throbber
Downloaded from https://academic.oup.com/mend/article/14/5/733/2747718 by Finnegan, Henderson, Farabow, Garrett & Dunner, LLP user on 05 November 2022
`
`Alteration of a Single Amino Acid in
`Peroxisome Proliferator-Activated
`Receptor-a (PPARa) Generates a
`PPARd Phenotype
`
`Ichiro Takada, Ruth T. Yu, H. Eric Xu, Millard H. Lambert,
`Valerie G. Montana, Steven A. Kliewer, Ronald M. Evans, and
`Kazuhiko Umesono*
`
`Graduate School for Biostudies
`Kyoto University (I.T., R.T.Y., K.U.)
`Kyoto 606-8507, Japan
`Glaxo Wellcome Inc. Research and Development (H.E.X., M.H.L.,
`V.G.M., S.A.K.)
`Research Triangle Park, North Carolina 27709
`Howard Hughes Medical Institute (R.M.E.)
`The Salk Institute for Biological Studies
`La Jolla, California 92037
`
`Three pharmacologically important nuclear recep-
`tors, the peroxisome proliferator-activated receptors
`(PPARs a, g, and d), mediate key transcriptional re-
`sponses involved in lipid homeostasis. The PPARa
`and gsubtypes are well conserved from Xenopus to
`man, but the b/d subtypes display substantial spe-
`cies variations in both structure and ligand activation
`profiles. Characterization of the avian cognates re-
`vealed a close relationship between chick (c) aand g
`subtypes to their mammalian counterparts, whereas
`the third chicken subtype was intermediate to Xeno-
`pus (x) band mammalian d, establishing that band d
`are orthologs. Like xPPARb, cPPARb responded ef-
`ficiently to hypolipidemic compounds that fail to ac-
`tivate the human counterpart. This provided the op-
`portunity to address the pharmacological problem as
`to how drug selectivity is achieved and the more
`global evolutionary question as to the minimal
`changes needed to generate a new class of receptor.
`X-ray crystallography and chimeric analyses com-
`bined with site-directed mutagenesis of avian and
`mammalian cognates revealed that a Met to Val
`change at residue 417 was sufficient to switch the
`human and chick phenotype. These results establish
`that the genetic drive to evolve a novel and function-
`ally selectable receptor can be modulated by a single
`amino acid change and suggest how nuclear recep-
`tors can accommodate natural variation in species
`physiology. (Molecular Endocrinology 14: 733–740,
`2000)
`
`0888-8809/00/$3.00/0
`Molecular Endocrinology 14(5): 733–740
`Copyright © 2000 by The Endocrine Society
`Printed in U.S.A.
`
`INTRODUCTION
`
`Lipid and glucose homeostasis involves the coordina-
`tion of signaling pathways mediated by transcription
`factors, among which the peroxisome proliferator-
`activated receptors (PPARs) have been shown to play
`a major role. The PPARs are members of the nuclear
`receptor superfamily of ligand-activated transcription
`factors. Several PPAR subtypes have been described
`and named PPARa, PPARb, PPARg, and PPARd. The
`different forms are expressed in tissue-specific pat-
`terns: PPARa is abundantly found in liver, kidney,
`heart, and muscle; PPARg is localized in fat, large
`intestine, and macrophages; and PPARs b and d are
`widely expressed. The PPARs form a subclass of fatty
`acid and eicosanoid sensors that are characterized by
`their distinct pharmacological profiles, a property that
`has allowed the identification of subtype-selective li-
`gands including the widely used fibrate and thiazo-
`lidinedione classes of drugs (for review, see Refs. 1–4
`and references therein).
