Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
J Cell Biol ; 154(3): 619-30, 2001 Aug 06.
Article in English | MEDLINE | ID: mdl-11489919

ABSTRACT

The murine genome is known to have two keratin 6 (K6) genes, mouse K6 (MK6)a and MK6b. These genes display a complex expression pattern with constitutive expression in the epithelia of oral mucosa, hair follicles, and nail beds. We generated mice deficient for both genes through embryonic stem cell technology. The majority of MK6a/b-/- mice die of starvation within the first two weeks of life. This is due to a localized disintegration of the dorsal tongue epithelium, which results in the build up of a plaque of cell debris that severely impairs feeding. However, approximately 25% of MK6a/b-/- mice survive to adulthood. Remarkably, the surviving MK6a/b-/- mice have normal hair and nails. To our surprise, we discovered MK6 staining both in the hair follicle and the nail bed of MK6a/b-/- mice, indicating the presence of a third MK6 gene. We cloned this previously unknown murine keratin gene and found it to be highly homologous to human K6hf, which is expressed in hair follicles. We therefore termed this gene MK6 hair follicle (MK6hf). The presence of MK6hf in the MK6a/b-/- follicles and nails offers an explanation for the absence of hair and nail defects in MK6a/b-/- animals.


Subject(s)
Hair Diseases/genetics , Hair Diseases/pathology , Keratins/genetics , Nail Diseases/genetics , Nail Diseases/pathology , Animals , Epithelial Cells/pathology , Gene Deletion , Hair Diseases/mortality , Hyperplasia , Isomerism , Keratins/chemistry , Mice , Mice, Knockout , Microscopy, Electron , Molecular Sequence Data , Mouth Diseases/genetics , Mouth Diseases/mortality , Mouth Diseases/pathology , Nail Diseases/mortality , Phenotype , Sequence Homology, Amino Acid , Skin/pathology , Starvation/genetics , Starvation/mortality , Starvation/pathology , Tongue/pathology , Tongue/ultrastructure , Wound Healing/genetics
2.
J Cell Biol ; 152(3): 645-9, 2001 Feb 05.
Article in English | MEDLINE | ID: mdl-11157989

ABSTRACT

Stem cells are crucial for the formation and maintenance of tissues and organs. The role of stem cells in the pathogenesis of mosaic skin disorders remains unclear. To study the molecular and cellular basis of mosaicism, we established a mouse model for the autosomal-dominant skin blistering disorder, epidermolytic hyperkeratosis (MIM 113800), which is caused by mutations in either keratin K1 or K10. This genetic model allows activation of a somatic K10 mutation in epidermal stem cells in a spatially and temporally controlled manner using an inducible Cre recombinase. Our results indicate that lack of selective pressure against certain mutations in epidermal stem cells leads to mosaic phenotypes. This finding has important implications for the development of new strategies for somatic gene therapy of dominant genodermatoses.


Subject(s)
Hyperkeratosis, Epidermolytic/genetics , Keratins/genetics , Mosaicism/genetics , Point Mutation/genetics , Skin/pathology , Stem Cells/physiology , Viral Proteins , Animals , Disease Models, Animal , Female , Gene Targeting , Humans , Hyperkeratosis, Epidermolytic/pathology , Hyperkeratosis, Epidermolytic/physiopathology , Integrases/genetics , Integrases/metabolism , Keratin-10 , Keratins/metabolism , Mice , Mice, Transgenic , Mifepristone/pharmacology , Skin/drug effects , Skin/physiopathology
3.
J Cell Biol ; 152(3): 651-6, 2001 Feb 05.
Article in English | MEDLINE | ID: mdl-11157990

ABSTRACT

The Dowling-Meara variant of epidermolysis bullosa simplex (EBS-DM) is a severe blistering disease inherited in an autosomal-dominant fashion. Here we report the generation of a mouse model that allows focal activation of a mutant keratin 14 allele in epidermal stem cells upon topical administration of an inducer, resulting in EBS phenotypes in treated areas. Using laser capture microdissection, we show that induced blisters healed by migration of surrounding nonphenotypic stem cells into the wound bed. This observation provides an explanation for the lack of mosaic forms of EBS-DM. In addition, we show that decreased mutant keratin 14 expression resulted in normal morphology and functions of the skin. Our results have important implications for gene therapy of EBS and other dominantly inherited diseases.


