throbber
CFAD V. Anacor, |PR201 5-01776 ANACOR EX. 2094 - “I/“I4
`
`CFAD v. Anacor, IPR2015-01776 ANACOR EX. 2094 - 1/14
`
`

`
`Editor-in-Chief
`
`Norton B. Gilula
`Department of Cell Biology, MB6, The Scripps Research Institute, 10550 North
`Torrey Pines Rd., La Jolla, CA 92037. Tel.: (619) 784-9880. Fax: (619) 784-2345.
`E-mail: thejcb@scripps.edu
`
`Associate Editors
`Don W. Cleveland
`Ludwig Institute for Cancer Research, University of California at San Diego,
`Room 3080 CMM-East, Mailstop 0660, 9500 Gilman Drive, La Jolla, CA 92093.
`Tel.: (619) 534-7870. Fax: (619) 534-7659. E-mail: jcb@ucsd.edu
`
`Werner Franke
`Division of Cell Biology, German Cancer Research Center, Im Neuenheimer
`Feld 280,69120 Heidelberg, F.R.G. Tel.: (49) 6221 42 3451.
`Fax: (49) 6221 42 3452. E-mail: jcb@dkfz-heidelberg.de
`
`Elaine Fuchs
`The University of Chicago, 5841 Maryland Ave. N-314, M/C 1028, Chicago, IL
`60637. Tel.: (773) 702-1347. Fax: (773) 702-0141. E-mail: nliptak@midway.
`uchicago.edu
`
`Ira Mellman
`Department of Cell Biology, Yale University School of Medicine, 333 Cedar
`Street, P.O. Box 208002, New Haven, CT 06520. Tel.: (203) 785-4302.
`Fax: (203) 785-4301. E-mail: ira.mellman@yale.edu
`
`Tl1e Journal of
`
`Volume 143, Number 7,
`December 28, 1998
`
`W. James Nelson
`Department of Molecular and Cellular Physiology, Beckman Center B 121,
`Stanford University School of Medicine, Stanford. CA 94305. Tel.: (650)
`725-7596. Fax: (650) 498-5286. E-mail: wjnelson@leland.stanford.edu
`Louis F. Reichardt
`Neuroscience Program, Howard Hughes Medical Institute, Departments of
`Physiology and Biochemistry & Biophysics, University of California, Rm. U426,
`San Francisco, CA 94143. Tel.: (415) 476-3976. Fax: (415) 476-9914. E-mail:
`lfr@cgl.ucsf.edu
`Randy Schekman
`Department of Molecular and Cell Biology, University of California, 401 Barker
`Hall, Berkeley, CA 94720. Tel.: (510) 643-8358. Fax: (510) 643-8493.
`E-mail: schekman@uclink4.berkeley.edu
`Kenneth M. Yamada
`Craniofacial Developmental Biology and Regeneration Branch, National
`Institute of Dental Research, National Institutes of Health, Building 30, Room
`421, 30 Convent Drive MSC 4370, Bethesda, MD 20892-4370. Fax: (301) 365-2656.
`E-mail: yamada@bellatlantic.net
`
`Editorial Board
`Kathryn Anderson
`Richard G.W. Anderson
`Vann Bennett
`Marianne Bronner-Fraser
`Eric J. Brown
`Lewis Cantley
`John S. Condeelis
`Caroline Damsky
`Pietro De Camilli
`Elisabetta Dejana
`David J. DeRosier
`
`David Drubin
`William Earnshaw
`Scott Emr
`Larry Gerace
`Larry Goldstein
`Alan Hall
`Ulrich Hartl
`Nobutaka Hirakawa
`H. Robert Horvitz
`Arthur Horwich
`
`Alan Rick Horwitz
`Tony Hunter
`Keith Joiner
`Eric Karsenti
`Hynda Kleinman
`Arthur D. Lander
`Jeanne Lawrence
`Leslie Leinwand
`Joan Mass ague
`Jacopo Meldolesi
`
`Timothy J. Mitchison
`Randall T. Moon
`Andrew Murray
`Bjorn Olsen
`Leslie Parise
`Mu-ming Poo
`Martin Raff
`Natasha V. Raikhel
`Tom A. Rapoport
`Howard Riezman
`
`Daniel B. Rifkin
`Mark Rose
`Joshua Sanes
`Richard Scheller
`Trina Schroer
`Jean Schwarzbauer
`Masatoshi Takeichi
`Shoichiro Tsukita
`Graham Warren
`Marino Zerial
`
`Managing Editor
`Michael T. Rossner
`The Rockefeller University Press, 1114 First Avenue, New York, NY 10021.