`The PPARb and -d forms posed a dilemma as to
`whether they constituted a single group or repre-
`sented distinct subtypes. Since Xenopus PPARb (xP-
`PARb) shares only approximately 75% amino acid
`identity in the ligand-binding domain with mouse and
`human (h) PPARd, it was not clear whether these re-
`ceptors are orthologs or paralogs. This lack of clarity
`was further exacerbated by the finding that human and
`mouse PPARds are functionally distinct from xPPARb
`in their response to ligands (5, 6). To better understand
`the evolutionary relationship between the PPARs, we
`have isolated the chick counterparts as a means for
`providing insight into the ancestral
`form of these
`
`733
`
`Bausch Health Ireland Exhibit 2038, Page 1 of 8
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`Downloaded from https://academic.oup.com/mend/article/14/5/733/2747718 by Finnegan, Henderson, Farabow, Garrett & Dunner, LLP user on 05 November 2022
`
`MOL ENDO · 2000
`734
`
`Vol 14 No. 5
`
`genes after divergence from amphibians. Our results
`demonstrate that chick and Xenopus PPARb and
`mammalian PPARd are orthologs. Moreover, we have
`exploited cross-species differences in the PPARb/d
`subtype to understand the molecular basis for impor-
`tant pharmacological differences in the ligand binding
`properties of the PPARs.
`
`RESULTS AND DISCUSSION
`
`Chick PPAR (cPPAR)-related gene products were ob-
`tained from cDNA libraries prepared from 2.5-day-old
`embryos and adult adipose tissue (see Materials and
`Methods). Isolation and characterization of multiple
`overlapping clones allowed the compilation of full-
`length cDNA sequences for all three cPPARs1 (Fig. 1A,
`GenBank accession nos. AF163809, AF163810, and
`AF163811), and an evolutionary tree comparing the
`ligand-binding domains (LBDs) was constructed using
`the ClustalW program (Ref. 7 and Fig. 1B). The phy-
`logenetic relationships reveal that PPARa and -g are
`highly conserved from Xenopus to human but greater
`divergence exists among the b/d subtypes, with the
`chick counterpart forming an intermediary link be-
`tween Xenopus PPARb and mouse/human PPARd.
`This alignment indicates clearly that the b and d forms
`constitute a single subtype as the conservation within
`individual subtypes is much higher than the similarity
`between that of a given species.
`To characterize the ligand response profiles of the
`cPPARs, we used GAL4 fusions of the LBDs to avoid
`background activation from endogenous PPARs. Se-
`rum-free conditions were chosen as some nuclear re-
`ceptors are modulated by the addition of FBS. The
`chick subtypes were found to exhibit distinct ligand
`response profiles (Fig. 2A; structures of compounds
`are shown in Table 1). Wy-14,643 and eicosatetro-
`enoic acid (ETYA) are selective PPARa activators; in
`chick, ETYA activates PPARa to a greater extent than
`Wy-14,643, as is the case in humans and Xenopus, but
`not in mouse (8, 9). Carbaprostacyclin is active on both
`PPARa and -b (but a .. b) and thiazolidinediones
`(BRL 49653, Glaxo Wellcome, Inc., Research Triangle
`Park, NC) are selective for PPARg as previously re-
`ported (5, 10). Among the fibrate derivatives, bezafi-
`brate and GW2331 (11) were capable of activating
`PPARb; other fibrates (fenofibrate and gemfibrozil)
`were active only on PPARa.
`the fibrates to activate
`This ability of some of
`cPPARb prompted us to compare the effect of beza-
`fibrate, GW2331, and carbaprostacyclin in the mam-
`malian and chick PPARb/ds. As shown in Fig. 2B, the
`responses elicited by bezafibrate and GW2331 were
`distinct between chick (c) and human (h). The potency
`of bezafibrate on cPPARbwas similar to that seen with
`
`1 The sequences reported in this paper have been depos-
`ited in the GenBank database [accession nos. AF163809
`(cPPARa), AF163810 (cPPARb/d), and AF163811 (cPPARg)].
`
`Fig. 1. Comparison of Chick PPARs with Human, Mouse,
`and Xenopus Counterparts
`A, Schematic of the DBDs and LBDs of cPPARs compared
`with the corresponding regions of the human, mouse, and
`Xenopus receptors. Numbers indicate percent identity to the
`corresponding cPPAR. B, Tree plot comparison of human (H),
`mouse (M), chick (C), and Xenopus (X) PPAR LBDs. Human
`RXRa was used as the root for this tree. 0.05 indicates the
`frequency of amino acid change (maximum is 1). The Clustal
`W program (7) was used for alignment.
`
`xPPARb(6). Carbaprostacyclin activated hPPARdand
`cPPARb with equal efficiency.
