboratory of Immunopathology, Research Center
`for Allergy and Immunology, Institute for Physical and
`Chemical Research, Yokohama 230 – 0045,
`Japan.
`2Department of Experimental Pathology, Institute for
`Frontier Medical Sciences, Kyoto University, Kyoto
`606 – 8507, Japan.
`
`*To whom correspondence should be addressed. E-
`mail: shimon@frontier.kyoto-u.ac.jp
`
`Fig. 1. Expression of Foxp3 in a subpopulation of CD4⫹ T
`cells in the thymus and periphery. (A) BALB/c thymocytes
`were sorted into CD4–CD8– (DN), CD4⫹CD8⫹ (DP), CD4–
`8⫹ (CD8) or CD4⫹8– (CD4) cells (left). CD4⫹8– thymo-
`cytes were further separated into CD25⫹ or CD25– cells
`(right). cDNA from each population was subjected to non-
`saturating PCR using Foxp3- or HPRT (hypoxanthine-
`guanine phosphoribosyl-transferase)–specific primers (21).
`(B) Pooled lymph node and spleen cells from BALB/c mice
`were sorted into the indicated compartments, and nonsat-
`urating RT-PCR analyses were carried out. CD4⫹ cells were
`further separated into CD25⫹ or CD25– cells. (C) Quanti-
`fication of relative Foxp3 mRNA levels in indicated CD4⫹ T
`cell subsets. cDNA samples were subjected to real-time
`quantitative PCR analyses using primers and an internal
`fluorescent probe specific for Foxp3 or HPRT. The relative
`quantity of Foxp3 in each sample was normalized to the
`relative quantity of HPRT (21). (D) CD25–CD4⫹ (open
`symbols) or CD25⫹CD4⫹ cells (closed symbols) were ac-
`tivated for indicated hours with plate-bound CD3 mAb in
`the presence of IL-2 (circles) or CD28 mAb (squares) and
`assessed for the expression of Foxp3 by real-time quanti-
`tative RT-PCR. (A) to (D) each show one representative
`result of three independent experiments.
`
`www.sciencemag.org SCIENCE VOL 299 14 FEBRUARY 2003
`
`1057
`
`Page 1 of 6
`
`YEDA EXHIBIT NO. 2070
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`R E P O R T S
`
`Fig. 2. Retroviral transduction of Foxp3 into naı¨ve CD4⫹ T cells. (A) Foxp3 cDNA was inserted
`into the MIGR1 retrovirus vector (21). This vector simultaneously expresses two cDNAs, Foxp3
`and GFP, with the use of an internal ribosomal entry site (IRES). (B) CD3 mAb–stimulated
`proliferative response of freshly isolated CD25–CD4⫹ cells and GFP⫹ Foxp3/MIGR1- or
`MIGR1-infected CD25–CD4⫹ cells. [3H] thymidine incorporation was measured as an indicator
`of cell proliferation and expressed as the mean (⫾ SD) of triplicate cultures. cpm, counts per
`IFN-␥,
`minute. (C) IL-2,
`IL-4, or IL-10 concentration in the culture supernatant of CD3
`mAb–stimulated culture of GFP⫹ Foxp3/MIGR1- or MIGR1-infected CD25–CD4⫹ cells (mean ⫾
`SD). (D) Foxp3/MIGR1- or MIGR1-infected T cells derived from CD25–CD4⫹ cells were stained
`with phycoerythrin (PE)–labeled mAb for CD25, GITR, CD103, CD4, CD44, or an irrelevant
`antigen (control), along with detection of GFP. Intracellular CTLA-4 molecules were detected
`with PE-labeled CTLA-4 or isotype-matched control mAb (21). (B) to (D) each show one
`representative result of three independent experiments.
`
`scurfy, also succumbs to similar X-linked
`recessive autoimmune and inflammatory dis-
`eases as a result of uncontrolled activation
`and expansion of CD4⫹ T cells (13–16).
`Recent efforts to identify the genetic defect in
`IPEX/XLAAD patients or scurfy mice have
`revealed mutations
`in a common gene,
`Foxp3, which encodes a forkhead-winged–
`helix transcription factor designated Scurfin
`(17–20). Immunological similarities between
`the autoimmunity and inflammation pro-
`duced by manipulating CD25⫹CD4⫹ TR
`cells and those induced by genetic defects in
`Foxp3 prompted us to investigate the possible
`contribution of Foxp3 to the development or
`function of regulatory T cells.
