throbber
W1 AN77G
`
`1995
`v.11
`0.91 —————— *"SEO: SROOBSSBQ
`
`TI: ANNUAL REVIEW OF CELL AND
`DEVELOPMENTAL BIOL 08/08/96
`
`
`
`Page 1 0f 35
`
`Kaken Exhibit 2062
`
`Acrux V. Kaken
`
`IPR2017-00190
`
`

`

`TX 4—159—264
`
`lllllllllllllll[Illflljjlflllfllljlljlllllilllllllllll
`M
`"
`FE -
`ANNUAL REVIEW OF
`CELL AND
`DEVELOPMENTAL
`BIOLOGY
`
`VOLUME 11, 1995
`
`JAMES A. SPUDICH, Editor
`
`Stanford University School of Medicine
`
`JOHN GERHART, Associate Editor
`
`University of California, Berkeley
`
`STEVEN L. MCKNIGHT, Associate Editor
`
`TULARIK, South San Francisco
`
`RANDY SCHEKMAN, Associate Editor
`
`University of California, Berkeley
`
`ANNUAL REVIEWS INC.
`Page 2 0f
`
`4139 EL GAMING \myma'tfifiltmflflag
`:1. the NLM and may be
`Sub-fiat USC'ooooright Laws
`
`PALO ALTO. CALIFORNIA 94303-0139
`
`

`

`l
`
`I
`
`ANNUAL REVIEWS INC.
`
`Palo Alto, California, USA
`
`COPYRIGHT © 1995 BY ANNUAL REVIEWS th., PALO ALTO, CALIFORNIA, USA. ALL
`
`RIGHTS RESERVED. The appearance of the code at the bottom of the first page of an
`
`article in this serial indicates the copyright owner's consent that copies of the
`
`article may be made for personal or internal use, or for the personal or intemai use
`
`of specific clients. This consent is given on the condition, however,
`
`that the copier
`
`pay the stated per-copy fee of $5.00 per article through the Copyright Clearance
`
`Center, Inc. (222 Rosewood Drive, Danvers, MA 01923) for copying beyond that
`
`permitted by Sections 107 or 108 of the US Copyright Law. The per~copy fee of
`
`$5.00 per article also applies to the copying, under the stated conditions, of articles
`
`published in any Annual Review serial before January 1, 1978. Individual readers,
`
`and nonprofit libraries acting for them, are permitted to make a single copy of an
`
`article without charge for use in research or teaching. 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. For such
`
`uses, written permission is required. Write to Permissions Dept., Annual Reviews
`
`Inc., 4139 El Camino Way, PO. Box 10139, Palo Alto, CA 94303-0139 USA.
`
`International Standard Serial Number: 1081—0706
`
`International Standard Book Number: 0—8243—311 1-7
`
`Annual Review and publication titles are registered trademarks of Annual Reviews Inc.
`
`® The paper used in this publication meets the minimum requirements of
`
`American National Standard for Information Sciences—uPermanence of Paper
`
`for Printed Library Materials, ANSI 239.48-1984.
`
`Annual Reviews Inc. and the Editors of its publications assume no responsibility for the
`
`statements expressed by the contributors to this Review.
`
`Typesetting by Kachina Typesetting Inc., Tempe, Arizona; John Olson. President;
`
`Toni Starr, Typesetting Coordinator; and by the Annual Reviews Inc. Editorial Staff
`
`PRINTED AND BOUND IN THE UNITED STATES OF AMERICA
`
`Page 3 of 35
`
`