`To determine whether these compounds could di-
`rectly bind to PPARd/b, we used protease digestion
`assays. Addition of increasing concentrations of tryp-
`sin in the presence of 100 mM bezafibrate or 1 mM
`GW2331 to 35S-labeled cPPARb resulted in the ap-
`pearance of protease-resistant fragments of approxi-
`
`Bausch Health Ireland Exhibit 2038, Page 2 of 8
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`Generation of PPARd Phenotype
`
`735
`
`Downloaded from https://academic.oup.com/mend/article/14/5/733/2747718 by Finnegan, Henderson, Farabow, Garrett & Dunner, LLP user on 05 November 2022
`
`Fig. 2. Transcriptional Activity of GAL-cPPAR Fusion Receptors
`CV-1 cells were cotransfected with a reporter gene containing four copies of a GAL4 binding site (MH-100x4-tk-Luc) in the
`presence or absence of a chimeric receptor (GAL4-cPPARa,b,g). Activation of the luciferase reporter gene was measured in
`relative light units with b-galactosidase activity as a control for transfection efficiency and presented as fold activation. Ligand
`response data are derived from triplicate points from two independent experiments and represented as the mean 6 SE; n5 6.
`A, Comparison of the fold activities of cPPARa, -b, and -g by the indicated compounds. Numbers within brackets represent the
`ligand concentration in micromoles. cPGI, carbaprostacyclin, Wy; Wy-14,643, BF; bezafibrate, FF; fenofibrate, GF; gemfibrozil,
`GW; GW2331, BRL; BRL 49653. B, Chick and human GAL4-PPARb/ds were analyzed in cotransfection assays with bezafibrate,
`GW2331, and carbaprostacyclin. Bezafibrate and GW2331 appear specific for cPPARb. C, 35S-human and chick PPARd/b were
`preincubated for 30 min at 37 C with either 100 mM bezafibrate, 1 mM GW2331, or 100 mM carbaprostacyclin before addition of
`trypsin (final concentrations of 20 and 40 mg/ml, respectively). Proteolytic digestions were carried out at 37 C for 10 min, and then
`samples were denatured and electrophoresed on a 12.5% SDS-polyacrylamide gel. In the presence of bezafibrate and GW2331,
`only cPPARb shows protected fragments (arrows). Addition of carbaprostacyclin results in protected fragments for hPPARd and
`cPPARb (arrows).
`
`Bausch Health Ireland Exhibit 2038, Page 3 of 8
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`Downloaded from https://academic.oup.com/mend/article/14/5/733/2747718 by Finnegan, Henderson, Farabow, Garrett & Dunner, LLP user on 05 November 2022
`
`MOL ENDO · 2000
`736
`
`Vol 14 No. 5
`
`Table 1. Chemical Structures of the Compounds Used in This Study
`
`mately 32 kDa, 29 kDa, and 27 kDa (Fig. 2C, arrows),
`but no protected bands were observed with hPPARb.
`With carbaprostacyclin, protease-resistant fragments
`of similar sizes were observed with both human and
`chick PPARd/b. It is thus apparent that PPARb/d li-
`gands can be classified into those with species-selec-
`tive activity (bezafibrate, GW2331) and those without
`(carbaprostacyclin).
`To determine the region essential for ligand-selec-
`tive recognition by PPARb/d, we examined the struc-
`tures of the chick, Xenopus, mouse, and human ho-
`mologs (12–14). Although cPPARb LBD and xPPARb
`LBD share only 71% amino acid identity (216/303) vs.
`90% (272/303) between chick and human, the ligand
`activation properties of the cPPARbLBD more closely
`resemble those of Xenopus (6, 11). Detailed compar-
`ison of the LBD sequences of cPPARb with those of
`human, mouse, and Xenopus revealed that 200 amino
`acids (a.a.) are conserved with the remaining 103 a.a.
`varying between species.
`
`Taking into consideration the similarity in ligand
`response between chick and Xenopus, we focused
`on 9 a.a. that are conserved between chick and
`Xenopus, but not between chick and human/mouse.
`A series of chimeric human and chick PPARd/b ex-
`pression constructs were made in an attempt to
`further localize the key residues involved in the li-
`gand specification (Fig. 3A). Examination of the re-
`sponse of these receptors to bezafibrate, GW2331
`and carbaprostacyclin indicated that the domain
`spanning from the hinge region to helix 9 is not
`critical
`for
`recognition of either bezafibrate or
`GW2331 by the cPPARb, but that helix 10, contain-
`ing a net change of 3 a.a., was essential for recog-
`nition of both compounds (Fig. 3B).
