Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 174(1): 115-25, 2006 Jul 03.
Article in English | MEDLINE | ID: mdl-16818723

ABSTRACT

Keratin 8 (K8) variants predispose to human liver injury via poorly understood mechanisms. We generated transgenic mice that overexpress the human disease-associated K8 Gly61-to-Cys (G61C) variant and showed that G61C predisposes to liver injury and apoptosis and dramatically inhibits K8 phosphorylation at serine 73 (S73) via stress-activated kinases. This led us to generate mice that overexpress K8 S73-to-Ala (S73A), which mimicked the susceptibility of K8 G61C mice to injury, thereby providing a molecular link between K8 phosphorylation and disease-associated mutation. Upon apoptotic stimulation, G61C and S73A hepatocytes have persistent and increased nonkeratin proapoptotic substrate phosphorylation by stress-activated kinases, compared with wild-type hepatocytes, in association with an inability to phosphorylate K8 S73. Our findings provide the first direct link between patient-related human keratin variants and liver disease predisposition. The highly abundant cytoskeletal protein K8, and possibly other keratins with the conserved S73-containing phosphoepitope, can protect tissue from injury by serving as a phosphate "sponge" for stress-activated kinases and thereby provide a novel nonmechanical function for intermediate filament proteins.


Subject(s)
Disease Models, Animal , Keratins/physiology , Liver Diseases/genetics , Animals , Apoptosis/drug effects , Apoptosis/physiology , Chemical and Drug Induced Liver Injury , Fas Ligand Protein , Genetic Predisposition to Disease , Genetic Variation , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Intermediate Filament Proteins/metabolism , Intermediate Filament Proteins/physiology , Keratin-8 , Keratins/antagonists & inhibitors , Keratins/deficiency , Liver Diseases/pathology , Liver Function Tests , Marine Toxins , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/chemistry , Mice , Mice, Knockout , Mice, Transgenic , Microcystins , Mitogen-Activated Protein Kinases/physiology , Mutation , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/chemistry , Phosphorylation , Tumor Necrosis Factors/administration & dosage , Tumor Necrosis Factors/chemistry
3.
Nature ; 441(7091): 362-5, 2006 May 18.
Article in English | MEDLINE | ID: mdl-16710422

ABSTRACT

Cell growth, an increase in mass and size, is a highly regulated cellular event. The Akt/mTOR (mammalian target of rapamycin) signalling pathway has a central role in the control of protein synthesis and thus the growth of cells, tissues and organisms. A striking example of a physiological context requiring rapid cell growth is tissue repair in response to injury. Here we show that keratin 17, an intermediate filament protein rapidly induced in wounded stratified epithelia, regulates cell growth through binding to the adaptor protein 14-3-3sigma. Mouse skin keratinocytes lacking keratin 17 (ref. 4) show depressed protein translation and are of smaller size, correlating with decreased Akt/mTOR signalling activity. Other signalling kinases have normal activity, pointing to the specificity of this defect. Two amino acid residues located in the amino-terminal head domain of keratin 17 are required for the serum-dependent relocalization of 14-3-3sigma from the nucleus to the cytoplasm, and for the concomitant stimulation of mTOR activity and cell growth. These findings reveal a new and unexpected role for the intermediate filament cytoskeleton in influencing cell growth and size by regulating protein synthesis.


Subject(s)
Cytoskeleton/metabolism , Epithelial Cells/cytology , Keratins/metabolism , Protein Biosynthesis , 14-3-3 Proteins/metabolism , Animals , Cell Growth Processes , Cells, Cultured , Cytoplasm/metabolism , Ectoderm/cytology , Epithelial Cells/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Keratins/deficiency , Keratins/genetics , Mice , Protein Binding , Protein Kinases/metabolism , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases
4.
Exp Dermatol ; 14(7): 481-90, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15946235

ABSTRACT

The purpose of this study is to reproduce in vitro a recessive keratinization defect of Norfolk terrier dogs characterized by a lack of keratin 10 (K10) production. Keratinocytes from skin biopsy samples of four normal dogs and two affected dogs were cultured organotypically with growth factor-supplemented media in order to stimulate cornification. The cultured epidermis from the normal dogs closely resembled the normal epidermis in vivo and cornified. The cultured epidermis from the affected dogs displayed many phenotypic alterations identified in skin biopsies from dogs with this heritable defect. Immunohistochemistry and immunoblotting showed a marked decrease in K10 from the cultures of the affected keratinocytes, compared to that in K10 from the cultures of the normal keratinocytes. Real-time reverse transcription polymerase chain reaction quantitation showed a 31-fold decrease in K10, a 1.75-fold increase in K1 and a 136-fold increase in K2e between the affected and the normal epidermis. Organotypic keratinocytes showed a 241-fold decrease in K10, a 31-fold decrease in K1 and a 1467-fold decrease in K2e between the affected and normal cultures. Although in vitro keratin expression did not precisely simulate in vivo, the morphology of the normal and the affected epidermis was largely preserved; thus, this culture system may provide an alternative to in vivo investigations for cutaneous research involving cornification.