`

`Alum. Rev. Cell Dev. Biol. I995. 11323—53
`
`Copyright © 1995 by Animal Reviews Inc. All rights reserved
`
`KERATINS AND THE SKIN
`
`EZaine Fuchs
`
`Howard Hughes Medical Institute, Department of Molecular Genetics and Cell
`Biology, The University of Chicago. 5841 South Maryland Avenue, Chicago.
`Illinois 6063’?
`
`KEY WORDS:
`
`keratin filaments. genetic disease, epidermis, hair follicles, multigene family
`
`
`
`CONTENTS
`
`SKIN AND ITS PROGRAMS OF EPITHELIAL DIFFERENTIATION . . . . . . . . . . . ..
`
`124
`
`PATTERNS OF KERATIN EXPRESSION IN EMBRYONIC AND ADULT
`MAMMALIAN SKIN . . . .
`.
`.
`. . . . . . . .
`.
`.
`.
`. . . . .
`.
`. . . . .
`.
`.
`. . . . .
`.
`
`.
`
`.
`
`.
`
`.
`
`.
`
`. ..
`
`REGULATION OF KERATIN GENE EXPRESSION IN THE SKIN . .
`
`.
`
`. .. .. .. .. ..
`
`ASSEMBLY AND STRUCTURE OF KERATIN FILAMENTS .
`
`.
`
`.
`
`. . .
`
`. . .
`
`.
`
`.
`
`.
`
`. . .
`
`.
`
`.
`
`.
`
`.
`
`ASSOCIATIONS BETWEEN KERATIN IFS AND OTHER PROTEINS AND
`STRUCTURES: FORMING A CYTOPLASMIC IF NETWORK IN
`EPIDERM IS AND HAIR . .
`.
`.
`.
`.
`.
`.
`.
`.
`. . . . . . .
`.
`. . . . .
`. . . . .
`.
`.
`.
`.
`.
`.
`. . . . . . .
`
`.
`. . . . .
`KERATIN MUTATIONS AND BLISTERING HUMAN SKIN DISORDERS .
`.
`.
`.
`.
`.
`.
`Epidermolysr's Buliosa Simplex .
`.
`.
`. .
`.
`.
`.
`.
`.
`.
`. .
`.
`.
`.
`.
`. .
`. .
`.
`.
`. .
`. . . .
`. .
`.
`.
`.
`.
`. .
`.
`.
`.
`.
`. .
`Epidermolyric Hyperkerarosi: . .
`. . .
`.
`. . . .
`.
`.
`. . . . .
`.
`. . .
`. . .
`.
`.
`.
`.
`.
`. . . . .
`. . .
`.
`. .
`.
`.
`. .
`Epidermal Nevr' of the EH Type .
`.
`.
`.
`.
`.
`. .
`. .
`.
`.
`.
`.
`. .
`.
`.
`.
`.
`. .
`. .
`.
`.
`. .
`.
`.
`.
`.
`.
`.
`. .
`I’ahnopianrar Keraroderma and Paehyonyehia Congem'm . . .
`.
`.
`. . . . .
`.
`.
`.
`. . . . .
`.
`. .
`KERATIN IFS FUNCTION TO IMPART MECHANICAL INTEGRITY TO CELLS:
`POSSIBLE INSIGHTS INTO ADDITIONAL GENETIC DISORDERS OF
`KERATIN .
`.
`.
`. .
`.
`.
`. .
`. .
`. . . . .
`. . .
`.
`.
`. . . . .
`.
`. . . . .
`.
`.
`.
`. . . . . . . . . . .
`.
`. . . . .
`.
`
`CONCLUSIONS AND PERSPECTIVES . . . . . .
`
`.
`
`. . . . .
`
`.
`
`. . . . . .
`
`.
`
`.
`
`. . . . .
`
`.
`
`.
`
`.
`
`. . .
`
`.
`
`.
`
`.
`
`.
`
`.
`
`128
`
`130
`
`135
`
`133
`
`I40
`I40
`144
`144
`14S
`
`146
`
`I46
`
`ABSTRACT
`
`Keratins are the major structural proteins of the vertebrate epidermis and its
`appendages, constituting up to 85% of a fully differentiated keratinocyte.
`Together with aetin microfilaments and microtubules, keratin filaments make
`up the cytoskeletons of vertebrate epithelial cells. Traced as far back in the
`evolutionary kingdom as mollusks, keratins belong to the superfamily of in-
`termediate filament (IF) proteins that form oc-helical coiled-coil dimers which
`associate laterally and end—to-end to form 10-nm diameter filaments. The
`evolutionary transition betWecn organisms bearing an exoskeleton and those
`with an endoskeleton seemed to cause considerable change in keratin. Keratins
`expanded from a single gene to a multigene family. Of the ~60 IF genes in
`
`123
`
`Page 4 Of35
`
`1081—0706/951’1115—0123$05.00
`
`