Subject(s)
Disease Models, Animal , Epidermolysis Bullosa Simplex/genetics , Gene Expression Regulation , Keratins/genetics , Skin/physiopathology , Viral Proteins , Animals , Blotting, Southern , Epidermolysis Bullosa Simplex/pathology , Epidermolysis Bullosa Simplex/physiopathology , Epidermolysis Bullosa Simplex/therapy , Genetic Therapy , Humans , Integrases/genetics , Integrases/metabolism , Keratin-14 , Keratins/metabolism , Luteolytic Agents/pharmacology , Mice , Mice, Transgenic , Microscopy, Fluorescence , Mifepristone/administration & dosage , Mifepristone/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Skin/drug effects , Skin/ultrastructure
4.
J Cell Biol ; 151(2): 401-12, 2000 Oct 16.
Article in English | MEDLINE | ID: mdl-11038186

ABSTRACT

Mutations in the cornified cell envelope protein loricrin have been reported recently in some patients with Vohwinkel syndrome (VS) and progressive symmetric erythrokeratoderma (PSEK). To establish a causative relationship between loricrin mutations and these diseases, we have generated transgenic mice expressing a COOH-terminal truncated form of loricrin that is similar to the protein expressed in VS and PSEK patients. At birth, transgenic mice (ML.VS) exhibited erythrokeratoderma with an epidermal barrier dysfunction. 4 d after birth, high-expressing transgenic animals showed a generalized scaling of the skin, as well as a constricting band encircling the tail and, by day 7, a thickening of the footpads. Histologically, ML. VS transgenic mice also showed retention of nuclei in the stratum corneum, a characteristic feature of VS and PSEK. Immunofluorescence and immunoelectron microscopy showed the mutant loricrin protein in the nucleus and cytoplasm of epidermal keratinocytes, but did not detect the protein in the cornified cell envelope. Transfection experiments indicated that the COOH-terminal domain of the mutant loricrin contains a nuclear localization signal. To determine whether the ML.VS phenotype resulted from dominant-negative interference of the transgene with endogenous loricrin, we mated the ML.VS transgenics with loricrin knockout mice. A severe phenotype was observed in mice that lacked expression of wild-type loricrin. Since loricrin knockout mice are largely asymptomatic (Koch, P.K., P. A. de Viragh, E. Scharer, D. Bundman, M.A. Longley, J. Bickenbach, Y. Kawachi, Y. Suga, Z. Zhou, M. Huber, et al., J. Cell Biol. 151:389-400, this issue), this phenotype may be attributed to expression of the mutant form of loricrin. Thus, deposition of the mutant protein in the nucleus appears to interfere with late stages of epidermal differentiation, resulting in a VS-like phenotype.


Subject(s)
Deafness/etiology , Keratosis/etiology , Membrane Proteins/genetics , Amino Acid Sequence , Animals , Cell Compartmentation , Cell Membrane/chemistry , Deafness/genetics , Frameshift Mutation , Keratosis/genetics , Mice , Mice, Mutant Strains , Mice, Transgenic , Molecular Sequence Data , Nuclear Localization Signals , Phenotype , Protein Transport , Skin/pathology , Skin Physiological Phenomena/genetics , Syndrome
5.
J Cell Biol ; 151(2): 389-400, 2000 Oct 16.
Article in English | MEDLINE | ID: mdl-11038185