Subject(s)
Dog Diseases/pathology , Keratins/deficiency , Keratins/genetics , Skin Diseases, Genetic/veterinary , Animals , Base Sequence , Cell Culture Techniques , Cells, Cultured , DNA/genetics , Dog Diseases/genetics , Dog Diseases/metabolism , Dogs , Keratin-10 , Keratinocytes/metabolism , Keratinocytes/pathology , Keratins/metabolism , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Skin Diseases, Genetic/genetics , Skin Diseases, Genetic/metabolism , Skin Diseases, Genetic/pathology
5.
Expert Opin Biol Ther ; 4(9): 1435-43, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15335311

ABSTRACT

Easy access to the organ and identification of underlying mutations in epidermolysis bullosa (EB) facilitated the first cutaneous gene therapy experiments in vitro in the mid-1990s. The leading technology was transduction of the respective cDNA carried by a retroviral vector. Using this approach, the genotypic and phenotypic hallmark features of the recessive forms of junctional EB, which are caused by loss of function of the structural proteins laminin-5 or bullous pemphigoid antigen 2/type XVII collagen of the dermo-epidermal basement membrane zone, have been corrected in vitro and in vivo using xenograft mouse models. Recently, this approach has also been shown to be feasible for the large COL7A1 gene (mutated in dystrophic EB), applying PhiC31 integrase or lentiviral vectors. Neither of these approaches has made it into a successful Phase I study on EB patients. Therefore, alternative approaches to gene correction, including modulation of splicing, are being investigated for gene therapy in EB.


Subject(s)
Epidermolysis Bullosa/therapy , Genetic Therapy , Animals , Autoantigens/genetics , Autoantigens/physiology , Carrier Proteins , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/physiology , Cytoskeletal Proteins , DNA, Complementary/genetics , DNA-Binding Proteins , Disease Models, Animal , Dystonin , Epidermolysis Bullosa/classification , Epidermolysis Bullosa/genetics , Genetic Heterogeneity , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Humans , Integrin beta4/genetics , Integrin beta4/physiology , Keratin-14 , Keratinocytes/metabolism , Keratinocytes/transplantation , Keratins/deficiency , Keratins/genetics , Keratins/physiology , Matrix Metalloproteinases/physiology , Mice , Mice, Nude , Nerve Tissue Proteins , Non-Fibrillar Collagens/deficiency , Non-Fibrillar Collagens/genetics , Non-Fibrillar Collagens/physiology , Protease Inhibitors/therapeutic use , RNA Splicing , RNA, Catalytic/therapeutic use , Telomerase/genetics , Telomerase/physiology , Kalinin , Collagen Type XVII
6.
J Comp Pathol ; 130(4): 246-54, 2004 May.
Article in English | MEDLINE | ID: mdl-15053927

ABSTRACT

Although well-characterized in man, abnormal cornification secondary to heritable superficial keratin defects is rarely reported in animals. This report describes a mild cornification defect in seven related Norfolk terrier dogs. Lesions were present at birth and pedigree analysis suggested an autosomal recessive mode of inheritance. The affected dogs had hyperpigmented skin with scaling following mild trauma. The lesions were generalized but most prominent in the glabrous skin of the axillary and inguinal regions-areas where the epidermis is not protected by hair and is subject to frequent trauma. The most striking histological change was vacuolation in the upper epidermis, which often resulted in epidermolysis and blister formation. All of the affected dogs showed similar gross and histological changes. Ultrastructural changes included abnormal keratin filament clumping, prominent clear spaces in the cytoplasm of suprabasal keratinocytes, and abnormal keratohyaline granules. Immunohistochemical labelling for keratin 10 demonstrated a lack of expression in the superficial epidermis of affected dogs. All of the morphological changes noted in the Norfolk terriers were consistent with a mild form of a heritable defect in superficial keratin synthesis.