`

`124
`
`FUCHS
`
`the human genome, half encode keratins, and at least 18 of these are expressed
`in skin. Vertebrate keratins are subdivided into two sequence types (I and II)
`that are typically coexpressed as specific pairs with complex expression pat-
`terns. The filament-forming capacity of a pair is dependent upon its intrinsic
`ability to self-assemble into coiled-coil heterodimers, a feature not required of
`the invertebrate keratins (Weber et a1 1988). Approximately 20,000 heterodi-
`mers of type I and type II keratins assemble into an IF. Mutations that perturb
`keratin filament assembly in vitro can cause blistering human skin disorders
`in vivo. From studies of these diseases, an important function of keratins has
`been unraveled. These filaments impart mechanical strength to a keratinocyte,
`without which the cell becomes fragile and prone to rupturing upon physical
`stress. In this review, studies on the pattern of expression, structure, and
`function of skin keratins are summarized, and new insights into the functions
`of these proteins and their involvement in human disease are postulated.
`
`SKIN AND ITS PROGRAMS OF EPITHELIAL
`DIFFERENTIATION
`
`The single-layered embryonic ectoderm of mammals receives mesenchymal
`cues that specify its programs of differentiation. Early cues influence epidermal
`cell fate, and later signals influence the ectodermal cell to become epidermis
`vs hair follicle. These later cues happen shortly after stratification, where
`condensates of Specialized mesenchyme, referred to as dermal papilla (anlage)
`cells. form in a dotted pattern beneath the embryonic basal layer (Figure I).
`Where contact is made, basal cells of the epidermis differentiate downward to
`craft what will ultimately be the hair follicles of the mammalian skin. In the
`absence of these mesenchymaI-epithelial interactions. basal cells conunit to
`an epidermal cell fate.
`At the single-layer stage, eetodermal proliferation occurs lateral 1y, with the
`mitotic plane perpendicular to the embryo surface (Figure 2). Upon stratifica—
`tion, mitotic activity occurs in all layers, with the mitotic plane often parallel
`
`
`
`Figure I The choica between epidermis and hair follicles during embryonic development in the
`skin. Shortly after stratification, condensates of specialized mescnchyme, or dermal papilla anlage.
`assemble beneath the embryonic basal
`layer of the skin. These condensates provide as—yet-
`undetermined external cues that stimulate basal cells to migrate downward and form a primary hair
`germ. This morphegenic process eventually gives rise to an adult hair follicle.
`
`Page 5 of 35
`
`

`

`KERATINS AND THE SKIN
`
`125
`
`mitosis basally
`single layered epithelium
`
`terminal differentiation suprahasally
`
`mitosis suprabasally
`stratification but no differentiation
`
`mitosis basally
`
`Figure 2 Cell division during embyronic development in the skin. At the single-layer epithelial
`stage, ectodermal proliferation occurs laterally, with the mitotic plane perpendicular to the embryo
`surface. The mitotic plane soon shifts 90°, causing stratification. Suprabasal mitoses are necessary
`during the time that the skin surface is rapidly expanding. Later, the mitotic plane reverts back,
`allowing dividing cells to move only laterally. At this stage, signs of differentiation appear in the
`suprabasal layers.
`
`to the embryo surface (Figure 2). Suprabasal mitoses occur during the time
`that the skin surface undergoes rapid expansion. Later, as suprabasal cells
`display morphological signs of differentiation, mitotic activity becomes re—
`stricted to a single layer of basal cells, with the mitotic plane reverting to a
`perpendicular orientation. This pattern persists in postnatal epidermis.
`In the adult, a basal epidermal cell responds to an as-yet-unidentified trigger
`of terminal differentiation. When it ceases to divide and begins its journey to
`the skin surface, it alters its adhesive properties, evoking changes that are likely
`to be central to the control of the differentiative program (for a recent review,
`see Watt et al 1993). In transit, the cell undergoes a series of morphological
`and biochemical changes culminating in the production of dead, flattened,
`enucleated squames, which are sloughed from the surface, continually replaced
`by inner cells differentiating outward (Figure 3). The trek takes 2 to 4 weeks
`in humans and continues throughout the life of the individual.
`In the adult hair follicle, differentiation is far more complex than in the
`epidermis (Figure 4; Hardy I992). The hair is surrounded by two sheaths, an
`outer root sheath (ORS), continuous with the epidermis but thought to have
`its own compartment of stem cells (Rochat ct al 1994), and an inner root sheath
`
`Page 6 of 35
`
`