`Tel.: (212) 327-8881. Fax: (212) 327-8576. E-mail: rossner@rockvax.rockefeller.edu
`
`Editorial Assistants
`
`Sati Motieram, Adaobi Obi, Leslie Thomas,
`and Brandy Murphy
`Tel.: (212) 327-8581. Fax: (212) 327-8576. E-mail: jcellbiol@rockvax.rockefeller.edu
`
`Copyediting and Production
`
`Justin Collins, Davide R. Dickson,
`and C. Kelly White
`Tel.: (212) 327-8552. Fax: (212) 327-8513. E-mail: jcb@rockvax.rockefeller.edu
`
`Published biweekly by The Rockefeller University Press
`
`The appearance of the code at the bottom of the first page of an article in this journal indicates that the Publisher gives consent for individual copies of that
`article to be made for personal or internal use. This consent is given on the condition, however, that-for copying beyond the limited quantities permitted
`under Sections 107 or 108 of the U.S. Copyright Law-the copier pay the stated per-copy fee, for this journal $2.00 per article, through the Copyright
`Clearance Center, Inc., 222 Rosewood Drive, Danvers, MA 01923, Tel.: (978) 750-8400, a nonprofit organization. This consent does not extend to other kinds
`of copying, such as copying for general distribution, for advertising or promotional purposes, for creating new collective works, or for resale.
`The Journal of Cell Biology (ISSN 0021-9525) is published biweekly for $715 (outside USA/Canada add $145 net postage) by The Rockefeller University
`Press, 1114 First Avenue, 4th Floor, New York, NY 10021. Periodical postage paid at New York, NY and additional mailing offices. POSTMASTER: Send
`address changes to The Joumal of Cell Biology, Circulation Department, The Rockefeller University Press, 1114 First Avenue, 4th Floor, New York, NY
`10021. Copyright© 1998 by The Rockefeller University Press, 1114 First Avenue, 4th Floor, New York, NY 10021.
`Subscription rates for 1998: Full paid $715, Individual $165, ASCB Society Members $125, Student $82.50, Air mail surcharge $145, Single copy $27.50.
`Please contact the ASCB for membership information: ASCB, 9650 Rockville Pike, Bethesda, MD 20814. Tel.: (301) 530-7153.
`
`CFAD v. Anacor, IPR2015-01776 ANACOR EX. 2094 - 2/14
`
`

`
`Contents:
`The Journal of Cell Biology
`Volume 143, Number 7, December 28, 1998
`
`In Brief
`Movement by cable or capture. Gathering
`together unfolded proteins. Not all sarcoglycans
`are equal. Nuclear pore complexes and spindle
`pole bodies share a component. Early functions
`for desmosomes. A sex -specific homeodomain
`protein in algae.
`W.A. Wells
`
`Regular Articles ·
`INCENP centromere and spindle targeting:
`Identification of essential conserved motifs and
`involvement of heterochromatin protein HP1.
`A.M. Ainsztein, S.E. Kandels-Lewis, A.M. Mackay,
`and W.C. Earnshaw
`
`1763
`
`177 5 Fission yeast Bub 1 is a mitotic centromere
`protein essential for the spindle checkpoint and
`the preservation of correct ploidy through
`mitosis.
`P. Bernard, K. Hardwick, and J.-P. Javerzat
`
`1789 Saccharomyces cerevisiae Ndclp is a shared
`component of nuclear pore complexes and
`spindle pole bodies.
`H.J. Chial, M.P. Rout, T.H. Giddings, Jr.,
`and M. Winey
`
`1801 Functional analysis of Tpr: Identification of
`nuclear pore complex association and nuclear
`localization domains and a role in mRNA
`export.
`P. Bangs, B. Burke, C. Powers, R. Craig, A. Purohit,
`and S. Doxsey
`
`1813 Specific binding of the karyopherin Kap121p to
`a subunit of the nuclear pore complex containing
`Nup53p, Nup59p, and Nup170p.
`M. Marelli, J.D. Aitchison, and R.W. Wozniak
`
`1831 Homotypic fusion of immature secretory
`granules during maturation in a cell-free assay.
`S. Urbe, L.J. Page, and S.A. Tooze
`
`1845 Biochemical and functional studies of cortical
`vesicle fusion: The SNARE complex and Ca2+
`sensitivity.
`J.R. Coorssen, P.S. Blank, M. Tahara,
`and J. Zimmerberg
`
`1859 Pex18p and Pex21p, a novel pair of related
`peroxins essential for peroxisomal targeting by
`the PTS2 pathway.
`P.E. Purdue, X. Yang, and P.B. Lazarow
`
`1871 Redundant and distinct functions for dynamin-1
`and dynamin-2 isoforms.