`We first examined the expression of Foxp3
`mRNA in the thymus and the periphery of
`normal mice by reverse transcriptase–polymer-
`ase chain reaction (RT-PCR) (21). In the thy-
`mus, CD25⫹CD4⫹CD8– thymocytes, which
`constitute 5% of CD4⫹CD8– thymocytes (7),
`predominantly transcribed Foxp3, whereas
`CD4–CD8⫹ and other immature thymocyte
`populations did not (Fig. 1A). In the periphery,
`CD4⫹ T cells specifically transcribed the gene,
`whereas CD8⫹ T cells and CD19⫹ B cells did
`not (Fig. 1B) (17). Among CD4⫹ T cells, the
`CD25⫹ subset, which constitutes 5 to 10% of
`CD4⫹ T cells in normal naı¨ve mice, exhibited
`predominant transcription. Real-time quantita-
`tive PCR analyses revealed that the Foxp3
`mRNA level in CD25⫹CD4⫹ cells was 100-
`fold more abundant than in CD25–CD4⫹ cells
`(Fig. 1C). A low level of expression in CD25–
`CD4⫹ cells was confined to the CD45RBlow
`population, which has been previously reported
`to contain regulatory activity (22, 23). These
`results indicate that Foxp3 expression is pre-
`dominantly restricted to the CD25⫹CD4⫹ pop-
`ulation in both the thymus and periphery.
`Expression of Foxp3 is not a mere conse-
`quence of T cell activation, because in vitro
`stimulation of CD25–CD4⫹ cells for 3 days
`by monoclonal antibodies (mAbs) to CD3 in
`the presence of IL-2 or CD28 mAb failed to
`elicit Foxp3 expression (Fig. 1D). Neither
`effector Th1 nor effector Th2 cells prepared
`from naı¨ve T cells expressed Foxp3 (fig. S1).
`Furthermore, stimulation of CD25⫹CD4⫹
`cells with CD3 mAb and IL-2 did not alter
`their expression levels of Foxp3. Thus, Foxp3
`expression is stable in CD25⫹CD4⫹ TR cells
`irrespective of the mode or state of activation.
`We next determined whether forced ex-
`pression of Foxp3 in naı¨ve T cells could
`convert these cells toward a regulatory T cell
`phenotype. Bicistronic retroviral vectors ex-
`pressing Foxp3 and green fluorescent protein
`(GFP)
`(Foxp3/MIGR1)
`or GFP alone
`(MIGR1) were generated (Fig. 2A). Periph-
`eral CD25–CD4⫹ cells from normal naı¨ve
`mice were stimulated with CD3 mAb and
`IL-2 and infected with either retrovirus. One
`week after infection,
`the proliferative re-
`
`1058
`
`14 FEBRUARY 2003 VOL 299 SCIENCE www.sciencemag.org
`
`Page 2 of 6
`
`YEDA EXHIBIT NO. 2070
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`with the level of GFP (Foxp3) expression in the
`infected population [Supporting Online Ma-
`terial (SOM) Text, fig. S3A]. These GFPhigh
`cells also suppressed IL-4 secretion by
`CD25–CD4⫹ responder T cells
`(fig.
`S3B). CD25⫹CD4⫹ cells, whether infected
`with Foxp3/MIGR1 or MIGR1,
`showed
`
`equally potent suppressive activity (Fig. 3A).