`

`126
`
`FUCHS
`
`
`
`
`Splnous Layers:
`Keratln filaments
`made of K1 and K10
`
`Basal Layers:
`Kerntin filaments
`made 01 K5 and K14
`
`-- Basement Membrane
`
`Dermil
`
`F'
`
`c
`
`l
`
`I
`
`.
`
`n
`
`u
`
`s
`
`.
`
`te‘f;::;d'germlnaldifferentiation in adult epidermis. The diagram illustrates the four stages of
`b
`I
`l
`l erenttation and the pattern of keratin expression. All cells above the innermost. i.e.
`
`333 . ayer of the epidermis are considered to be suprabasal.
`
`(IRS), whose cells are derived from the same precursors as the hair shaft. The
`((31138 18 composed of multiple layers, with the outermost layer being the least
`1 ferentiated (Coulombe et a1 1989). In the lower part of the ORS, cells move
`upward and inward as they differentiate, whereas in the upper part of the
`lf)(])llicle, movement 'is largely inward, and differentiation more closely resem-
`es that of the epidermis. The IRS is composed of three layers: the outer
`(1612:: layer, the Huxley’s layer, and the cuticle of the IRS. These three layers
`theg“first;C1,113 5:1: Liliipesrhpptrtior;1 of the follicle..Another layer of cuticle forms
`surface (Figure 4).
`a
`an remains With it as it breaks through the skin
`
`Page 7 of 35
`
`

`

`
`
`
`KERATINS AND THE SKIN
`
`127
`
`I Epidermis
`spinous layers K1, K10
`basal layer -— WKS, K14
`
`fi—l r3
`,‘ '
`
`— Sebaeeous
`Gland
`
`Arrector
`
`_
`
`’3
`
`‘
`
`Muscle
`
`7-- :H ‘-
`
`Bulge -—fi——% K5, K14
`
`Isthmus
`
`
`
`—~ Outer Root Sheath
`
`Hgirr wee—k -—---
`ulicle
`Cortex
`Medulla
`
`inner rool sheath:
`Henle‘s layer
`Huxley's layer
`Cuticle of inner root shealh
`
`("1 K5' K” 'hmugmm
`K6, K16 inner layers
`- Ha. Hb Keratins
`HS. HGT lFAP Keratins
`
`-
`
`-~ 7
`
`K1. K10
`
`
`
`‘ Marix cells *‘
`
`—— no major keratin network
`
`Dermal papilla
`
`vimenlin
`
`Figure 4 Terminal differentiation in the hair follicle. In the growth phase of the hair cycle, cells
`from the dermal papilla interact with proliferating matrix cells. Under an unidentified trigger, matrix
`cells cease to divide, begin to migrate upward, and commit to at least five concentric rings of
`differentiated states: The inner root sheath (IRS), consisting of Henle's layer, Huxley's layer, and
`outer cuticle, guides the hair shaft as it emerges from the immature cortex cells. The shaft consists
`of the inner cuticle and medulla. The outer root sheath (ORS) is contiguous with the basal epidermal
`layer and has a proliferating compartment distinct from matrix cells. These cells move upward and
`inward as they grow and differentiate. The bulge (see text) is a possible silo for stem cells. The
`keratins expressed in different cells of the hair follicle are indicated according to the nomenclature
`of Moi] et al (1982).
`
`The cells of the IRS and the hair shaft are thought to arise from upward
`modes of differentiation that are controlled by the adult dermal papilla cells,
`which maintain a condensate at the base of the follicle. The precursors of the
`hair shaft and IRS are matrix cells, which are relatively undifferentiated epi-
`thelial cells that surround the dermal papilla to form the hair bulb. In their
`mitotically active state, matrix cells maintain close association with the dermal
`
`papilla. As they move upward and away from this compartment, they cease to
`divide and begin to differentiate. These postmitotic cortical cells then give rise
`to the hair shaft as transcriptional activity comes to a halt.
`
`Page 8 of 35
`
`