ABSTRACT

The epidermal cornified cell envelope (CE) is a complex protein-lipid composite that replaces the plasma membrane of terminally differentiated keratinocytes. This lamellar structure is essential for the barrier function of the skin and has the ability to prevent the loss of water and ions and to protect from environmental hazards. The major protein of the epidermal CE is loricrin, contributing approximately 70% by mass. We have generated mice that are deficient for this protein. These mice showed a delay in the formation of the skin barrier in embryonic development. At birth, homozygous mutant mice weighed less than control littermates and showed skin abnormalities, such as congenital erythroderma with a shiny, translucent skin. Tape stripping experiments suggested that the stratum corneum stability was reduced in newborn Lor(-/-) mice compared with wild-type controls. Isolated mutant CEs were more easily fragmented by sonication in vitro, indicating a greater susceptibility to mechanical stress. Nevertheless, we did not detect impaired epidermal barrier function in these mice. Surprisingly, the skin phenotype disappeared 4-5 d after birth. At least one of the compensatory mechanisms preventing a more severe skin phenotype in newborn Lor(-/-) mice is an increase in the expression of other CE components, such as SPRRP2D and SPRRP2H, members of the family of "small proline rich proteins", and repetin, a member of the "fused gene" subgroup of the S100 gene family.


Subject(s)
Epidermis/physiology , Membrane Proteins/genetics , Skin Physiological Phenomena/genetics , Adaptation, Biological , Amino Acid Sequence , Animals , Biomechanical Phenomena , Cell Membrane , Cloning, Molecular , Cornified Envelope Proline-Rich Proteins , Intermediate Filament Proteins/biosynthesis , Membrane Proteins/deficiency , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Permeability , S100 Proteins/biosynthesis , Up-Regulation
6.
Exp Dermatol ; 9(1): 16-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10688370

ABSTRACT

Epidermolytic hyperkeratosis (EHK) is a hereditary skin disorder typified by blistering due to cytolysis. One in 100,000 individuals is affected by this autosomal-dominant disease. The onset of the disease phenotype is typically at birth. Histological and ultrastructural examination of the epidermis shows a thickened stratum corneum and tonofilament clumping around the nucleus of suprabasal keratinocytes. Linkage studies localized the disease genes on chromosomes 12q and 17q which contain the type II and type I keratin gene clusters. Recently, several point mutations in the genes encoding the suprabasal keratins, K1 and K10, have been reported in EHK patients. We have investigated a large kindred affected by EHK and identified a new point mutation in the 2B region of keratin 1 (I107T), resulting from a T to C transition in codon 478.


Subject(s)
Hyperkeratosis, Epidermolytic/genetics , Keratins/genetics , Point Mutation , Alleles , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , DNA Primers/genetics , Female , Genotype , Heterozygote , Humans , Male , Pedigree , Phenotype , Polymerase Chain Reaction
7.
Differentiation ; 65(2): 97-112, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10550543

ABSTRACT

Mouse keratin 6a (MK6a) is constitutively expressed in a single cell layer of the outer root sheath (ORS) of hair follicles, but its synthesis can be induced in interfollicular epidermis including the basal cell layer in response to perturbing stimuli. A basally inducible human K6 (HK6) isoform has not been described, and it is not clear which of the known HK6 isoforms is expressed in the ORS. In this study we show that expression of a dominant-negative MK6a construct (Delta2B-P) in the interfollicular epidermis caused severe blistering and neonatal lethality, suggesting that mutations in a yet to be identified basally expressed HK6 isoform might result in a severe blistering phenotype. Surviving Delta2B-P animals showed transgene expression only in isolated epidermal cells and not in all cells of the ORS, but nevertheless developed severe alopecia. Expression of two different C-terminal mutant transgenes also caused alopecia while a third C-terminal mutant had no phenotypic conse- quences. Electron microscopy revealed that Delta2B-P expression resulted in the collapse of keratin filaments, while destruction of hair follicles in the two phenotypic C-terminal mutant lines occurred in the absence of filament abnormalities. The latter finding indicates that the innermost ORS cells are uniquely sensitive to expression of even slightly altered K6 proteins, suggesting that mutations affecting an HK6 isoform expressed in this cell layer could result in alopecia in humans as well.