`To examine whether antigen-specific na-
`ı¨ve T cells could be converted to regulatory T
`cells by ectopic Foxp3 expression, we used
`recombination-activating gene (RAG)–defi-
`cient DO11.10 CD4⫹ T cells, which express
`ovalbumin (OVA) peptide–specific trans-
`
`R E P O R T S
`
`CD25+CD4+
`
`B
`
`150
`
`IL-2
`
`100
`
`50
`
`pg/ml
`
`CD25-CD4+
`
`0.4
`
`0.3
`
`0.2
`
`0.1
`
`0.0
`
`+ MIG R1
`
`+C D25+
`
`0
`
`C D25- alone
`+ Foxp3/MIG R1
`
`anti-CD3
`
`0.0
`
`0.5
`
`1.0
`
`1.5
`
`2.0
`2.5
`0.0
`0.5
`No. of GFP+ cells (x10-4)
`
`1.0
`
`1.5
`
`2.0
`
`2.5
`
`1.5
`
`1.2
`
`0.9
`
`0.6
`
`0.3
`
`0.0
`
`OVA peptide
`
`1.5
`
`1.2
`
`0.9
`
`0.6
`
`0.3
`
`0.0
`
`D
`
`cpm (x10-5)
`
`DO.RAG-
`
`2.0
`0.0
`0.5
`1.0
`1.5
`No. of GFP+ cells (x10-4)
`
`0.0
`
`0.5
`
`1.0
`
`1.5
`
`2.0
`2.5
`0.0
`0.5
`1.0
`No. of GFP+ cells (x10-4)
`
`1.5
`
`2.0
`
`2.5
`
`1.0
`
`0.4
`
`0.3
`
`0.2
`
`0.1
`
`0.0
`
`1.4
`
`1.0
`
`0.6
`
`0.2
`
`0.1
`
`0.0
`
`A
`
`cpm (x10-5)
`
`C
`
`cpm (x10-5)
`
`E
`
`1.5
`
`1.0
`
`0.5
`
`cpm (x10-5)
`
`0.75
`
`0.50
`
`0.25
`
`0.00
`
`PBS
`
`ααααIL-10R
`
`ααααTGF-ββββ
`
`ααααIL-10R
`+ααααTGF-ββββ
`
`0
`
`C D25- alone
`+Foxp3/MIG R1
`+Foxp3/MIG R1
`(transwell)
`
`Fig. 3. Suppressive activity of Foxp3-transduced naı¨ve CD4⫹ T cells. (A) Graded doses of GFP⫹ cells
`infected with either Foxp3/MIGR1 (closed circles) or MIGR1 (open circles) derived from BALB/c
`CD25–CD4⫹ (left) or CD25⫹CD4⫹ cells (right) were cultured with 2.5 ⫻ 104 freshly prepared
`CD25–CD4⫹ cells for 72 hours with CD3 mAb and 5.0 ⫻ 104 antigen-presenting cells (APCs).
`Proliferation of cells was assessed as in Fig. 2B. Freshly isolated CD25⫹CD4⫹ cells were also
`included in the assay (open squares). (B) Freshly prepared CD25–CD4⫹ cells alone or mixed with
`the same number of GFP⫹ Foxp3/MIGR1-(⫹Foxp3/MIGR1) or MIGR1-infected CD25–CD4⫹ cells
`(⫹MIGR1) or freshly isolated CD25⫹CD4⫹ cells (⫹CD25⫹) were stimulated with CD3 mAb, and
`IL-2 concentration in the culture supernatant was measured as in Fig. 2C. (C) GFP⫹ cells from
`DO11.10/RAG-2–/– CD4⫹ cells infected with Foxp3/MIGR1 (closed circles) or MIGR1 (open circles)
`were cultured with 2.0 ⫻ 104 freshly prepared DO11.10 CD4⫹ cells in the presence of OVA peptide
`and 4.0 ⫻ 104 APCs. (D) BALB/c CD25–CD4⫹ cells were transduced with Foxp3 (closed circles) or
`GFP alone (open circles) and sorted for GFP. Indicated numbers of GFP⫹ cells were mixed with
`2.5 ⫻ 104 DO11.10 TCR transgenic CD4⫹ cells and stimulated with either specific OVA peptide
`(left) or CD3 mAb (right). (E) GFP⫹ Foxp3/MIGR1-infected T cells derived from BALB/c CD25–CD4⫹
`cells and an equal number of freshly prepared CD25–CD4⫹ cells were separated or nonseparated
`by a semipermeable membrane and stimulated with CD3 mAb and APCs on each side (left). IL-10
`receptor (IL-10R) mAb, TGF-␤ mAb, or the mixture of these two was added to the culture of freshly
`prepared CD25–CD4⫹ cells either alone (white bars) or in the presence of GFP⫹ Foxp3/MIGR1-
`infected CD25–CD4⫹ cells (gray bars) or freshly prepared CD25⫹CD4⫹ cells (black bars) (right)
`(21). (A) to (E) each show one representative result of three independent experiments.
`
`sponses of GFP⫹ cells to T cell receptor
`(TCR) stimulation, cytokine production, and
`expression of cell surface molecules were
`examined. Retroviral transduction led to ex-
`pression of GFP in 30 to 60% of CD4⫹ cells.