`

`128
`
`FUCHS
`
`Hairs follow rhythmic periods of growth and quiescence. In anagen, matrix
`cells are highly mitotically active, and the differentiating hair shaft moves
`Upward at about 0.3-0.4 mm/day in humans (Kobori & Montagna 1976).
`Matrix cells are inactive in catagen, a period where the hair follicle degenerates
`and regresses. This is followed by telogen, the period of rest in the hair cycle.
`In humans, the resting period is variable: relatively short for scalp hairs, and
`longer for body hairs. The hair cycle is controlled in part by FGFS, a member
`of the fibroblast growth factor (FGF) family. Mice homozygous for a null
`FGFS mutation have an extended cycle, resulting in the production of unusually
`long hairs (Hebert et a] 1994).
`Throughout an individual’s life, the epidermis and hair follicles must main-
`tain a balance of dividing and differentiating cells. Given the continuous
`renewal programs and ability to respond to injury, it is not surprising that these
`structures have reservoirs of cells capable of generating tremendous prolifera-
`tion. The pepulation of stem cells in the epidermis is likely to reside within
`the basal layer itself, and as judged by analysis of newborn human foreskin
`keratinocyte cultures, there is one clonal subtype, holoclones, whose cells
`possess extraordinary proliferative potential (>100 doublings) (Barrandon &
`Green 1987, Jones & Watt 1993, Jones et a1 1995). Holoclones thus have the
`capacity to generate enough cells from a single clone to completely cover an
`adult human (Rochat et al 1994 and references therein). In the follicle, matrix
`cells are able to amplify as long as they maintain contact with dermal papilla
`condensates. These cells could be the source of stem cells for the IRS, cuticle,
`and hair shaft. However, follicle stem cells have also been hypothesized to
`reside in the bulge, a region at the midpoint of the follicle where the arrector
`pili muscle attaches (Cotsarelis et al 1990). This region contains 95% of the
`keratinocyte colony-forming cells isolated from rat vibrissae, and these cells
`are slow cycling, features generally ascribed to stem cells (reviewed in Rocha;
`et al 1994). The location is attractive because the bulge effectively resides
`outside the hair follicle, which periodically degenerates. Cyclic stimulation of
`new stem cells in the bulge could then take place as the hair regresses upward,
`perhaps bringing dermal papilla cells with it (Cotsarelis et a1 1990). The nature
`of the epithelial-mesenchymal interactions that control the complex differen~
`tiation programs in the follicle are not yet known, although keratinocyte growth
`factor (KGF), another member of the FGF family, has been implicated (Finch
`et a1 1989, Guo et al 1993).
`
`PATTERNS OF KERATIN EXPRESSION IN EMBRYONIC
`AND ADULT MAMMALIAN SKIN
`
`The epidermis and its appendages devote the majority of their protein synthe-
`srzmg machinery to making keratins. Figure 5 provides a schematic of a
`
`Page 9 of 35
`
`T}! i: mzf'nrizll mm: rrxmine‘l
`
`

`

`KERATINS AND THE SKIN
`
`129
`
`Mr(x10'3)
`
`7.4
`
`6.4
`
`5.4
`
`pKi
`
`Figure 5 Two-dimensional gel profile of keratins expressed in the skin. The schematic illustrates
`the electrophoretic mobilities and isoelectric points of the various keratins found in the skin at
`different stages of development and differentiation. Keratins are expressed as specific pairs of type
`I and type 11 proteins. which form obligatory heteropolymers. Each pair is designated by like
`symbols, and the patterns of keratin expression are described in the text. The smaller sizes of like
`symbols indicate minor isoelectric variants of the same keratin (Expression patterns are according
`to Moll et al 1982.)
`
`two-dimensional gel, showing the type, size, and approximate isoelectric points
`of human skin keratins, which are divided into types I and II based on isoelec-
`tric point and sequence. Figures 3 and 4 indicate the pattern of expression of
`the major keratins of adult mammalian epidermis and hair follicles. K8 and
`K18 are the first skin keratins expressed, coincident with the emergence of a
`single-layered ectoderm. These keratins are typically associated with adult
`simple epithelial tissues and are not characteristic of adult stratified squamous
`epithelia. K5 and K14 are then induced (E95 in mice) in a defined pattern
`that is influenced by mesenchymc (Byrne et al 1994). As judged by expression
`of 21 K5 promoter driven B-galactosidase transgene in mice, it is not until E14.5
`that the entire surface ectoderm expresses these genes. In embryo and adult
`mice, K5 and K14 mRNAs seem to be restricted to cells that maintain their
`proliferative capacity (Byrne ct al 1994 and references therein). A minor type
`I keratin, K15, is also expressed in basal keratinocytes (Lloyd et a1 1995). As
`basal cells differentiate in adult skin, they downregulate expression of K5, K14,
`
`Page 10 of 35
`
`This material was {opted
`
`