`Y. Altschuler, S.M. Barbas, L.J. Terlecky, K. Tang,
`S. Hardy, K.E. Mostov, and S.L. Schmid
`
`1883 Aggresomes: A cellular response to misfolded
`proteins.
`J.A. Johnston, C.L. Ward, and R.R. Kopito
`
`1899 Visualization of melanosome dynamics within
`wild-type and dilute melanocytes suggests a
`paradigm for myosin V function in vivo.
`X. Wu, B. Bowers, K. Rao, Q. Wei,
`and J.A. Hammer III
`
`1919 Visualization and molecular analysis of actin
`assembly in living cells.
`D.A. Schafer, M.D. Welch, L.M. Machesky,
`P.C. Bridgman, S.M. Meyer, and J.A. Cooper
`
`1931 Tropomyosin-containing actin cables direct the
`Myo2p-dependent polarized delivery of secretory
`vesicles in budding yeast.
`D.W. Pruyne, D.H. Schott, and A. Bretscher
`
`194 7 Dual function of Cyk2, a cdc15/PSTPIP family
`protein, in regulating actomyosin ring dynamics
`and septin distribution.
`J. Lippincott and R. Li
`
`1961 Nonuniform microtubular polarity established
`by CH01/MKLP1 motor protein is necessary for
`process formation of podocytes.
`N. Kobayashi, J. Reiser, W. Kriz, R. Kuriyama,
`and P. Mundel
`
`1971 A gamete-specific, sex-limited homeodomain
`protein in Chlamydomonas.
`V. Kurvari, N.V. Grishin, and W.J. Snell
`
`Contents continued
`
`Cover picture: An ES .0 day mouse embryo was labeled with E-cadherin
`(red), desmoplakin (green), and DAPI (blue) and viewed using confocal
`microscopy. Adherens junctions are located between cells of the extraem(cid:173)
`bryonic tissues (ectoplacental cone and visceral endoderm) and embry(cid:173)
`onic tissue (primitive ectodermal), while desmosomes at this stage are lo(cid:173)
`cated only between cells in the extraembryonic tissues. See related article
`in this issue by Gallicano et al., 2009-2022.
`
`CFAD v. Anacor, IPR2015-01776 ANACOR EX. 2094 - 3/14
`
`

`
`1981 ARFl 1nediates paxillin recruitment to focal
`adhesions and potentiates Rho-stimulated stress
`fiber formation in intact and permeabilized Swiss
`3T3 fibroblasts.
`J.C. Norman, D. Jones, S.T. Barry, M.R. Holt,
`S. Cockcroft, and D.R. Critchley
`
`1997
`
`Regulation of the cell cycle by focal adhesion
`kinase.
`J.-H. Zhao, H. Reiske, and J.-L. Guan
`
`2009 Desmoplakin is required early in development
`for assembly of· desmosomes and cytoskeletal
`linkage.
`G.I. Gallicano, P. Kouklis, C. Bauer, M. Yin,
`V. Vasioukhin, L. Degenstein, and E. Fuchs
`
`2023 Mutation of a major. keratin phosphorylation site
`predisposes to hepatotoxic injury in transgenic
`mice.
`N.-0. Ku, S.A. Michie, R.M. Soetikno,
`E.Z. Resurreccion, R.L. Broome, and M.B. Omary
`
`2033 Molecular organization of sarcoglycan complex
`in mouse myotubes in culture.
`Y.-m. Chan, C.G. Bonnemann, H.G.W. Lidov,
`and L.M. Kunkel
`
`2045 Functional characteristics of ES cell-derived
`cardiac precursor cells identified by
`tissue-specific expression of the green
`fluorescent protein.
`E. Kolossov, B.K. Fleischmann, Q. Liu, W. Bloch,
`S. Viatchenko-Karpinski, 0. Manzke, G.J. Ji,
`H. Bohlen, K. Addicks, and J. Hescheler
`
`2057 Autocrine tumor necrosis factor (TNF) and
`lymphotoxin (L T) a differentially modulate
`cellular sensitivity to TNF/LT-a cytotoxicity in
`L929 cells.
`E. Decoster, S. Cornelis, B. Vanhaesebroeck,
`and W. Fiers
`
`2067 The cell adhesion molecule L1 is
`developmentally regulated in the renal
`epithelium and is involved in kidney branching
`morphogenesis.
`H. Debiec, E.I. Christensen, and P.M. Ronco
`2081 Activation of avl33 on vascular cells controls
`recognition of prothrombin.