`During the course of this work, we solved the crystal
`structure of the fibrate GW2331 bound to hPPARd
`(Fig. 4). As in the case of other PPAR ligands, the
`carboxylic acid of GW2331 (see Table 1) was found to
`form an intricate series of hydrogen bonds with histi-
`
`Bausch Health Ireland Exhibit 2038, Page 4 of 8
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`Generation of PPARd Phenotype
`
`737
`
`Downloaded from https://academic.oup.com/mend/article/14/5/733/2747718 by Finnegan, Henderson, Farabow, Garrett & Dunner, LLP user on 05 November 2022
`
`Fig. 3. The C-Terminal Region of cPPAR Is Required for Fibrate-Dependent Activity
`A, Schematic representations of chimeric GAL4-hPPARd/cPPARb fusion proteins (numbers indicate the a.a. position from the
`first methionine). Chimera 1 (open triangle) is a fusion of hPPARd (137–261) and cPPARb (264–443), chimera 2 (open circle)
`encodes a fusion of hPPARd (137–261), cPPARb (264–383), and hPPARd (382–441), and chimera 3 (solid circle) is a fusion of
`hPPARd(137–381) and cPPARb(384–443). B, Cotransfection experiments were performed as described in Fig. 2 with the addition
`of bezafibrate, GW2331, and carbaprostacyclin as indicated. Chimeras 1 and 3 showed response to bezafibrate and GW2331,
`and all constructs were responsive to carbaprostacyclin. Vertical axis represents fold activation.
`
`dine residues in helices 5 and 10 and a tyrosine in the
`AF-2 helix (15, 16). We postulate that this network of
`interactions effectively locks the receptor into a con-
`formation permissive for coactivator interactions. No-
`tably, M417 in helix 10 is bent into an unfavorable
`conformation for accommodation of the gem-dialkyl
`constituent of the GW2331 fibrate headgroup. The
`steric interference between M417 and the fibrate may
`explain the relatively low-affinity binding of GW2331 to
`hPPARd.
`
`Sequence comparison revealed that both the
`chick and Xenopus PPARb subtypes have a valine
`residue at the position analogous to M417. The
`shorter side chain of valine would be expected to
`better accommodate the gem-dialkyl substituent of
`the fibrate headgroup. These data suggested that
`this single residue might be a key determinant in the
`binding of fibrates to PPARs. To test this hypothe-
`sis, we constructed the following mutants (depicted
`in Fig. 5A); one in which M417 of hPPARd was
`
`Bausch Health Ireland Exhibit 2038, Page 5 of 8
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`Downloaded from https://academic.oup.com/mend/article/14/5/733/2747718 by Finnegan, Henderson, Farabow, Garrett & Dunner, LLP user on 05 November 2022
`
`MOL ENDO · 2000
`738
`
`Vol 14 No. 5
`
`Fig. 4. The hPPARd-GW2331 Cocrystal Structure
`Left panel, The hPPARdpolypeptide backbone is shown as a yellow ribbon. The carbon atoms of GW2331 are shown in green.
`The oxygen and fluorine atoms of GW2331 are shown in red and orange, respectively. Right panel, Close-up view of the fibrate
`headgroup of GW2331 bound to the hPPARd LBD. The carbon and oxygen atoms of GW2331 are shown in green and red,
`respectively. The side chains of M416 and M417 are shown with the carbon and sulfur atoms depicted in blue and orange,
`respectively. The side chain of M417 is bent into a high-energy configuration by the gem-dialkyl of GW2331.
`
`changed to valine (hPPARd417V) and the reverse
`where the corresponding valines of cPPARb and
`hPPARa were altered to methionine (cPPARb419M,
`hPPARa444M). Reporter assays confirmed that the
`substitution of a valine confers the ability for a fi-
`brate response to hPPARd417V and, reciprocally,
`replacement of
`the more compact valine with
`methionine results in loss of response to the fibrates
`(Fig. 5B). This alteration in ligand responsiveness
`was found to correlate with changes in ligand
`binding as inferred from protease digestion assays
`using functionally equivalent constructs (data not
`shown).