`

`130
`
`FUCHS
`
`and K15 and induce new sets of differentiation-specific keratins (Fuchs &
`Green 1980, M011 et a1 1982, Sun et a] 1984, Lloyd et al 1995). Most body
`regions express K1 and K10 suprabasally, along with a second type I keratin,
`K11. K2e is also quite broadly expressed suprabasally, but its production is
`delayed relative to K] and K10 (Collin et a1 1992). K9 is confined to suprabasal
`palmo and plantar skin (Fuchs & Green 1980, Langbein et al 1993). K6 and
`K16 are unusual in that they are induced suprabasally during wound healing,
`upon retinoic acid treatment, or in hyperproliferative diseases of the skin,
`including various skin cancers (Sun et a1 1984). K6, K16, and K17 are also
`induced when skin keratinocytes are cultured in vitro. The fianctional signifi-
`cance of. the multiplicity of keratins has not yet been resolved; however, the
`assembly properties of keratins differ as do their differential interactions with
`lF-associated proteins.
`Keratin expression in the hair follicle is as complex as its differentiation
`programs suggest (Figure 4). In the ORS, as cells differentiate and move
`inward, they maintain mitotic activity, upregulate expression of K5 and K14,
`and also initiate expression of K6 and K16. In the IRS, K1 and K10 are
`expressed. The IF network in the matrix cells of the hair follicle has been
`difficult to discern either ultrastructurally or biochemically. K19 and possibly
`very low levels of K14 have been the only keratins identified in the matrix,
`and whether they produce a bona fide IF network remains to be determined.
`As these cells differentiate, however, they initiate abundant expression of Ha
`and Hb keratins, which are exclusive to the cortex of the follicle (Lynch et a]
`1986, Heid et a1 1986, M011 et a1 1988, Kopan & Fuchs 1989, Rogers & Powell
`1993). In addition to Ha and Hb keratins, there are high sulfur (HS) and high
`glycine-tyrosine (HGT) proteins, which are also called keratins. These proteins
`share no sequence or structural homology to the IF keratins, but rather they
`are small IF-associated proteins that seal together the IFS into large macrofi-
`brillar structures. Expression of the Ha/Hb, HS, and HGT keratins in the cortex
`is complex, and not all of the genes encoding these proteins are induced
`simultaneously (Rogers & Powell 1993).
`
`REGULATION OF KERATIN GENE EXPRESSION IN THE
`SKIN
`
`All keratins seem to be encoded by separate genes, and as yet, no evidence of
`differential splicing has been reported. Human K14 was the first keratin whose
`cDNA was cloned (Hanukoglu & Fuchs 1982) and whose gene was sequenced
`(Marchuk et a1 1984). Following these studies, a flood of additional epidermal
`and hair follicle cDNA and gene sequences were reported (for review, see
`Fuchs & Weber 1994). As judged by nuclear run-off experiments, expression
`of skin keratin mRNAs is largely regulated at the transcriptional level (Stell-
`Page 11 of35
`
`

`

`KERATINS AND THE SKIN
`
`131
`
`mach et a1 1991). Transgenic mouse studies have revealed that for many skin
`keratin genes, the sequences involved in regulating their expression reside in
`the 5' upstream sequences of the genes (Vassar et a1 1989, Bailleu] et a1 1990,
`Powell et a1 1992, Takahashi et a1 1994). A knowledge of the major transcrip-
`tion factors controlling skin keratin gene expression is of central importance
`in the quest to elucidate the molecular mechanisms underlying keratinocyte
`specificity and epidermal and follicle differentiation.
`Among the candidates implicated in orchestrating epidermal gene expres-
`sion is the sequence 5’-GCCTGCAGGC—3’, first identified 5’ from the TATA
`box of vertebrate K14 genes (Leask et al 1990, Snape et al 1990). For the K14
`gene, the sequence acts in synergy with a distal element to regulate transcrip-
`tion in keratinocytes (Leask et al 1990). Epidermal nuclear extracts contain a
`protein(s) that binds to this sequence and cross-reacts with antibodies against
`the transcription factor AP2 (Leask et a1 1990, Snape et a] 1991). APZ-binding
`sites have now been found in the promoters of most epidermal- and some
`hair—specific genes, and where tested, they are functionally important for gene
`expression (Leask et a] 1990, Snape et a1 1990, Byrne & Fuchs 1993 and
`references within).
`During embryogenesis, multiple AP2 mRNAs are synthesized in the skin
`(Snape et a1 1991 and references within; Buettner et a1 1993), and an AP2
`CRNA recognizing these forms hybridizes to tissues of ectodermal and neural
`crest lineages (Mitchell et al 1991). As judged by in situ hybridizations of
`whole mouse embryos, the patterns of AP2 mRNA on the embryo surface are
`strikingly similar to and precede by about 1 day those patterns of K5 and K14
`mRNAs (Byrne et al 1994). In contrast, a dominant negative inhibitor of AP2,
`referred to as AP2B (Buettner et al 1993), is not in the ectoderm of developing
`skin (Byrne et al 1994). Thus APZ mRNAs are positioned temporally and
`spatially to play a role in controlling gene expression during skin development
`and differentiation.
`How important is AP2 in controlling epidermal—specific gene expression?
`In at least one case, AP2 appears to be central. Thus recombinant AP2A (the
`active form of AP2) can impart to cultured hepatocytes the ability to express
`K5 and K14 promoter-driven transgenes when cotransfected into these cells
`(Byrne et a1 1994). APZB counteracts these effects. However, the mere pres—
`ence of AP2 does not mandate the expression of endogenous K5 and K14
`genes in cultured cells or in animals (Leask et al 1990), and thus AP2 cannot
`be the key to unleashing the cascade of keratinocyte—specific genes. Neverthe—
`less,
`these findings do suggest that
`the AP2 forms present in embryonic
`ectoderm are likely to play a role in KS and K14 activation in vivo, and in
`some cells, the absence of these factors may be sufficient to maintain these
`genes in an inactive state.
`Members of the API family of transcription factors are also found in the
`
`Page 12 of 35
`
`Thai: m=+r=ri=l WIPES rnniafl
`
`