`Upon TCR stimulation with CD3 mAb,
`GFP⫹ cells from Foxp3/MIGR1-infected cul-
`tures proliferated poorly in contrast to the
`vigorous proliferation of GFP⫹ cells from
`control MIGR1-infected cells or freshly pre-
`pared CD25–CD4⫹ cells (Fig. 2B). In addi-
`tion, Foxp3/MIGR1-infected GFP⫹ cells pro-
`duced very little but detectable IL-2, IFN-␥,
`IL-4, and IL-10 as compared to GFP⫹
`MIGR1-infected cells, which secreted large
`amounts of these cytokines (Fig. 2C). Al-
`though these Foxp3-expressing cells pro-
`duced higher amounts of cytokines than
`freshly isolated CD25⫹CD4⫹ cells (fig. S2),
`cytokine-producing cells were largely con-
`fined to cells expressing low levels of GFP
`(and hence Foxp3).
`Naturally arising CD25⫹CD4⫹ TR cells
`characteristically express CD25, cytotoxic T
`lymphocyte-associated antigen– 4 (CTLA-4),
`glucocorticoid-induced tumor necrosis factor
`receptor family–related gene (GITR), and
`CD103 (␣
`(6–10, 24–28). Al-
`E integrin)
`though the activation of CD25–CD4⫹ cells
`for retroviral infection led to expression of
`these molecules in both Foxp3/MIGR1- and
`MIGR1-infected cells, GFP⫹ cells in the
`former expressed CD25, GITR, and CTLA-4
`at higher levels than GFP– cells or GFP⫹
`MIGR1-infected cells (Fig. 2D, table S1).
`Among the GFP⫹ Foxp3-transduced cells,
`the higher the level of GFP (Foxp3) expres-
`sion, the higher the expression was of these
`molecules
`(Fig. 2D). Furthermore, only
`GFPhigh (Foxp3high) cells in the Foxp3/
`MIGR1-infected cells expressed CD103,
`with no expression on MIGR1-infected cells.
`Because Foxp3 transduction did not affect the
`expression levels of either CD4 or CD44, the
`Foxp3-induced high expression of these TR-
`associated molecules is most likely due to
`specific genetic instruction and not simply
`excessive cell activation by Foxp3. From
`these experiments, we conclude that trans-
`duction of Foxp3 could render naı¨ve T cells
`hyporesponsive to TCR stimulation, inhibit
`cytokine production, and up-regulate expres-
`sion of cell surface molecules closely associ-
`ated with the regulatory function of naturally
`occurring CD25⫹CD4⫹ TR cells (29, 30).
`We next assessed the potential of Foxp3-
`transduced T cells to show suppressive activity.
`Foxp3/MIGR1-infected CD25–CD4⫹ T cells
`could specifically reduce proliferation of fresh-
`ly prepared CD25–CD4⫹ responder T cells
`when stimulated with CD3 mAb (Fig. 3A).
`This suppression was associated with the inhi-
`bition of IL-2 production of the responder pop-
`ulation in a fashion similar to that of natural
`CD25⫹CD4⫹ TR cells (Fig. 3B) and correlated
`
`www.sciencemag.org SCIENCE VOL 299 14 FEBRUARY 2003
`
`1059
`
`Page 3 of 6
`
`YEDA EXHIBIT NO. 2070
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`of T cell regulation. For this, we used a model
`of IBD and autoimmune gastritis that can be
`induced
`in
`severe
`combined
`immuno-
`deficiency (SCID) mice by the transfer of
`CD25–CD45RBhighCD4⫹ T cells from normal
`BALB/c mice and prevented by cotransfer of
`CD25⫹CD4⫹ TR cells (10, 31) (Fig. 4). CD25–
`CD45RBhighCD4⫹ cells from BALB/c mice
`were infected with Foxp3/MIGR1 or MIGR1,
`and GFP⫹ cells were cotransferred with freshly
`prepared CD25–CD45RBhighCD4⫹ cells. The
`GFP⫹ Foxp3-transduced cells inhibited weight
`loss, diarrhea, and histological development of
`colitis and gastritis induced by the transfer of
`CD25–CD45RBhighCD4⫹ cells as effectively
`as naturally occurring CD25⫹CD4⫹ cells. By
`contrast,
`the cells transduced with GFP
`alone failed to prevent disease, and rather
`enhanced weight loss and the development
`of colitis. Thus, transduction of Foxp3 can
`render naı¨ve T cells capable of preventing
`autoimmune gastritis and IBD caused by
`dysregulated immune responses
`toward
`gastric self-antigens and commensal bacte-
`ria, respectively (SOM Text).