`In summary, the isolation and characterization of the
`cPPAR homologs have provided an opportunity to
`study how this class of receptors may have evolved.
`The chimeric analyses together with x-ray crystallo-
`graphic studies of
`the fibrate GW2331 bound to
`hPPARd allowed us to localize a difference in ligand
`responsiveness to M417 in helix 10 of the human
`receptor. This work illustrates that a single amino acid
`change may be sufficient to acquire a new ligand
`binding specificity as well as to suppress recognition
`of a previous ligand. Our data agree with and extend
`the observations of others who showed that changes
`in one or several amino acids can result in marked
`alterations in the ligand selectivity of nuclear receptors
`(9, 17). Alteration of a single amino acid would be the
`minimum change necessary to generate a functionally
`distinct receptor. In combination with a preexisting
`gene duplication, this represents the simplest concep-
`tual mechanism for the formation of a new receptor
`gene family.
`
`MATERIALS AND METHODS
`
`Isolation of cPPAR cDNAs
`
`cDNA clones encoding the cPPARs were obtained from
`7-week-old male broiler adipose tissue (Stratagene) and
`stage 17–18 chick embryonic lZAP cDNA libraries (18) with
`mouse PPARg (13) and mouse retinoid X receptor-b (RXRb)
`(19) cDNAs as probes using standard low-stringency hybrid-
`ization procedures. Insert cDNA fragments were recovered
`from purified positive clones into pBluescript vectors for re-
`striction enzyme mapping and DNA sequencing. Full-length
`cDNA sequences for each of three PPAR subtypes were
`assembled and analyzed by DNA sequencing using ALFex-
`press (Pharmacia Biotech, Piscataway, NJ).
`
`Plasmid Construction
`
`Full length coding sequences for cPPARa, PPARb, and PPARg
`were inserted into a pCMX expression vector (20), giving rise to
`pCMX-cPPARa, pCMX-cPPARb/d, and pCMX-cPPARg, re-
`spectively. GAL4 fusions of the PPAR LBDs were prepared by
`PCR amplification of the DNA fragments encoding the respec-
`tive LBDs from corresponding pCMX plasmid templates. Se-
`quences of primers used are TTGGGTTTGTCGACGGAATGT
`CACATAATGCAATACGT (forward) and TTTGGGTTTGGA-
`TCCAAAAATCCTTAATACATG TCCCT (reverse) for PPARa,
`TTGGTTGAATTCGGCATGTCACATAACGCAAT (forward) and
`TTTGGGTTTGTCGACAAGAGGTCCTTAGTACATGTCCTTGTA
`(reverse) for PPARb/d, and TTTGGGTTTGAATTCGGAATGT-
`CACATAATGCCATC (forward) and TGGGGTTTGGATC CGA-
`ACTACTATCGCCATTAATATAAGTC (reverse) for PPARg. The
`amplified DNA fragments were digested with SalI and BamHI for
`PPARa, EcoRI and SalI for PPARband PPARg, and inserted at
`the respective sites in the pCMX-GAL4 derivatives to prepare
`pCMX-GAL4-cPPARa, pCMX-GAL4-cPPARb, and pCMX-
`GAL4-cPPARg. GAL4 fusion constructs for the hPPARs were
`previously described (11).
`
`Bausch Health Ireland Exhibit 2038, Page 6 of 8
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`Generation of PPARd Phenotype
`
`739
`
`Downloaded from https://academic.oup.com/mend/article/14/5/733/2747718 by Finnegan, Henderson, Farabow, Garrett & Dunner, LLP user on 05 November 2022
`
`Fig. 5. A Single Amino Acid Residue Confers Subtype-Specific Ligand Recognition
`Data are represented as the mean 6 SE; n5 6. A, A schematic representation of GAL4-fusion point mutants; hPPARd417V (Met to
`Val change at position 417) and cPPARb419M, hPPARa444M (Val to Met change at positions 419 and 444, respectively). B The above
`constructs were analyzed in cotransfection experiments with 10 mM bezafibrate, 10 nM GW2331, and 10 mM carbaprostacyclin.