`

`132
`
`FUCHS
`
`epidermis ofskin. These includejunB (Wilkinson et a1 1989) and cfos (Smeyne
`et al 1992 and references therein) in postnatal skin, andfosB in effl‘bryotnfi‘:
`epidermis (Redemann-Fibi et a1 1991). JunB and cfos resrde prrmarr I): lnAPI
`differentiating layers of epidermis, implicating these members of 1 6 m
`family in regulating differentiation-Specific functions in skin. ConSIStent W.l
`this notion is the finding that promoters active in terminally qlffcfem‘at'lng
`keratinocytes contain APl sites (Lu et al 1994 and references Within, see a so
`Casatorres et a] 1994). Among the best evidence that these srtes are funCtlonal
`comes from studies on the AP] site 3' to the K1 gene, which .haS been
`implicated in mediating the calcium-inducible, differentiation-specrftc expres-
`sion of this gene (Lu et al 1994).
`.
`A number of other factors have been localized to the epidermis, although
`their roles in controlling keratinocyte-Specific gene expression. have not been
`fully elucidated. One of these proteins is a zinc finger proteln, basonuclln,
`which is expressed in the basal layer of epidermis (Tseng & Green 1994)-
`Basonuclin is interesting in that it persists in cells that have withdrawn from
`the cell cycle, but it is absent in terminally differentiating cells. BaSOHUCIin
`has been postulated to be a key to the switch controlling the balance between
`growth and differentiation in keratinocytes.
`Several POU-specific proteins have also been found in epidermis. A new
`class II POU sequence, called Skn-la but related or identical to Oct-11, was
`detected in epidermis and hair follicles (Anderson et al 1993). Skn-la can
`specifically upregulate expression of a human KlO-luciferase reporter gene,
`suggesting a role in terminal differentiation (Anderson et a] 1993). XLPOU],
`a Xenopus class III POU protein, is also expressed in adult skin (Agarwal &
`Sato 1991). XLPOUI shares ~90% sequence homology with human Oct-6,
`which has been cloned from epidermal keratinocytes (Faus et a1 1994). Oct-6
`is intriguing in that it can act in both positive and negative fashions, depending
`on the gene and the tissue. In basal keratinocytes, Oct—6 suppresses keratin
`gene expression, and it may possibly have a role in downregulation of these
`genes during terminal differentiation (Faus et al 1994).
`Researchers are just beginning to decipher the sequences and transcription
`factors involved in controlling the expression of the hair-specific keratin genes.
`A number of potential regulatory motifs have been identified on the basis of
`sequence comparisons (reviewed in Rogers & Powell 1993). One of these sites,
`5'-C'ITI‘GAAGA-3’, referred to as the HK—l motif, was detected in four
`published hair keratin promoters. When the HK—l sequence motif was seen to
`overlap with a sequence identical to the motif of a known pair of lymphoid
`enhancer factors, LEF—l and TCF-l, it became apparent that all thirteen pub-
`lished hair keratin promoters contained LEF—l/I‘CF—l binding sites (Zhou et
`al 1995). Further studies showed that LEF—l plays a functional role in hair-
`specific morphogenesis and keratin gene expression. LEF—l is expressed early
`
`Page 13 of35
`
`Tth.l'lH-V5"1r:—|I ....,—n— _._..:....t
`
`