`The present study shows that Foxp3 is pre-
`dominantly expressed in the CD25⫹CD4⫹ TR
`population naturally arising in the thymus and
`periphery and that Foxp3 expression in naı¨ve T
`cells can convert these cells to a regulatory T
`cell phenotype functionally similar to naturally
`occurring CD25⫹CD4⫹ TR cells. This result
`suggests that Foxp3 may be a master regulatory
`gene for cell-lineage commitment or develop-
`mental differentiation of regulatory T cells in
`the thymus and the periphery. Our results also
`indicate that, in defining the naturally occurring
`CD4⫹ regulatory T cells engaged in preventing
`autoimmune disease and immunopathology,
`Foxp3 represents a more specific marker than
`currently used cell-surface molecules (such as
`CD25, CD45RB, CTLA-4, and GITR), which
`are unable to completely discriminate between
`regulatory T cells and activated, effector, or
`memory T cells.
`Mutations in the Foxp3 gene culminate in
`the development of a fatal lymphoprolifera-
`tive disorder associated with multiorgan pa-
`thology both in mice and humans (12–20).
`FOXP3 is predominantly expressed in human
`CD25⫹CD4⫹ T cells as well (32). Further-
`more, transduction of a mutant Foxp3 lacking
`the forkhead domain, similar to the mutated
`Foxp3 in scurfy mice (17), failed to confer
`suppressive activity to CD25–CD4⫹ T cells
`(fig. S7). The present results therefore sug-
`gest that mutations of the Foxp3 gene may
`cause
`these disorders
`through develop-
`mental or
`functional abnormality of
`the
`CD25⫹CD4⫹ TR population.
`Potentially, generation of TR cells by Foxp3
`transduction of naı¨ve T cells may provide a
`previously unstudied therapeutic mode for
`treatment of autoimmune and inflammatory
`diseases and in transplantation tolerance.
`
`R E P O R T S
`
`natural
`in
`deficient
`are
`genic TCRs,
`CD25⫹CD4⫹ TR cells (7), and scarcely ex-
`press Foxp3 (SOM Text, fig. S4). In the
`absence of specific antigen, almost all T cells
`from these animals remain in a naı¨ve state.
`Transgenic CD4⫹ T cells infected with
`Foxp3/MIGR1 suppressed the proliferation
`of freshly prepared noninfected transgenic T
`cells upon stimulation with specific OVA
`peptides, whereas those infected with MIGR1
`did not (Fig. 3C). These results collectively
`indicate that ectopic expression of Foxp3 was
`sufficient to convert otherwise nonregulatory
`naı¨ve T cells toward a regulatory T cell phe-
`notype capable of suppressing proliferation
`of other T cells, presumably through inhibi-
`tion of IL-2 production (SOM Text, fig. S5).
`Foxp3-transduced CD4⫹ T cells appeared
`to exert the suppressive activity in a similar
`manner
`to
`that
`of
`naturally
`occurring
`CD25⫹CD4⫹ TR cells (SOM Text, fig. S6).
`First, Foxp3-transduced cells derived from
`BALB/c CD25–CD4⫹ cells failed to suppress
`the proliferative response of DO11.10 TCR
`transgenic CD4⫹ T cells when stimulated with
`
`OVA peptide, whereas polyclonal stimulation
`with CD3 mAb induced suppression (Fig. 3D).
`This indicates that Foxp3-transduced T cells
`require stimulation through TCRs to exert sup-
`pression (29, 30). Second, Foxp3-infected T
`cells failed to suppress other T cells when sep-
`arated by a semipermeable membrane, in con-
`trast to effective suppression that was observed
`when cell-cell contact was allowed (Fig. 3E).
`These separated Foxp3-infected T cells even
`enhanced T cell responses across the mem-
`brane, presumably via cytokines they pro-
`duced (fig. S2). In addition, neutralization of
`transforming growth factor–␤ (TGF-␤) or
`blocking of IL-10 receptor, either alone or in
`combination, failed to abrogate suppression,
`as was the case with natural CD25⫹CD4⫹ TR
`cells. These results indicate that in vitro sup-
`pression by Foxp3-transduced T cells may
`not be mediated by humoral factors but re-
`quires cell contact (29, 30).