Subject(s)
Epidermis/metabolism , Genes, Dominant , Hair Follicle/metabolism , Keratins/genetics , Transgenes , Age of Onset , Alopecia/genetics , Amino Acid Sequence , Animals , Animals, Newborn , Epidermis/pathology , Epidermis/ultrastructure , Gene Expression , Hair Follicle/pathology , Hair Follicle/ultrastructure , Keratins/biosynthesis , Keratins/ultrastructure , Mice , Mice, Inbred BALB C , Mice, Transgenic , Microscopy, Electron , Molecular Sequence Data , Mutagenesis, Site-Directed , Phenotype , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Skin Diseases/genetics , Skin Diseases/pathology , Time Factors
8.
Differentiation ; 65(2): 119-30, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10550545

ABSTRACT

Keratin 6 (K6) is expressed constitutively in a variety of internal stratified epithelia as well as in palmoplantar epidermis and in specialized cells of the hair follicle. K6 expression can also be induced by hyperproliferative conditions as in wound healing or by conditions that perturb normal keratinocyte function. The functional significance of the expression of K6 on keratinocyte biology under these disparate conditions is not known. Here we report on the characterization of two isoforms of mouse K6 that are encoded by separate genes. The two genes (denoted K6a and K6b) are linked, have the same orientation and are actively transcribed. Sequence analysis revealed, that although they encode almost identical products, they have distinctly different regulatory regions, suggesting that the two K6 genes would be differentially expressed. In an attempt to define the expression characteristics of the K6 isoforms, we produced transgenic mice with each gene after modifying the C-terminal sequences to enable detection of the transgenic proteins with specific antibodies. The constitutive expression of the K6a transgene paralleled that of the endogenous genes in all K6 expressing tissues, except in the tongue. The K6b transgene was also expressed in these tissues but, in contrast to K6a, was only expressed in suprabasal cells. Both K6 transgenes were also induced in the interfollicular epidermis in response to phorbol esters, with K6a induced in all layers of the treated epidermis, while K6b was expressed only in suprabasal cells. These studies suggest that the K6 isoforms have overlapping yet distinct expression profiles.


Subject(s)
Epidermis/metabolism , Hair Follicle/metabolism , Hindlimb/metabolism , Keratins/biosynthesis , Keratins/genetics , Tongue/metabolism , Animals , Blotting, Western , Fluorescent Antibody Technique , Gene Expression Regulation , Genetic Linkage , Mice , Mice, Inbred BALB C , Mice, Transgenic , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
9.
Differentiation ; 64(4): 225-35, 1999 May.
Article in English | MEDLINE | ID: mdl-10365440

ABSTRACT

We have previously shown that the promoter of a 6.5 kb mouse loricrin clone contains a functional AP-1 element and directs tissue-specific, but not differentiation-specific, expression. We now report the isolation of a 14-kb genomic clone containing an additional 7 kb of genomic sequence. The additional sequences limit expression of a reporter construct to differentiated keratinocytes in culture. The expression of the 6.5-kb and 14-kb loricrin constructs were also analyzed in transgenic mice. Significantly, loricrin was found in all layers of the epidermis of the 6.5-kb transgenics, including basal and spinous cells. The expression of the 14-kb clone was indistinguishable from that of the endogenous gene, confirming that the additional sequences contain negative regulatory elements that restrict loricrin expression to the granular layer in vivo. In addition, we show the AP-1 element localized in the loricrin proximal promoter is necessary but not sufficient for expression of the loricrin gene in vivo in transgenic mice. Finally, to gain further insight into how AP-1 family members regulate expression of the loricrin gene, we co-transfected the loricrin reporter constructs with expression plasmids for various fos and jun family members and demonstrated that c-Fos/Jun-B heterodimers could mimic the differentiation-specific induction of loricrin.


Subject(s)
Gene Expression Regulation , Keratinocytes/cytology , Membrane Proteins/genetics , Animals , Animals, Newborn , Cell Differentiation , Cells, Cultured , Chloramphenicol O-Acetyltransferase/genetics , Cloning, Molecular , Dimerization , Genes, Reporter , Genomic Library , Keratinocytes/physiology , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Recombinant Proteins/biosynthesis , Regulatory Sequences, Nucleic Acid , Restriction Mapping , Skin/cytology , Skin/metabolism , Transcription Factor AP-1/metabolism , Transfection , beta-Galactosidase/genetics
10.
Exp Dermatol ; 8(2): 124-7, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10232403