`

`KERATINS AND THE SKIN
`
`133
`
`.
`
`a
`
`in the development of embryonic skin, and it is produced in cells that first
`Induce ha1r-_specrfic gene expression (Zhou et a1 1995). Interestingly, LEF—l
`knockout mice have no whiskers and few hair follicles, further strengthening
`thelrgiggpn that LEF—l is involved in follicle morphogenesis (van Genderen et
`LEF-l belongs to the high-mobility group (HMG) family of proteins that
`are not conventional transcription factors, but rather act by bending DNA and
`altering chromatin to create a structure conducive for conventional transcrip-
`tion factor binding (Giese et a1 1992). It has been postulated that in hair
`follicles, LEF-l is involved in creating a DNA structure conducive to initiation,
`but not necessarily maintenance, of gene expression, and that additional factors
`are likely to be necessary for hair-specific gene BXpl‘ession (Zhou et al 1995).
`one POSSiblc candidate iS a Zinc finger protein recently shown to be the product
`0f the hairless gene in mice (Cachon-Gonzalez et a1 1994). Like the LEF-I
`knOCROUt mice, hairless mice Show a marked reduction and malformation of
`hair follicles, suggesting that they may be involved in the same pathway.
`What governs transcriptional regulation early in development? Taking cues
`from other systems, researchers have begun to investigate skin-specific ex-
`pression of the Hex class of transcription factors, known to specify positional
`information in segmentally derived structures, as well as in appendages such
`as limbs. Although epidermis is not a segmental structure, it overlies and is
`influenced by a dermis, which is, in part, segmentally derived. During em—
`bryogenesis, patterning and regionalization of the epidermis is likely to be
`under the influence of both dermal and epidermal Hox genes. An example of
`the former may be the segmental expression of the Moxl homeoprotein in
`E85 dermamyotome in mouse embryos, in a pattern that parallels E9.5 ex~
`pression of basal epidermal keratin genes in the overlying ectoderm (Byrne et
`al 1994 and references therein). At later developmental stages, mesodermal
`gradients of Hox proteins have been found in epidermal appendages of other
`vertebrates (reviewed in Chuong 1993), which suggests that positional infor-
`mation directing skin appendage patterning is determined by dermal Hox
`genes, perhaps in a fashion analogous to their mode of action in body append-
`
`ages.
`Hox genes are also expressed temporally in the ectodermal component of
`skin. Early in development, Hox 2 gene family members are expressed in the
`branchial arches of the head (Hunt et al 1991), and later the pattern of transient
`expression of additional members of this family suggests a role in specifying
`the differentiation status of epidermis (Mathews et a1 1993). This notion is
`strengthened by the recent discovery of two additional differentiation-activated
`homeoproteins, Xdll-3/Dlx—3 (Morasso et al 1993) and HOXC4 (Rieger et al
`1994).
`It is well established that retinoids influence patterns of homeobox gene
`Page 14 of 35
`
`

`

`134
`
`FUCHS
`
`ave been shown
`
`ms of epithelial differen-
`expression, and thus it is not surprising that progra
`ids. Retinoic acid at high
`.
`tiation in the skin are intricately controlled by retino
`nhibitory effects on
`(10‘6 M) concentration has long been known to have .1
`mRNAs (Fuchs &
`epidermal differentiation and on the expression of keratin
`Green 1981). Recently, lower concentrations of retinoic acid h
`f
`'
`to have a beneficial effect on the differentiation process. The effects 0 reti-
`noids on keratin expression are transcriptionally regulated (StellmaCh 3t 31
`1991, Tomic-Canic et a1 1992, Lu et al 1994), and keratinocytes express a
`number of retinoid receptors. These include retinoic acid receptors on and “Y
`(RAROL and RARy) and RXRoc (Viallet & Dhouailly 1994 and references
`therein). RARY and RXROL are expressed in neonatal skin and 1n only a few
`other organs, whereas RAROL is more broadly expressed. Developmentally,
`RARy mRNAs appear in epidermis prior to RXROL mRNAS, and In neonatal
`skin, RAR'y mRNAs are most abundant in the keratinizing layers of epidermis.
`The control of gene transcription by RARs and RXRs is complex. involving
`a multitude of both indirect and direct mechanisms (see Kastner et a1 1993).
`RARs can heterodimerize with thyroid hormone receptors and With RXRS, and
`at least some of these interactions change the DNA affinity and activity of
`RARs. RARs and/or RXRs can bind to thyroid response elements, retinoic
`acid response elements (RAREs), and retinoid X response elements (RXREs),
`and the repertoire of complex interactions is further expanded by the capacity
`of RARs and RXRs to interact with AP] proteins. The direct binding of RARs
`to epidermal keratin genes has not yet been demonstrated, although RA-me—
`diated biochemical changes in differentiation do appear to involve DNA se-
`quences in keratin genes that are responsive to retinoids (Tomic-Canic

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