`Finally, we examined whether Foxp3/
`MIGR1-infected T cells could suppress in vivo
`the inflammation and the autoimmune disease
`that have been shown to develop in the absence
`
`3
`
`2
`
`1
`
`0
`
`gastritis score
`
`012345
`
`C
`
`colitis score
`
`*
`
`*
`
`*
`
`*
`
`*
`
`0
`
`1
`
`2
`3
`4
`5
`Weeks after transfer
`
`6
`
`7
`
`None
`
`25-45R Bhi alone
`+Foxp3/MIG R1
`+ MIG R1
`
`25-45R Bhi alone
`+Foxp3/MIG R1
`+ MIG R1
`
`None
`
`CD25-CD45RBhi alone
`
`+Foxp3/MIGR1
`
`+MIGR1
`
`None
`
`A
`
`120
`
`110
`
`100
`
`90
`
`80
`
`70
`
`60
`
`% of initial body weight
`
`B
`
`Colon
`
`Stomach
`
`Fig. 4. Prevention of IBD and autoimmune gastritis by Foxp3-transduced T cells. (A) C.B-17 scid
`mice received 4 ⫻ 105 fresh CD25–CD45RBhighCD4⫹ cells either alone (n ⫽ 6, where n is the
`number of mice) (open squares) or together with 1.2 ⫻ 106 GFP⫹ sorted cells derived from
`CD25–CD45RBhighCD4⫹ cells infected with Foxp3/MIGR1 (n ⫽ 7) (closed circles) or MIGR1 (n ⫽ 5)
`(open circles). Body weight is represented as the percentage of initial weight (mean ⫾ SD).
`Astericks indicate significant difference, P ⬍ 0.01, Foxp3/MIGR1 versus other two groups by
`Mann-Whitney test. (B) Histopathology of the colon and stomach in each group and in an
`unreconstituted SCID mouse (None). (C) Colitis (left) and gastritis (right) were histologically
`scored. Two mice in the group cotransferred with MIGR1-infected cells and one transferred with
`CD25–CD45RBhighCD4⫹ cells alone died of debilitation before histological examination. Results
`shown in (A) to (C) are from a total of three independent experiments.
`
`1060
`
`14 FEBRUARY 2003 VOL 299 SCIENCE www.sciencemag.org
`
`Page 4 of 6
`
`YEDA EXHIBIT NO. 2070
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`R E P O R T S
`
`31. E. Suri-Payer, H. Cantor, J. Autoimmun. 16, 115
`(2001).
`32. S. Hori, T. Nomura, S. Sakaguchi, unpublished data.
`33. We thank K. J. Wood, Z. Fehervari, and T. Takahashi
`for critically reading the manuscript; W. S. Pear and T.
`Kitamura for reagents; and T. Matsushita for histol-
`ogy. Supported by grants-in-aid from the Ministry of
`Education, Sports and Culture, the Ministry of Human
`Welfare, and the Organization for Pharmaceutical
`Safety and Research of Japan.
`
`Supporting Online Material
`www.sciencemag.org/cgi/content/full/1079490/DC1
`Materials and Methods
`SOM Text
`Figs. S1 to S7
`Table S1
`References
`
`17 October 2002; accepted 23 December 2002
`Published online 9 January 2003;
`10.1126/science.1079490
`Include this information when citing this paper.
`
`placed with a heterologous protein-protein
`interaction (Fig. 2A): the well-characterized
`heterodimerization interaction between PDZ
`domains from the mammalian proteins neu-
`ronal nitric oxide synthase (nNOS) and syn-
`trophin (13, 14). PDZ domains are interaction
`modules involved in assembly of diverse sig-
`naling complexes in higher eukaryotes (15,
`
`References and Notes
`1. L. Van Parijs, A. K. Abbas, Science 280, 243 (1998).
`2. S. Sakaguchi, Cell 101, 455 (2000).
`3. K. J. Maloy, F. Powrie, Nature Immunol. 2, 816 (2001).
`4. A. Coutinho, S. Hori, T. Carvalho,
`I. Caramalho, J.
`Demengeot, Immunol. Rev. 182, 89 (2001).
`5. E. M. Shevach, Nature Rev. Immunol. 2, 389 (2002).
`6. S. Sakaguchi, N. Sakaguchi, M. Asano, M.
`Itoh, M.
`Toda, J. Immunol. 155, 1151 (1995).
`7. M. Itoh et al., J. Immunol. 162, 5317 (1999).
`8. M. Asano, M. Toda, N. Sakaguchi, S. Sakaguchi, J. Exp.
`Med. 184, 387 (1996).
`9. E. Suri-Payer, A. Z. Amar, A. M. Thornton, E. M.