`T.V. Byzova and E.F. Plow
`
`2093 ADDITIONS AND CORRECTIONS
`2095 AUTHOR INDEX FOR VOLUMES 140-143
`2131 SUBJECT INDEX FOR VOLUMES 140-143
`2157 ACKNOWLEDGMENT
`
`CFAD v. Anacor, IPR2015-01776 ANACOR EX. 2094 - 4/14
`
`

`
`Mutation of a Major Keratin Phosphorylation Site
`Predisposes to Hepatotoxic Injury in Transgenic Mice
`Nam-On Ku,*ll Sara A. Michie,+ll Roy M. Soetikno,*ll Evelyn Z. Resurreccion,+ll Rosemary L. Broome,§ll
`and M. Bishr Omary*ll
`*Department of Medicine, *Department of Pathology, and §Department of Veterinary Medicine, Veterans Administration Palo
`Alto Health Care System, Palo Alto, CA 94304; and the IIDigestive Disease Center, Stanford University School of Medicine,
`Palo Alto, California 94305
`
`Abstract. Simple epithelia express keratins 8 (K8) and
`18 (K18) as their major intermediate filament (IF) pro(cid:173)
`teins. One important physjologic function of K8/18 is to
`protect hepatocytes from drug-induced liver injury. Al(cid:173)
`though the mechanism of this protection is unknown,
`marked K8/18 hyperphosphorylation occurs in associa(cid:173)
`tion with a variety of cell stresses and during mitosis.
`This increase in keratin phosphorylation involves mul(cid:173)
`tiple sites including human K18 serine-( ser )52, which is
`a major K18 phosphorylation site. We studied the sig(cid:173)
`nificance of keratin hyperphosphorylation and focused
`on K18 ser52 by generating transgenic mice that over(cid:173)
`express a human genomic K18 ser52-?ala mutant
`(S52A) and compared them with mice that overexpress,
`at similar levels, wild-type (WT) human K18. Abroga(cid:173)
`tion of K18 ser52 phosphorylation did not affect fila(cid:173)
`ment organization after partial hepatectomy nor the
`
`ability of mouse livers to regenerate. However, expo(cid:173)
`sure of S52A-expressing mice to the hepatotoxins,
`griseofulvin or microcystin, which are associated with
`K18 ser52 and other keratin phosphorylation changes,
`resulted in more dramatic hepatotoxicity as compared
`with WT K18-expressing mice. Our results demonstrate
`that K18 ser52 phosphorylation plays a physiologic role
`in protecting hepatocytes from stress-induced liver in(cid:173)
`jury. Since hepatotoxins are associated with increased
`keratin phosphorylation at multiple sites, it is likely that
`unique sites aside from K18 ser52, and phosphorylation
`sites on other IF proteins, also participate in protection
`from cell stress.
`
`Key words: keratins • phosphorylation • intermediate
`filaments • transgenic mice • liver
`
`I NTERMEDIATE filament (IF)1 proteins are one of the
`
`three major cytoskeletal protein groups, that also in(cid:173)
`clude microfilaments and microtubules (reviewed in
`reference 14, 34, 49). A comparison of the three major cy(cid:173)
`toskeletal protein groups shows several distinguishing
`unique features of IF proteins: they have nuclear (i.e., the
`lamins) and many cytoplasmic members, their cytoplasmic
`members appear to be expressed only in higher eucaryotes
`in a tissue preferential manner, and mutations of IF pro(cid:173)
`teins cause a variety of human diseases (15). Among the
`diverse IF protein family, the keratin subgroup is the larg-
`
`Address correspondence to Bishr Omary, Palo Alto VA Medical Center,
`3801 Miranda Avenue, 154J, Palo Alto, CA 94304. Fax: (415) 852-3259.
`Address reprint requests to Nam-On Ku, Palo Alto VA Medical Cen(cid:173)
`ter, 3801 Miranda Avenue, 154J, Palo Alto, CA 94304. Fax: (415) 852-
`3259.
`
`1. Abbreviations used in this paper: Emp, Empigen BB; GF, griseofulvin;
`IF, intermediate filament(s); K, keratin; MLR, microcystin LR; pS, phos(cid:173)
`pho-serine; PVDF, polyvinylidene difluoride; S52A, transgenic mice that
`overexpress ser52--7ala K18; TG2, transgenic mice that overexpress wild(cid:173)
`type human K18; WT, wild-type.
`
`est and is specifically expressed in epithelial cells ( 44).
`Aside from the "hard" keratins that are found in epider(cid:173)
`mal appendages (e.g., hair and nails), the "soft" keratins
`(K) consist of >20 members that are divided into rela(cid:173)
`tively basic type II (K1-K8) and acidic type I (K9-K20)
`keratins ( 44). All epithelial cells express at least one type I
`and one type II keratin that associate, noncovalently, to
`form extended filamentous arrays. In general, epithelial
`cells express a dominant unique keratin pair depending on
`the epithelial cell type. For example, glandular epithelia
`express K8 and K18 (with variable levels of K19 and K20),
`whereas keratinocytes preferentially express K5/14 basally
`and Kl/10 suprabasally. Although keratin function(s) re(cid:173)
`main poorly understood, one clear function is to provide
`mechanical integrity to cells including those in epidermis
`(15), cornea (19) and liver ( 46). This was clearly demon(cid:173)
`strated by the phenotypes of several human diseases that
`are caused by keratin mutations and by several transgenic
`animal models that express mutant keratins (15, 43, 46).