`
`Transactivation Assays
`
`Monkey kidney CV-1 cells were used for transfection assays
`in 24-well cluster tissue culture plates by calcium phosphate
`precipitation (21). Transfection mixtures contained 50 ng of
`receptor expression plasmid, 150 ng of MH100x4-tk-luc re-
`porter plasmid, 350 ng of pCMX-bGAL as control for trans-
`fection efficiency, and 200 ng of pGEM4 carrier plasmid.
`Cells were transfected for 7 h, washed, and incubated for
`approximately 36 h in serum-free media containing 5 mg/ml
`insulin, 5 mg/ml transferrin, 0.01% fatty acid free BSA, plus
`ligand compounds where indicated, before harvesting and
`luciferase and b-galactosidase activity. All
`assaying for
`points were performed in triplicate and repeated at least
`twice in independent experiments with variations of less than
`10%.
`
`Mutagenesis
`
`GAL4 fusions of chimeric constructs encoding hPPARd and
`cPPARb LBDs were prepared as follows. Chimera 1 was
`prepared by digestion of cPPARb/d with SacI/BglII and liga-
`tion into respective sites of pCMX-GAL4 hPPARd. Chimera 2
`was prepared by PCR amplification of chimera 1 and ligation
`to the C-terminal region of hPPARd. Sequences of primers
`used are TTTGTCGACGGCATGTCACACAACGCTATCCG
`(forward) and GGACTGCAGGTGGAATTCCAGTG (reverse).
`Amplified DNA fragments were digested with SalI/EcoRI and
`ligation into respective sites of pCMX-GAL4-hPPARd. Chi-
`mera 3 was prepared by PCR amplification from pCMX-
`cPPARb and ligation to pCMX-hPPARd. Primer sequences
`used are CACTGGAATTCCACCTGCAGTCC (forward) and
`TTTGGGTTTGTCGACAAGAGG TCCTTAGTACATGTCCTT-
`
`GTA (reverse). The amplified DNA fragments were digested
`with EcoRI and BglII and inserted at the respective sites in
`pCMX-GAL4-hPPARd.
`GAL4 fusions of the mutant hPPARa and -d and cPPARb
`LBD were prepared by PCR amplification first on N-terminal and
`mutated sites and subsequently on mutated sites and C termini.
`N-terminal and C-terminal primers were used to obtain the
`full-length construct. Primer sequences used for hPPARd417V,
`TTTGTCGACGGCATGTCACACAACGCTATCCG (forward) and
`CCGCTGAACCATCTGGGCGTGCTCG (reverse), were for N-
`terminal fragment; CGAGCACGCCCAGATGGTTCAGCGG (for-
`ward) and TTTGGATCCTTAGTACATGTCCT TGTAGATCTC-
`CTGGAGC (reverse) were for C-terminal fragment. In cPPAR-
`b419M, TGGGTTTGAATTCGGCATGTCACATAACGCAATCC
`(forward) and CTGCATCAGCTGGG CGTGC (reverse) were for
`N-terminal fragment; GCACGCCCAGCTGATGCAG (forward)
`and TTTGGGTTTGTCGACAAGAGGTCCTTAGTACATGTCCT-
`TGTA (reverse) were for C-terminal fragment. In hPPARa444M,
`TTTGGGGGTCGACTCACACAACGCGATTCGTTT TGG (for-
`ward) and CTGCATCAGCTGCGCATGCT (reverse) are for N-
`terminal
`fragment; AGCATGCGCAGCTGATGCAG (forward)
`and TTTGGGGATCCTCAGTACATGTCCCTG TAGATCT (re-
`verse) are for C-terminal fragment. All constructs were con-
`firmed by DNA sequencing using ALFexpress (Pharmacia
`Biotech).