ABSTRACT

Epidermolytic hyperkeratosis (EHK) is a congenital, autosomal dominant disorder of cornification characterized by hyperkeratosis and blister formation. The clinical manifestations are heterogeneous, with respect to the extent of body surface involvement, palmar and plantar hyperkeratosis and the presence of erythroderma. Point mutations in the genes encoding the suprabasal-specific keratins, keratins 1 and 10 have been identified in EHK patients. The inappropriate amino acid substitutions cause a collapse of the keratin filament network, resulting in cytolysis of the involved keratinocytes. We report a severe case of EHK with a single base pair mutation that causes a threonine for asparagine substitution in residue 8 (N8T) of the 1A region of the keratin 1 protein. This is the region involved in molecular overlaps between neighboring keratin heterodimers. These findings suggest that even conservative amino acid substitutions in overlap regions can cause tonofilament clumping.


Subject(s)
Amino Acid Substitution , Asparagine , Hyperkeratosis, Epidermolytic/genetics , Keratins/genetics , Point Mutation , Threonine , Amino Acid Sequence , Base Sequence , Female , Follow-Up Studies , Humans , Infant, Newborn , Keratins/chemistry , Male , Pedigree
11.
J Invest Dermatol ; 112(4): 506-8, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10201536

ABSTRACT

Epidermolytic hyperkeratosis is characterized by tonofilament clumping, cytolysis, and blister formation in suprabasal keratinocytes. It has been shown that the tonofilament aggregates in these areas are composed of keratin 1 (K1) and keratin 10 (K10), and several K1 and K10 point mutations have been identified as the molecular basis of epidermolytic hyperkeratosis. In this report we identify a novel, single base pair substitution resulting in an amino acid exchange from tyrosine to serine at residue 14 within the conserved 1A region of K10 (Y14S). This A to C transversion in codon 160 was only present in the affected individual and was associated with a very severe disease phenotype. Our observations are in agreement with previous reports documenting that this tyrosine residue, located at the beginning of the rod domain of type I keratins, is particularly sensitive to amino acid substitutions, and that alterations in this residue can have deleterious effects on filament assembly and stability.


Subject(s)
Hyperkeratosis, Epidermolytic/genetics , Keratins/genetics , Point Mutation , Child, Preschool , Female , Humans , Hyperkeratosis, Epidermolytic/pathology , Keratin-10 , Keratins/chemistry , Male
12.
J Invest Dermatol ; 112(3): 380-2, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10084318

ABSTRACT

Ichthyosis bullosa of Siemens (IBS) is a rare autosomal dominant skin disorder with clinical features similar to epidermolytic hyperkeratosis (EHK). Both diseases have been linked to the type II keratin cluster on chromosome 12q. Hyperkeratosis and blister formation are relatively mild in IBS compared with EHK, and the lysis of keratinocytes is restricted to the upper spinous and granular layers of the epidermis of IBS patients, whereas in EHK lysis occurs in the lower spinous layer. Recently, mutations in the helix initiation and termination motifs of keratin 2e (K2e) have been described in IBS patients. The majority of the mutations reported to date lie in the 2B region. In this report, we have examined a large kindred in which the disease was originally diagnosed as EHK and mapped to the type II keratin cluster on chromosome 12q. Molecular analysis revealed a novel amino acid substitution at the beginning of the conserved 1A region of the rod domain (I4N) of K2e, resulting from a T to A transversion in codon 188.


Subject(s)
Ichthyosis/genetics , Keratins/genetics , Mutation/genetics , Skin Diseases, Vesiculobullous/genetics , Amino Acid Sequence/genetics , Base Sequence/genetics , DNA Mutational Analysis , Female , Humans , Keratin-2 , Male , Pedigree
13.
Prenat Diagn ; 18(8): 826-30, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9742571

ABSTRACT

Epidermolytic hyperkeratosis (bullous congenital ichthyosiform erythroderma) is an autosomal dominant skin disorder caused by mutations in keratins 1 and 10. We have used direct gene sequencing to ascertain the status of a 15 week fetus of parents whose first child was affected with this disorder. The parents show no clinical signs of epidermolytic hyperkeratosis but were concerned about the possibility of transmitting the disorder due to germline mosaicism. Molecular analysis of the affected son revealed a G to A mutation in codon 156 of keratin 10, resulting in an arginine to histidine substitution within the highly conserved 1A region. Codon 156 has been previously identified as a mutational hot spot and substitutions of this arginine residue are very common in epidermolytic hyperkeratosis patients. Analysis of genomic DNA isolated from amniotic cells showed that the fetus did not harbour this mutation and a healthy infant was eventually born that was unaffected by this disorder.