`All IF proteins have a prototype structure consisting of
`a central coiled-coil a.-helix (310-350 amino acids) that is
`surrounded by globular NHz-terminal head and COOH-
`
`The Journal of Cell Biology, Volume 143, Number 7, December 28, 1998 2023-2032
`http://www.jcb.org
`2023
`
`CFAD v. Anacor, IPR2015-01776
`ANACOR EX. 2094 - 5/14
`
`

`
`terminal tail domains of variable length (14, 34, 49). The
`globular end domains provide most of the structural het(cid:173)
`erogeneity among IF proteins and contain all currently
`known phosphorylation and glycosylation sites, in contrast
`with the nonmodified rod domain (reviewed in 18, 26, 47).
`The exclusive presence of keratin phosphorylation within
`the structurally heterogeneous head and tail domains sug(cid:173)
`gests that this modification may play a regulatory role in
`the presumed tissue-specific function of these proteins. In
`the case of K18, ser52 is its major phosphorylation site
`during interphase. K18 ser52 phosphorylation is highly dy(cid:173)
`namic, increases two to three times during mitosis, and ap(cid:173)
`pears to be important for filament reorganization (23, 37).
`A potential role for K18 ser52 in filament organization
`was implicated based on transfection of a K18 ser52--7ala
`mutant, which results in a keratin filamentous array that is
`twofold less able to reorganize its filaments, after treat(cid:173)
`ment with colcemid or okadaic acid, as compared with WT
`K18-transfected cells (23). Several other K8 and Kl8 phos(cid:173)
`phorylation sites have been identified including K18 ser33
`that regulates binding to 14-3-3 proteins (30), K8 ser23
`that is highly conserved among all type II keratins (24), K8
`ser431 that is phosphorylated by MAP kinase upon EGF
`stimulation (24), and K8 ser73 that becomes phosphory(cid:173)
`lated during mitosis, cell stress, and apoptosis ( 40).
`Two observations led us to investigate keratin phosphor(cid:173)
`ylation in transgenic mice in situ. First, disruption of K8/18
`filaments in transgenic mice after expression of a domi(cid:173)
`nant negative K18 arg89--7cys results in hepatocyte keratin
`IF collapse in association with chronic hepatitis (25) and
`increased susceptibility to drug-induced liver injury (27).
`This, coupled with the abnormal liver phenotype of K8-
`null mice (4, 5, 41) and mice that express K14 ectopically in
`the liver (3) indicated that K8/18 play an important role in
`helping hepatocytes cope with cell stress. Such a functional
`role for keratin was not discernible before the use of trans(cid:173)
`genic animal models as was done first for K14 (54) and
`subsequently several other IF proteins (15), and the initial
`identification of keratin mutations in the human blistering
`skin disease epidermolysis bullosa simplex (6, 13, 33). Sec(cid:173)
`ond, a variety of stress conditions in cultured cells and
`intact animals are associated with significant K8/18 hy(cid:173)
`perphosphorylation, including rotavirus infection or heat
`stress in cultured cells (38), apoptosis (8, 28), and drug(cid:173)
`induced liver injury using the anti-fungal agent griseoful(cid:173)
`vin (27) or the phosphatase inhibitor microcystin LR (MLR;
`45, 51, 52). Given that ser52 is a major K18 phosphoryla(cid:173)
`tion site, we sought to determine its physiologic function
`by generating transgenic mice that overexpress a K18
`ser52--7ala mutant and comparing the phenotype and ker(cid:173)
`atin phosphorylation in these mice with the well described
`transgenic mice that overexpress WT human K18 (termed
`TG2; references 1, 25, 27). This represents the first report
`that we are aware of that utilizes transgenic animals to
`study the significance of a single phosphorylation site of a
`protein.