`Shevach, J. Immunol. 160, 1212 (1998).
`10. S. Read, V. Malmstrom, F. Powrie, J. Exp. Med. 192,
`295 (2000).
`11. M. A. Curotto de Lafaille et al., J. Exp. Med. 194, 1349
`(2001).
`12. R. S. Wildin, S. Smyk-Pearson, A. H. Filipovich, J. Med.
`Genet. 39, 537 (2002).
`13. V. L. Godfrey, J. E. Wilkinson, L. B. Russell, Am. J.
`Pathol. 138, 1379 (1991).
`
`14. P. J. Blair et al., J. Immunol. 153, 3764 (1994).
`15. L. B. Clark et al., J. Immunol. 162, 2546 (1999).
`16. J. L. Zahorsky-Reeves, J. E. Wilkinson, Eur. J. Immunol.
`31, 196 (2001).
`17. M. E. Brunkow et al., Nature Genet. 27, 68 (2001).
`18. T. A. Chatila et al., J. Clin. Investig. 106, R75 (2000).
`19. R. S. Wildin et al., Nature Genet. 27, 18 (2001).
`20. C. L. Bennett et al., Nature Genet. 27, 20 (2001).
`21. Materials and methods are available as supporting
`material on Science Online.
`22. L. A. Stephens, D. Mason, J. Immunol. 165, 3105 (2000).
`23. O. Annacker et al., J. Immunol. 166, 3008 (2001).
`24. T. Takahashi et al., J. Exp. Med. 192, 303 (2000).
`25. B. Salomon et al., Immunity 12, 431 (2000).
`26. J. Shimizu, S. Yamazaki, T. Takahashi, Y.
`Ishida, S.
`Sakaguchi, Nature Immunol. 3, 135 (2002).
`27. R. S. McHugh et al., Immunity 16, 311 (2002).
`28. J. Lehmann et al., Proc. Natl. Acad. Sci. U.S.A. 99,
`13031 (2002).
`29. T. Takahashi et al., Int. Immunol. 10, 1969 (1998).
`30. A. M. Thornton, E. M. Shevach, J. Exp. Med. 188, 287
`(1998).
`
`Rewiring MAP Kinase Pathways
`Using Alternative Scaffold
`Assembly Mechanisms
`
`Sang-Hyun Park, Ali Zarrinpar, Wendell A. Lim*
`
`How scaffold proteins control information flow in signaling pathways is poorly
`understood: Do they simply tether components, or do they precisely orient and
`activate them? We found that the yeast mitogen-activated protein (MAP) kinase
`scaffold Ste5 is tolerant to major stereochemical perturbations; heterologous pro-
`tein interactions could functionally replace native kinase recruitment interactions,
`indicating that simple tethering is largely sufficient for scaffold-mediated sig-
`naling. Moreover, by engineering a scaffold that tethers a unique kinase set, we
`could create a synthetic MAP kinase pathway with non-natural input-output
`properties. These findings demonstrate that scaffolds are highly flexible orga-
`nizing factors that can facilitate pathway evolution and engineering.
`
`Scaffold proteins are known to play a critical
`role in a growing number of signaling path-
`ways, including several mitogen-activated pro-
`tein kinase (MAPK) cascades (1–4). In the
`budding yeast Saccharomyces cerevisiae, the
`scaffold proteins Ste5 and Pbs2 are essential for
`the mating and high–osmolarity response
`MAPK pathways, respectively (5–8). These
`scaffold proteins contain binding sites for each
`of the pathway kinases, as well as for upstream
`signaling input proteins (Fig. 1).
`Despite their importance, little is known
`about the mechanism by which scaffold pro-
`teins such as Ste5 contribute to efficient and
`specific signaling (9). One model is that scaf-
`fold proteins simply tether pathway compo-
`nents, increasing their likelihood of acting on
`one another. However, one might expect a
`simple tethering scaffold to enhance but not
`
`Department of Cellular and Molecular Pharmacology
`and Department of Biochemistry and Biophysics, Uni-
`versity of California, 513 Parnassus Avenue, San Fran-
`cisco, CA 94143, USA.
`
`*To whom correspondence should be addressed. E-
`mail: wlim@itsa.ucsf.edu
`
`be required for signaling. Thus, because Ste5
`is essential for signaling, and because of ev-
`idence supporting conformational changes in-
`duced by scaffold-kinase association (10, 11),
`an alternative model is that Ste5 plays a more
`complex catalytic role, precisely orienting
`and/or allosterically regulating pathway ki-
`nases (4). One way to distinguish between
`these models would be to probe pathway
`sensitivity to perturbations
`in assembly
`mechanisms. If pathway function depended
`on precise catalytic participation of the scaf-
`fold, then strict stereochemical requirements
`for kinase recruitment would be expected.