`
`Protease Digestion Assay
`
`35S-radiolabeled proteins were synthesized from 1 mg of
`pCMX-PPARs by the reticulocyte lysate system (Promega
`Corp.). Of the total 40 ml of labeled in vitro translated PPAR
`proteins, 15 ml were preincubated for 30 min at 37 C in 40 ml
`of binding buffer [final 10 mM Tris-HCl, pH 8.0, 80 mM KCl,
`
`Bausch Health Ireland Exhibit 2038, Page 7 of 8
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`Downloaded from https://academic.oup.com/mend/article/14/5/733/2747718 by Finnegan, Henderson, Farabow, Garrett & Dunner, LLP user on 05 November 2022
`
`MOL ENDO · 2000
`740
`
`Vol 14 No. 5
`
`0.1% NP40, 7% glycerol, 1 mM dithiothreitol (DTT)] with ac-
`tivators (bezafibrate, carbaprostacyclin, GW2331) that were
`dissolved in 13 binding buffer. Protease digestion assays
`were initiated by the addition of 2 ml of 53 stock solution of
`trypsin to 8 ml of translation products and carried out for 10
`min at 37 C. Reactions were stopped by addition of 10 ml of
`23 loading buffer (62.5 mM Tris HCl, pH 6.8, 2% SDS, 10%
`glycerol, 50 mM DTT, 5 mg/ml bromophenol blue). Samples
`were loaded and electrophoresed on a 12% acrylamide-SDS
`gel, and the gel was dried under vacuum for 2 h and analyzed
`using BAS Imager (FUJIX, Tokyo, Japan).
`
`Crystallography
`
`The procedures for determining the cocrystal structure of
`GW2331 bound to the hPPARd LBD, including the protein
`purification, crystal growth, and structure refinement, were
`performed as previously described (16). The structure was
`determined at 2.5 A resolution and was refined with an R
`factor of 28.3% (31.4% for the free R), which revealed clear
`electron density for GW2331 and the LBD pocket residues
`surrounding the compound.
`
`Acknowledgments
`
`We thank K. Yasuda for his support during the early part of
`this project, T. Willson, H. Oizumi, and members of the Ume-
`sono laboratory for valuable advice and discussion, and E.
`Stevens for administrative assistance.
`
`Received December 10, 1999. Revision received January
`25, 2000. Accepted January 27, 2000.
`Institute
`Address requests for reprints to: Ruth T. Yu,
`for Virus Research, Kyoto University, 53 Kawaharacho,
`Shogoin, Sakyoku, Kyoto 606-8507, Japan. E-mail: rtyu@
`virus.kyoto-u.ac.jp.
`This work was supported in part by grants from Japan
`Society for Promotion of Science and Human Frontiers Sci-
`ence Program. This paper is dedicated to K. Umesono.
`* Deceased April 12, 1999.
`
`REFERENCES
`
`1. Sorensen HN, Treuter E, Gustafsson JA 1998 Regulation
`of peroxisome proliferator-activated receptors. Vitam
`Horm 54:121–166
`2. Kliewer SA, Lehmann JM, Willson TM 1999 Orphan nu-
`clear receptors: shifting endocrinology into reverse. Sci-
`ence 284:757–760
`3. Berger J, Leibowitz MD, Doebber TW, Elbrecht A, Zhang
`B, Zhou G, Biswas C, Cullinan CA, Hayes NS, Li Y, Tanen
`M, Ventre J, Wu MS, Berger GD, Mosley R, Marquis R,
`Santini C, Sahoo SP, Tolman RL, Smith RG, Moller DE
`1999 Novel peroxisome proliferator-activated receptor
`(PPAR)g and PPARd ligands produce distinct biological
`effects. J Biol Chem 274:6718–6725
`4. Brown PJ, Smith-Oliver TA, Charifson PS, Tomkinson
`NC, Fivush AM, Sternbach DD, Wade LE, Orband-Miller
`L, Parks DJ, Blanchard SG, Kliewer SA 1997 Identifica-
`tion of peroxisome proliferator-activated receptor ligands
`from a biased chemical library. Chem Biol 4:909–18
`5. Forman BM, Chen J, Evans RM 1997 Hypolipidemic
`drugs, polyunsaturated fatty acids, and eicosanoids are
`ligands for peroxisome proliferator-activated receptors a
`and d Proc Natl Acad Sci USA 94:4312–4317
`6. Krey G, Braissant O, L’Horset F, Kalkhoven E, Perroud M,