Subject(s)
Hyperkeratosis, Epidermolytic/diagnosis , Hyperkeratosis, Epidermolytic/genetics , Keratins/genetics , Mutation , Prenatal Diagnosis , Amniotic Fluid/cytology , DNA Mutational Analysis , DNA Restriction Enzymes , Female , Humans , Mosaicism , Point Mutation , Polymerase Chain Reaction , Pregnancy , Sequence Analysis, DNA
14.
J Invest Dermatol ; 108(3): 357-61, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9036939

ABSTRACT

Annular epidermolytic ichthyosis has recently been delineated as a distinct clinical phenotype within the spectrum of epidermolytic keratinization disorders. The pattern of inheritance of the disorder is consistent with an autosomal dominant mode of transmission. Here we report a second incidence of this disorder in a family with two affected generations. The proband suffered from bullous ichthyosis and had bouts of disease activity associated with the development of numerous annular and polycyclic erythematous, hyperkeratotic plaques on the trunk and the proximal extremities. Histologic examination showed the typical pathology of epidermolytic hyperkeratosis, and ultrastructural analysis revealed abnormal keratin filament networks and tonofilament clumping with a perinuclear distribution. Molecular analysis revealed a novel tandem CG to GA 2-bp mutation in the same allele of keratin 10 in affected individuals, resulting in an arginine to glutamate substitution at residue 83 (R83E) of the 2B helical segment. We conclude that annular epidermolytic ichthyosis should be considered a variant of bullous congenital ichthyosiform erythroderma.


Subject(s)
Hyperkeratosis, Epidermolytic/genetics , Keratins/genetics , Point Mutation , Adult , Alleles , Biopsy , Extremities/pathology , Female , Genetic Variation , Humans , Hyperkeratosis, Epidermolytic/diagnosis , Hyperkeratosis, Epidermolytic/pathology , Male , Pedigree , Phenotype , Sequence Analysis, DNA , Skin/ultrastructure
15.
Differentiation ; 61(2): 129-39, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8983179

ABSTRACT

Keratins are the major structural proteins of keratinocytes, which are the most abundant cell type in the mammalian epidermis. Mutations in epidermal keratin genes have been shown to cause severe blistering skin abnormalities. One such disease, epidermolytic hyperkeratosis (EHK), also known as bullous congenital ichthyosiform erythroderma, occurs as a result of mutations in highly conserved regions of keratins K1 and K10. Patients with EHK first exhibit erythroderma with severe blistering, which later is replaced by thick patches of scaly skin. To assess the effect of a mutated K1 gene on skin biology and to produce an animal model for EHK, we removed 60 residues from the 2B segment of HK1 and observed the effects of its expression in the epidermis of transgenic mice. Phenotypes of the resultant mice closely resembled those observed in the human disease, first with epidermal blisters, then later with hyperkeratotic lesions. In neonatal mice homozygous for the transgene, the skin was thicker, with an increased labeling index, and the spinous cells showed a collapse of the keratin filament network around the nuclei, suggesting that a critical concentration of the mutant HK1, over the endogenous MK1, was required to disrupt the structural integrity of the spinous cells. Additionally, footpad epithelium, which is devoid of hair follicles, showed blistering in the spinous layer, suggesting that hair follicles can stabilize or protect the epidermis from trauma. Blisters were not evident in adult mice, but instead they showed a thick, scaly hyperkeratotic skin with increased mitosis, resulting in an increased number of corneocytes and granular cells. Irregularly shaped keratohyalin granules were also observed. To date, this is the only transgenic model to show the typical morphology found in the adult form of EHK.