`
`Materials and Methods
`
`Cell Culture, Antibodies, and Reagents
`NIH-3T3 (mouse fibroblast) and HT29 (human colon) cells (American
`
`Type Culture Collection, Manassas, VA) were cultured as recommended
`by the supplier. Antibodies (Ab) used were: L2A1 mouse mAb that rec(cid:173)
`ognizes human K18 without cross-reacting with mouse keratins (11, 25);
`Troma I rat mAb that recognizes mouse K8 (Developmental Studies Hy(cid:173)
`bridoma Bank, University of Iowa, Iowa City, IA); rabbit Ab 8592 that
`was raised against human K8/18 (25); rabbit Ab 3055 that recognizes
`phospho-ser52 (pS52) of human K18 and does not cross-react with the
`corresponding phosphorylation site in mouse K18 that has a different anti(cid:173)
`genic context (37); rabbit Ab 8250 that recognizes human K18 pS33 and
`does cross-react with the equivalent mouse phosphorylation site (30);
`mAb LJ4 that recognizes K8 pS73 ( 40); anti-mouse K18 (provided by
`Robert Oshima); and anti-plectin and anti-desmoplakin Abs (provided by
`Harald Herrmann and Manijeh Pasdar, respectively). Other reagents used
`were: microcystin-LR (Alexis Corp., San Diego, CA), griseofulvin (GF;
`Sigma Chemical Company, St. Louis, MO), calf intestine alkaline phos(cid:173)
`phatase (Boehringer Mannheim, Indianapolis, IN), orthophosphate (32P04;
`Dupont-New England Nuclear, Wilmington, DE), Empigen (Emp) BB
`detergent (Calbiochem-Novabiochem Corp., La Jolla, CA), collagenase
`type I (Worthington Biomedical Corp., Freehold, NJ), TransformerTM
`mutagenesis kit (CLONTECH Laboratories, Palo Alto, CA), and pow(cid:173)
`dered Lab Diet (PMI Feeds Inc., St. Louis, MO).
`
`Transgene Construct and Generation of
`Transgenic Lines
`The human K18 eDNA ser52 codon (AGC) was mutated to a GCC (ala)
`codon in a pBluescript SK + plasmid using the Transformer kit as recom(cid:173)
`mended by the manufacturer. Confirmation of the mutation was done by
`sequencing both strands of the mutated region. The mutant eDNA was
`then digested with AlwN I to generate a 250-bp fragment that was then
`substituted for the corresponding wild-type segment of exon 1 in a 10-kb
`genomic K18 clone (provided by Dr. Robert Oshima, The Burnham Insti(cid:173)
`tute, La Jolla, CA), exactly as we had done in generating the arg89-7cys
`K18 genomic mutant (25). The 10-kb K18 ser52-7ala genomic DNA was
`injected into pronuclei of fertilized FVB/N mouse eggs. Progeny mice
`carrying the human K18 gene were then chosen, after PCR screening,
`followed by breeding to select for germline transmission using stan~
`dard methods. Two mouse lines (S52A1 and S52A2) that express K18
`ser52-7ala were expanded and used for subsequent studies. The control
`mice that were used (TG2 mice) overexpressed the wild-type 10-kb ge(cid:173)
`nomic K18 (1, 25, 27). PCR screening of mouse tail genomic DNA for the
`presence of human K18 involved amplification of a 270-bp fragment that
`corresponds to the COOH-terminal region of K18. The primers used
`were:(+) 5'-CAGAAGGCCAGCTTGGAGAAC-3' and(-) 5'-ATC(cid:173)
`TCCTGATCCCAGCACGTG-3'. All mice were housed in the same
`room with standard infection control precautions.
`
`Transgenic Mice, Liver and Serum Testing, and
`Partial Hepatectomy
`For all transgenic mouse hepatectomy and liver toxicity experiments, age
`(all >6 wk old, no more than 2 wk difference in age among the various
`groups) and sex-matched mice were weighed just before use. Liver and
`blood were collected after killing the mice using C02 inhalation, then
`bleeding via intracardiac puncture (0.5-1.0 ml). The liver was them imme(cid:173)
`diately harvested. Serum was analyzed from TG2 and S52A1 and S52A2
`control diet-fed mice (8 mice/line), GF-fed TG2 and S52A1 mice (10 mice/
`line), MLR-treated mice (10 mice/line) for creatinine, glucose, total pro(cid:173)
`tein, alkaline phosphatase, albumin, triglycerides, cholesterol, total biliru(cid:173)
`bin, and alanine and aspartate aminotransferases. Partial hepatectomy
`was done by removing the lateral, left, and right median lobes, as de(cid:173)
`scribed (55, 56) from TG2, S52A1 and S52A2 mice (16 mice/per transgenic
`line). Mice were then killed 24 h (4 mice/line), 48 h (6 mice/line), and 72 h
`(6 mice/line) afterwards and the liver:s were processed as below. For the
`hepatectomy experiments, sham-hepatectomized mice (i.e., mice that had
`anesthesia, an abdominal wall and peritoneal incision, exposure of the
`liver then closure of the incisions) from each transgenic line served as con(cid:173)
`trols. Resected livers (from control diet-fed, sham-hepatectomized, post(cid:173)
`hepatectomy, GF-fed, or MLR-administered) were cut into several pieces
`depending on the experiment and used for one or more of the following:
`(a) fixation with 10% formalin followed by paraffin embedding, section(cid:173)
`ing, then hematoxylin and eosin staining (done by Histo-tec Laboratory,
`Hayward, CA), (b) snap freezing in O.C.T. compound, sectioning then fix(cid:173)
`ing briefly in cold acetone for subsequent immunofluorescence staining,
`(c) snap freezing in liquid nitrogen for subsequent biochemical analysis,
`
`The Journal of Cell Biology, Volume 143, 1998
`
`2024
`
`CFAD v. Anacor, IPR2015-01776
`ANACOR EX. 2094 - 6/14
`
`

`
`and (d) cutting into small fragments then metabolic labeling with 32P04 in
`phosphate-free medium. Liver perfusion was performed as described (12)
`using 0.025% collagenase type I, and hepatocyte viability was determined
`using trypan blue exclusion.