Subject(s)
Epidermis/pathology , Keratins/deficiency , Skin Diseases/genetics , Skin Diseases/veterinary , Animals , Dogs , Epidermis/ultrastructure , Female , Immunohistochemistry , Keratin-10 , Male , Microscopy, Electron , Pedigree
7.
Biol Reprod ; 70(5): 1400-10, 2004 May.
Article in English | MEDLINE | ID: mdl-14724135

ABSTRACT

Acrosome biogenesis involves the transport and fusion of Golgi-derived proacrosomal vesicles along the acroplaxome, an F-actin/keratin 5-containing cytoskeletal plate anchored to the spermatid nucleus. A significant issue is whether the acroplaxome develops in acrosomeless mutant mice. Male mice with a Hrb null mutation are infertile and both spermatids and sperm are round-headed and lack an acrosome. Hrb, a protein that contains several NPF motifs (Asn-Pro-Phe) and interacts with proteins with Eps15 homology domains, is regarded as critical for the docking and/or fusion of Golgi-derived proacrosomal vesicles. Here we report that the lack of an acrosome in Hrb mutant spermatids does not prevent the development of the acroplaxome. Yet the acroplaxome in the mutant contains F-actin but is deficient in keratin 5. We also show that the actin-based motor protein myosin Va and its receptor, Rab27a/b, known to be involved in vesicle transport, are present in the Golgi and Golgi-derived proacrosomal vesicles in wild-type and Hrb mutant mouse spermatids. In the Hrb mutant, myosin-Va-bound proacrosome vesicles tether to the acroplaxome, where they flatten and form a flat sac, designated pseudoacrosome. As spermiogenesis advances, round-shaped spermatid nuclei of the mutant display several nuclear protrusions, designated nucleopodes. Nucleopodes are consistently found at the acroplaxome- pseudoacrosome site. Our findings support the interpretation that the acroplaxome provides a focal point for myosin-Va/ Rab27a/b-driven proacrosomal vesicles to accumulate, coalesce, and form an acrosome in wild-type spermatids and a pseudoacrosome in Hrb mutant spermatids. We suggest that nucleopodes develop at a site where a keratin 5-deficient acroplaxome may not withstand tension forces operating during spermatid nuclear shaping.


Subject(s)
Acrosome/physiology , Carrier Proteins/genetics , Golgi Apparatus/metabolism , Mutation , Myosin Heavy Chains/metabolism , Myosin Type V/metabolism , Spermatids/physiology , rab GTP-Binding Proteins/metabolism , Actins/metabolism , Adaptor Proteins, Vesicular Transport , Animals , Fluorescent Antibody Technique, Indirect , Immunohistochemistry , Keratin-15 , Keratin-5 , Keratins/deficiency , Male , Mice , Microscopy, Electron , Microscopy, Immunoelectron , Spermatids/ultrastructure , rab27 GTP-Binding Proteins
10.
J Invest Dermatol ; 121(4): 674-80, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14632181

ABSTRACT

Reduced coat 3 (Rco3) is a new spontaneous autosomal recessive mutation with defects in hair structure and progressive alopecia. Here we describe chromosomal mapping and molecular identification of the Rco3 mutation. The murine Rco3 locus maps to a 2-Mb interval on chromosome 15 encompassing the keratin type II gene cluster. Recently, mK6irs1 was described as a type II keratin expressed in Henle's and Huxley's layer of the murine inner root sheath. Genomic sequencing revealed a 10-bp deletion in exon 1 of mK6irs1 resulting in a frameshift after 58 amino acid residues and, therefore, the absence of 422 carboxy-terminal amino acid residues containing the complete alpha-helical rod domain. Henle's and Huxley's layers show no immunoreactivity with mK6irs1-specific antibodies and the absence of intermediate filament formation in electron microscopic images. These results indicate that the expression of functional mK6irs1 is indispensable for intermediate filament formation in the inner root sheath and highlights the importance of the keratinization of the inner root sheath in the normal formation of the hair shaft.


Subject(s)
Alopecia/genetics , Frameshift Mutation , Keratins/genetics , Alopecia/physiopathology , Amino Acid Sequence , Animals , Base Sequence , Chromosomes, Mammalian , Cloning, Molecular , Disease Models, Animal , Keratins/deficiency , Mice , Mice, Inbred AKR , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Molecular Sequence Data , Phenotype
11.
Am J Pathol ; 163(5): 1771-9, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14578178

ABSTRACT

Recessive epidermolysis bullosa simplex (REBS) is characterized by generalized cutaneous blistering in response to mechanical trauma. This results from fragility of the basal keratinocytes that lack keratin tonofilaments because of homozygote null mutation in the keratin 14 gene. REBS patients display in addition focal dyskeratotic skin lesions with histology of epidermolytic hyperkeratosis (EHK) and tonofilament clumping in the suprabasal layers of the epidermis. In this study we examined whether it is possible to mimic in vitro the bullous and dyskeratotic cellular phenotype. For this purpose, fibroblasts from nondyskeratotic (K14-/-) and dyskeratotic (K14-/-) skin of a REBS patient and fibroblasts from a healthy donor (K14+/+) were isolated and incorporated into collagen matrices. Subsequently, fresh biopsies originating from the nondyskeratotic and dyskeratotic skin of the patient and from a healthy donor were placed onto the collagen matrices and cultured at the air-liquid interface. Epidermal morphogenesis was evaluated on the basis of tissue morphology and the expression of a series of keratins. The results of the present study indicate that basal cell vacuolization in REBS can be mimicked in vitro but not the EHK. Fibroblasts seem to play an important regulatory role in establishing the REBS phenotype. These findings suggest that wild-type fibroblasts may enhance the stability of K14-/- keratinocytes in vitro.