`Parker MG, Wahli W 1997 Fatty acids, eicosanoids, and
`
`hypolipidemic agents identified as ligands of peroxisome
`proliferator- activated receptors by coactivator-depen-
`dent receptor ligand assay. Mol Endocrinol 11:779–791
`7. Thompson JD, Higgins DG, Gibson TJ 1994 CLUSTAL
`W: improving the sensitivity of progressive multiple se-
`quence alignment through sequence weighting, position-
`specific gap penalties and weight matrix choice. Nucleic
`Acids Res 22:4673–4680
`8. Mukherjee R, Jow L, Noonan D, McDonnell DP 1994
`Human and rat peroxisome proliferator activated recep-
`tors (PPARs) demonstrate similar tissue distribution but
`different responsiveness to PPAR activators. J Steroid
`Biochem Mol Biol 51:157–166
`9. Keller H, Devchand PR, Perroud M, Wahli W 1997 PPARa
`structure-function relationships derived from species-
`specific differences in responsiveness to hypolipidemic
`agents. J Biol Chem 378:651–655
`10. Lehmann JM, Moore LB, Smith-Oliver TA, Wilkison WO,
`Willson TM, Kliewer SA 1995 An antidiabetic thiazo-
`lidinedione is a high affinity ligand for peroxisome prolif-
`erator-activated receptor g ( PPAR g). J Biol Chem 270:
`12953–12956
`11. Kliewer SA, Sundseth SS, Jones SA, Brown PJ, Wisely
`GB, Koble CS, Devchand P, Wahli W, Willson TM, Len-
`hard JM, Lehmann JM 1997 Fatty acids and eicosanoids
`regulate gene expression through direct interactions with
`peroxisome proliferator-activated receptors a and g.
`Proc Natl Acad Sci USA 94:4318–4323
`12. Dreyer C, Krey G, Keller H, Givel F, Helftenbein G, Wahli
`W 1992 Control of the peroxisomal b-oxidation pathway
`by a novel family of nuclear hormone receptors. Cell
`68:879–887
`13. Kliewer SA, Forman BM, Blumberg B, Ong ES, Borg-
`meyer U, Mangelsdorf DJ, Umesono K, Evans RM 1994
`Differential expression and activation of a family of mu-
`rine peroxisome proliferator-activated receptors. Proc
`Natl Acad Sci USA 91:7355–7359
`14. Schmidt A, Endo N, Rutledge SJ, Vogel R, Shinar D,
`Rodan GA 1992 Identification of a new member of the
`steroid hormone receptor superfamily that is activated by
`a peroxisome proliferator and fatty acids. Mol Endocrinol
`6:1634–1641
`15. Nolte RT, Wisely GB, Westin S, Cobb JE, Lambert MH,
`Kurokawa R, Rosenfeld MG, Willson TM, Glass CK, Mil-
`burn MV 1998 Ligand binding and co-activator assembly
`of the peroxisome proliferator-activated receptor-g. Na-
`ture 395:137–43
`16. Xu HE, Lambert MH, Montana VG, Parks DJ, Blanchard
`SG, Brown PJ, Sternbach DD, Lehmann JM, Wisely GB,
`Willson TM, Kliewer SA, Milburn MV 1999 Molecular rec-
`ognition of fatty acids by peroxisome proliferator-acti-
`vated receptor. Mol Cell 3:397–403
`17. Peet DJ, Doyle DF, Corey DR, Mangelsdorf DJ 1998
`Engineering novel specificities for ligand-activated tran-
`scription in the nuclear hormone receptor RXR. Chem
`Biol 5:13–21
`18. Yu RT, McKeown M, Evans RM, Umesono K 1994 Re-
`lationship between Drosophila gap gene tailless and a
`vertebrate nuclear receptor Tlx. Nature 370:375–379
`19. Mangelsdorf DJ, Borgmeyer U, Heyman RA, Zhou JY,
`Ong ES, Oro AE, Kakizuka A, Evans RM 1992 Charac-
`terization of three RXR genes that mediate the action of
`9-cis retinoic acid. Genes Dev 6:329–344
`20. Umesono K, Murakami KK, Thompson CC, Evans RM
`1991 Direct repeats as selective response elements for
`the thyroid hormone, retinoic acid, and vitamin D3 recep-
`tors. Cell 65:1255–1266
`21. Umesono K, Evans RM 1989 Determinants of target gene
`specificity for steroid/thyroid hormone receptors. Cell
`57:1139–1146
`
`Bausch Health Ireland Exhibit 2038, Page 8 of 8
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

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