`
`by immunoblotting of serial dilutions of total liver tissue homogenates
`using anti-pankeratin or anti-phosphokeratin antibodies. Northern blot
`analysis was done using total liver RNA.
`
`Griseofulvin and Microcystin-LR Experiments
`A pilot experiment was initially done using TG2, S52A1, and S52A2 mice
`that were fed powered diet ±1.25% GF (6 mice/group). Since both S52A1
`and S52A2 mice gave similar results in terms of having a worse histologic
`score upon GF feeding as compared with TG2 mice, we only used the
`S52A 1 for subsequent experiments. Mice (10/line) were then fed control
`or GF-supplemented diet for 17 d followed by isolation, weighing then
`processing of the livers as described above. Alternatively, mice were fed
`control or OF-containing diet for 2, 5, or 8 d (2 mice/line) followed by the
`processing of the livers. For the MLR experiments, pilot experiments
`were carried out to determine the optimal dose that does not cause rapid
`lethality. A dose of 30 J..Lg/Kg was chosen, and as found for the GF experi(cid:173)
`ments, the S52A 1 and S52A2 had a worse histology than the TG2 mice that
`led us to use the S52A 1 line for the subsequent quantitative experiments.
`Mice (10/line) from each group were given 30 J..Lg/~g of MLR intraperito(cid:173)
`neally, followed by processing of the livers after 195 min. MLR was pre(cid:173)
`pared in dimethylsulfoxide, as a 1 mg/ml stock solution, then diluted in
`PBS (10 mM sodium phosphate/0.15 M NaCl, pH 7.4).
`
`Histology Grading and Statistical Analysis
`All histology slides were examined by a pathologist (S.A. Michie) without
`knowledge of the treatment, diet, or transgenic line. Grading of the OF(cid:173)
`related slides was done by counting the number of necrotic cells in a 20X
`field (10 fields per slide) and is presented as a necrosis score. Grading of
`the MLR-related slides was done by estimating the extent of vacuolization
`(vacuolization score) and area of hemorrhage/necrosis (hemorrhage score).
`The vacuole score was estimated by assigning a 0 to 3+ score depending
`on the percentage of vacuole volume as compared with overall hepatocyte
`cytoplasmic volume such that: 0 = <10%, 1 + = 10-29%, 2+ = 30-49%,
`and 3+ = :::::50%. The hemorrhage score was estimated by assigning a 0 to
`3+ score depending on extent of the hemorrhage and necrosis: 0 =none(cid:173)
`crosis, 1 + =small foci (mostly midzonal), 2+ =confluent areas, and 3+ =
`diffuse hemorrhage and necrosis with rare intact islands of hepatocytes.
`All scores and laboratory parameters are expressed as means ±SO. The
`Student's t test and nonparametric Wilcoxon method were used to calcu(cid:173)
`late the statistical significance between the means. Statistical analysis was
`performed using JMP version 3.1 (SAS Institute Inc., Cary, NC).