Subject(s)
Hyperkeratosis, Epidermolytic/genetics , Keratins/genetics , Mice, Transgenic/genetics , Age Factors , Animals , Animals, Newborn , Disease Models, Animal , Epidermis/metabolism , Epithelium/pathology , Female , Humans , Hyperplasia/genetics , Keratins/biosynthesis , Keratins/ultrastructure , Male , Mice , Mice, Inbred ICR , Mice, Inbred Strains , Mutation , Phenotype , Skin/pathology , Transgenes
16.
J Biol Chem ; 270(18): 10792-9, 1995 May 05.
Article in English | MEDLINE | ID: mdl-7738016

ABSTRACT

Loricrin gene expression is limited to terminally differentiating keratinocytes of stratified squamous epithelia. To define the regulatory elements that mediate the expression of the loricrin gene, we replaced the loricrin coding sequences from a 6.5-kilobase genomic fragment with the chloramphenicol acetyltransferase gene and transfected this construct into cultured mouse keratinocytes. High expression levels were observed in both undifferentiated as well as differentiating cells. Transgenic mice bearing a similar construct, but with beta-galactosidase as the reporter gene, corroborated these in vitro findings and showed tissue- and cell type-specific, but not differentiation-specific expression. Deletion analysis of the promoter region determined that sequences up to -60 base pairs from the start of transcription could be removed without significant loss of promoter activity. Within these proximal 60 base pairs is an evolutionarily conserved AP-1 element that is recognized by both purified c-Jun and AP-1 factors from keratinocytes in vitro. Mutation of this AP-1 site abolished the activity of the loricrin promoter. These studies show that elements directing expression of the loricrin gene to the stratified squamous epithelia are contained within a 6.5-kilobase genomic fragment, and those elements required to restrict expression to differentiated keratinocytes lie outside this region.


Subject(s)
Genes , Membrane Proteins/genetics , Promoter Regions, Genetic , Transcription Factor AP-1/metabolism , Animals , Base Sequence , Binding Sites , Cell Differentiation , DNA Primers/chemistry , Gene Expression Regulation , Keratinocytes/physiology , Mice , Molecular Sequence Data , Sequence Alignment , Sequence Homology, Nucleic Acid , Transcription, Genetic
17.
J Invest Dermatol ; 104(3): 313-7, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7860993

ABSTRACT

To assess the potential of an in vivo, adenovirus-mediated gene therapy approach for the treatment of malignant melanoma, the efficacy of adenovirus-mediated herpes simplex virus thymidine kinase gene (HSV-Ek) transfer and administration of ganciclovir (GCV) was investigated using a nude mouse model. Initially, B16 murine melanoma cells were efficiently transduced in vitro by a recombinant replication-defective adenovirus containing the HSV-tk gene (ADV/RSVtk), and rendered sensitive to cell killing by 10 micrograms/ml GCV. A significant "bystander effect" was observed at low multiplicity of infection in comparison of cell killing to control B16 transduction by adenovirus containing the beta-galactosidase gene (ADV/RSV-beta-gal). In vivo, melanomas established from subcutaneous injection of 4 x 10(5) B16 cells were injected after 14 d with 1 x 10(10) ADV/RSV-tk viral particles. Subsequent treatment for 6 d with GCV resulted in an inhibition of melanoma growth, with an approximately 40-50% reduction in melanoma volume in comparison to controls in repeated experiments. These data demonstrate that adenovirus-mediated gene transfer can function as an efficient delivery system to reduce established tumor burden in vivo. This result may hold significant promise for the development of effective in situ gene therapy for melanoma in humans.


Subject(s)
Adenoviridae/genetics , Genetic Therapy , Genetic Vectors , Melanoma, Experimental/prevention & control , Simplexvirus/genetics , Thymidine Kinase/genetics , Animals , Ganciclovir/therapeutic use , Gene Transfer Techniques , Melanoma, Experimental/genetics , Mice , Transduction, Genetic , Tumor Cells, Cultured
18.
Genomics ; 23(2): 450-6, 1994 Sep 15.
Article in English | MEDLINE | ID: mdl-7835895