`We therefore tested whether non-native
`protein-protein interactions could be used to
`build a scaffolded assembly capable of me-
`diating proper mating pathway connectivity
`and function. We took advantage of several
`known mutations in Ste5 that selectively de-
`stroy recruitment of the MAPK kinase kinase
`(MAPKKK) Ste11 and the MAPK kinase
`(MAPKK) Ste7. These mutations, respective-
`ly termed Ste5* and Ste5**, each resulted in
`a nonfunctional mating pathway (12). Defec-
`tive recruitment interactions were then re-
`
`Fig. 1. Yeast mating and high-osmolarity MAPK
`pathways require scaffold proteins Ste5 and
`Pbs2. Both pathways require the shared MAP-
`KKK Ste11 but exhibit no cross-signaling under
`normal conditions. Ste5 has distinct docking
`sites for Ste11, the MAPKK Ste7, and the MAPK
`Fus3 (or the partially redundant MAPK Kss1,
`not shown for simplicity) (5–7). Input to Ste5
`occurs through a docking site for Ste4, the G␤
`subunit of the heterotrimeric guanine nucleo-
`tide–binding protein activated upon phero-
`mone binding to the ␣-factor receptor, Ste2.
`Pbs2 functions as both the scaffold and MAPKK
`of the osmolarity pathway:
`It has a MAPKK
`domain, and it has been shown to bind Ste11
`and the MAPK Hog1 ( precise binding sites have
`not been identified) (8). Pbs2 also binds the Src
`homology 3 (SH3) domain from the upstream
`osmosensor Sho1 through a proline-rich dock-
`ing site (residues 94 to 100), indicated by PxxP
`(26). A second branch of the osmoresponse
`pathway involving the two-component sensor
`protein Sln1 has been omitted for simplicity
`(27). This branch of the pathway does not
`require Sho1 or Ste11. All of the studies de-
`scribed here were performed with strains lack-
`ing this pathway branch (ssk2⌬ and ssk22⌬).
`
`www.sciencemag.org SCIENCE VOL 299 14 FEBRUARY 2003
`
`1061
`
`Page 5 of 6
`
`YEDA EXHIBIT NO. 2070
`MYLAN PHARM. v YEDA
`IPR2015-00644
`
`

`
`Control of Regulatory T Cell Development by the Transcription Factor
`Foxp3
` et al.
`Shohei Hori
` 299 Science
`
`, 1057 (2003);
`DOI: 10.1126/science.1079490
`
`
`
` This copy is for your personal, non-commercial use only.
`
` on September 28, 2015
`
`www.sciencemag.org
`
`Downloaded from
`
`If you wish to distribute this article to others
`, you can order high-quality copies for your
`
`clicking here.
`colleagues, clients, or customers by
`
`Permission to republish or repurpose articles or portions of articles
`
` here.
`following the guidelines
`
` can be obtained by
`
`The following resources related to this article are available online at
`September 28, 2015
`
` www.sciencemag.org (this information is current as of
`
` ):
`
`
`
`including high-resolution figures, can be found in the onlineUpdated information and services,
`version of this article at:
` http://www.sciencemag.org/content/299/5609/1057.full.html
`
`
`
`can be found at: Supporting Online Material
`
`http://www.sciencemag.org/content/suppl/2003/02/13/1079490.DC1.html
`
`
`
`related to this article A list of selected additional articles on the Science Web sites
`found at:
` http://www.sciencemag.org/content/299/5609/1057.full.html#related
`
`
`
`can be
`
`This article , 16 of which can be accessed free:cites 30 articles
`
`
` http://www.sciencemag.org/content/299/5609/1057.full.html#ref-list-1
`
`
`
`
`cited by This article has been
`
`2284 article(s) on the ISI Web of Science
`
`cited by This article has been
`
`100 articles hosted by HighWire Press; see:
`
` http://www.sciencemag.org/content/299/5609/1057.full.html#related-urls
`
`
`
`subject collections:This article appears in the following
`Immunology
` http://www.sciencemag.org/cgi/collection/immunology
`
`
`
`(print ISSN 0036-8075; online ISSN 1095-9203) is published

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