`
`Keratin Isolation, Immunoprecipitation, and
`Other Methods
`Liver pieces (rv3 X 4 X 4 mm) were used to isolate keratins exactly as de(cid:173)
`scribed previously, either by immunoprecipitation (26) or high salt extrac(cid:173)
`tion (2, 25) except that NaF and sodium pyrophosphate were not added to
`the solubilization buffer. For immunoprecipitation, liver pieces were solu(cid:173)
`bilized in 1% Emp or 1% NP-40 in PBS containing a protease/phos(cid:173)
`phatase inhibitor cocktail (25). Dephosphorylation of keratin immunopre(cid:173)
`cipitates was done with alkaline phosphatase (20 units/J..Ll stock enzyme
`mix) using 20 units in 20 J..Ll of buffer (supplied by the manufacturer with
`the enzyme) for 1 h (30°C) followed by washing 2X with 1% NP-40 in
`PBS. SDS-PAGE was done using 10% acrylamide gels (31). Radiolabel(cid:173)
`ing of NIH-3T3 or HT29 cells, or liver tissue fragments (250 J..LCi/ml) was
`done in 5 ml or 2 ml, respectively, of phosphate free RPMI-1640 media
`containing 10% dialyzed FCS and 1-2% normal medium. Immunoblotting
`(53) was done by transferring immunoprecipitates or total celllysates (sol(cid:173)
`ubilized in 2% SDS/5% glycerol-containing Laemmli sample buffer) to
`polyvinylidene difluoride (PVDF) membranes followed by blotting with
`1:1,000 dilution of anti-keratin antibodies then visualization of the reac(cid:173)
`tive bands using enhanced chemiluminescence. Tryptic phosphopeptide
`mapping of K18 that was purified by immunoprecipitation from 32P04-
`prelabeled detergent solubilized HT29 or transfected NIH-3T3 cells or
`mouse freshly isolated liver fragments was done exactly as described (7).
`IEF of the keratin precipitates (39) was done using a Mini-PROTEAN II
`apparatus (BioRad Laboratories, Cambridge, MA) and an ampholine pH
`mix of 3-10 and 5-7 as recommended by the manufacturer. Immunofluo(cid:173)
`rescence staining was done as described (26).
`Comparative quantitation of keratin protein levels and phosphoryla(cid:173)
`tion was done using densitometric scanning of Coomassie-stained gels, or
`
`Results
`
`Characterization of Transgenic Mice That Overexpress
`Human K18 ser52~la
`To address the physiologic function of keratin phosphory(cid:173)
`lation, we targeted a specific keratin phosphorylation site
`in the context of an intact transgenic mouse. Our hypothe(cid:173)
`sis was that transgene expression of a keratin that is mu(cid:173)
`tated at a particular phosphorylation site may clarify the
`potential role of keratin phosphorylation during mitosis
`and/or in providing protection from drug-induced liver in(cid:173)
`jury. This hypothesis is based on the increase in K8/18
`phosphorylation that is observed in association with a
`variety of cell stresses in mice and cultured cells (26, 47).
`To test this hypothesis, we generated a genomic K18
`ser52~ala mutant and injected it into mouse embryos
`then selected two lines (S52A1 and S52A2). The wild-type
`(WT) 10-kb genomic K18 construct that we used to gener(cid:173)
`ate the point mutation was identical to what we used pre(cid:173)
`viously to generate a genomic K18 arg89~cys, which af(cid:173)
`forded tissue specific overexpression of human K18 that
`paralleled that of the endogenous mouse K18 (25). For
`comparison, and to serve as a control for WT human K18
`overexpression, we used the well-characterized TG2 mice
`that overexpress WT human K18 (1, 25). To isolate the
`overexpressed human keratin, we used mAb L2A1 that
`recognizes human K18 and does not cross-react with
`mouse keratins (25). As shown in Fig. 1 A, mAb L2A1 im(cid:173)
`munoprecipitated nearly equal levels of keratins from liv(cid:173)
`ers of TG2 mice and mice from the two S52A transgenic
`lines. As would be expected, immunoblotting of these pre(cid:173)
`cipitates with the previously described Ab 3055, which
`specifically recognizes an epitope that contains human
`phospho-ser52 (pS52; reference 37), showed binding only
`to K18 from TG2 mice but not to K18 from S52A mice
`(Fig. 1 A).
`The relative levels of keratin expression were compared
`in the livers of the transgenic lines and normal nontrans(cid:173)
`genic FVB/N mice by examining the total keratin pool us(cid:173)
`ing high salt extraction (Fig. 1 B) and total liver homage(cid:173)
`nates (not shown). In TG2 mice, A-/80% of the type I
`keratins (i.e., the total human ectopic and endogenous
`mouse K18) is human K18 whereas in the S52A mice
`A-/70% of the total keratin is human K18. Both S52A lines
`express similar human K18 levels that in turn are slightly
`lower than those noted in TG2 mice. The endogenous lev(cid:173)
`els of mouse K18 in TG2 and S52A mice, relative to wild(cid:173)
`type mice, are decreased in proportion to the increased
`levels of ectopic human K18 such that the total type I ker(cid:173)
`atins remains essentially constant (not shown). Overall
`and upon normalization of protein levels, type II keratin
`levels are also similar in TG2, S52A, and wild-type mice.
`Assignment of individual keratin bands was done by im(cid:173)
`munoblotting with anti-human and -mouse keratin-spe(cid:173)
`cific antibodies (e.g., Fig. 1 B and data not shown). The

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