ABSTRACT

Loricrin is the major component of a specialized structure, termed the cornified cell envelope, that is formed beneath the plasma membrane of stratified squamous epithelial cells and is coexpressed with profilaggrin in terminally differentiating epidermal keratinocytes. Full-length cDNAs for both mouse and human loricrin have been cloned and characterized, as has the human gene. Here we report the isolation and characterization of the mouse loricrin gene. The gene has a simple structure consisting of a single intron of 1091 bp within the 5' noncoding sequence and an uninterrupted open reading frame. Using PCR analyses of DNAs isolated from mouse x Chinese hamster somatic cell hybrids, we have mapped both the loricrin and the profilaggrin genes to chromosome 3. Genetic linkage analysis has shown that mouse loricin and profilaggrin lie within 1.5 +/- 1.1 centimorgans of each other. We have further shown that both genes map in the vicinity of the flaky tail (ft) and soft coat (soc) loci. These mouse mutants exhibit a number of changes in their integument, suggesting that abnormalities in these genes may contribute to the mutant phenotype.


Subject(s)
Genetic Linkage , Intermediate Filament Proteins/genetics , Membrane Proteins/genetics , Protein Precursors/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Cloning, Molecular , Cricetinae , DNA, Complementary/genetics , Filaggrin Proteins , Humans , Hybrid Cells , Introns , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Molecular Sequence Data , Mutation , Open Reading Frames , Phenotype , Polymerase Chain Reaction , Rats
19.
J Dermatol Sci ; 7 Suppl: S164-9, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7528048

ABSTRACT

Recent advances have challenged the prevailing view that keratins are merely passive bystanders of keratinocyte biology. With the exciting discovery that three autosomal dominant genetic skin disorders, epidermolysis bullosa simplex (EBS), epidermolytic hyperkeratosis (EHK) and palmoplantar keratoderma (PPK), are in fact disorders of keratins comes the realization that the integrity of the keratin filament network is crucial to the structural integrity of the skin. Since it has been recently established that mutations in keratins K5/K14, K1/K10 and K9 are causative for these keratinocyte disorders, it is very likely that mutations in K6 or in its obligate partner, K16 will result in disease. In order to test this we have produced transgenic mice that express a mutant K6 gene. These mice develop a progressive scarring alopecia at about 6 months of age. Later, the denuded areas developed a keratosis which was prone to infection. Ultrastructural analysis suggests that hair loss is due to the destruction of the outer root sheath. We believe that these mice are models of another keratin disorder.


Subject(s)
Alopecia/genetics , Keratins/genetics , Alopecia/classification , Alopecia/pathology , Animals , Disease Models, Animal , Epidermis/metabolism , Epidermis/pathology , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Keratins/chemistry , Keratins/metabolism , Mice , Mice, Transgenic , Molecular Structure , Multigene Family , Mutation
20.
Cell Growth Differ ; 5(6): 667-75, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7522035

ABSTRACT

In order to create a transgenic model for human papilloma virus (HPV)-associated carcinogenesis, we have used the regulatory elements of a human keratin K1 (HK1) gene to target the expression of the E6 and E7 oncogenes of HPV-18 exclusively to the epidermis. All murine expressors were viable and lived normal lifetimes; older mice (> 1 year) possessed numerous small lesions with a verrucous (wart-like) histotype. Analysis of newborn epidermis and lesions revealed that the HPV-18 E6/E7 genes were being expressed with a predominance of the E6*/E7 transcript over the full length E6/E7 message. The long latency in lesion appearance may reflect the low level of intact E6 transcripts and the requirement for additional genetic or epigenetic events before production of an overt lesion. In agreement with this proposal, spontaneous papillomas developed that expressed an activated rasHa oncogene (codon 61, A-->T; codon 13, G-->T). All lesions expressed keratin genes K1, K6, and K13 in a fashion characteristic of hyperproliferative or benign tumors with no evidence of malignant conversion. Our results demonstrate that the mouse epidermis represents a relevant in vivo model system to analyze the interaction between HPV and cellular genes in neoplasia.


Subject(s)
DNA-Binding Proteins , Mice, Transgenic , Oncogene Proteins, Viral/genetics , Papilloma/genetics , Papillomaviridae/genetics , Animals , Base Sequence , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , DNA Probes, HPV , Epidermis/pathology , Genes, ras , Keratins/genetics , Mice , Molecular Sequence Data , Papilloma/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...