Subject(s)
Epidermis/pathology , Epidermolysis Bullosa/genetics , Keratinocytes/physiology , Keratins/genetics , Biopsy , Cell Culture Techniques/methods , Cells, Cultured , Epidermis/physiology , Epidermis/ultrastructure , Epidermolysis Bullosa/pathology , Fibroblasts , Genes, Recessive , Humans , Immunohistochemistry , Keratin-14 , Keratins/biosynthesis , Keratins/deficiency , Microscopy, Fluorescence , Mutation , Phenotype
12.
J Cell Sci ; 115(Pt 22): 4341-51, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12376565

ABSTRACT

The intermediate filament cytoskeleton is thought to confer physical resilience on tissue cells, on the basis of extrapolations from the phenotype of cell fragility that results from mutations in skin keratins. There is a need for functional cell assays in which the impact of stress on intermediate filaments can be induced and analyzed. Using osmotic shock, we have induced cytoskeleton changes that suggest protective functions for actin and intermediate filament systems. Induction of the resulting stress response has been monitored in keratinocyte cells lines carrying K5 or K14 mutations, which are associated with varying severity of epidermolysis bullosa simplex. Cells with severe mutations were more sensitive to osmotic stress and took longer to recover from it. Their stress-activated response pathways were induced faster, as seen by early activation of JNK, ATF-2 and c-Jun. We demonstrate that the speed of a cell's response to hypotonic stress, by activation of the SAPK/JNK pathway, is correlated with the clinical severity of the mutation carried. The response to hypo-osmotic shock constitutes a discriminating stress assay to distinguish between the effects of different keratin mutations and is a potentially valuable tool in developing therapeutic strategies for keratin-based skin fragility disorders.


Subject(s)
Cytoskeleton/metabolism , Epidermis/enzymology , Epidermolysis Bullosa Simplex/enzymology , Epidermolysis Bullosa Simplex/genetics , Keratinocytes/enzymology , Keratins/deficiency , Stress, Physiological/genetics , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Actin Cytoskeleton/ultrastructure , Activating Transcription Factor 2 , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Cytoskeleton/pathology , Cytoskeleton/ultrastructure , Epidermis/pathology , Epidermis/ultrastructure , Epidermolysis Bullosa Simplex/physiopathology , Humans , Intermediate Filaments/metabolism , Intermediate Filaments/pathology , Intermediate Filaments/ultrastructure , Keratinocytes/pathology , Keratinocytes/ultrastructure , Keratins/genetics , Microscopy, Electron, Scanning , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinases/metabolism , Mutation/genetics , Osmotic Pressure , Proto-Oncogene Proteins c-jun/metabolism , Stress, Physiological/enzymology , Transcription Factors/metabolism
13.
Exp Dermatol ; 11(4): 327-36, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12190941

ABSTRACT

The clinical diagnosis of ichthyosis vulgaris (IV) can be difficult. Abnormalities in the granular layer and the ultrastructure of keratohyalin granules (KHG) suggest that morphology may be helpful. To clarify morphologic findings in IV, 41 clinically affected individuals and 21 unaffected family members or age- and sex-matched controls were studied by light microscopy. In these, the granular layer was totally absent in approximately 50% of affected individuals, while present in all controls. Forty-seven individuals in the light microscopy group were also studied by electron microscopy. Keratohyalin granules were absent in all affected individuals lacking the granular layer by light microscopy. Clinical severity usually correlated with the lack of a granular layer and KHG. Absence of the granular layer was consistent in different anatomic sites and in serial biopsies taken over a 1-3-year period. In a subset of clinically affected, unrelated subjects with moderate to severe involvement, four out of 11 (36%) had similar findings. Keratinocytes cultured from affected individuals with no KHG expressed virtually no detectable profilaggrin protein in vitro. The data suggest that a subset of individuals with moderate to severe IV have a consistently absent granular layer and KHG. Absence of the granular layer and lack of KHG correlated almost perfectly; thus light microscopy offers a convenient means of identifying this subtype of IV. However, both morphologic types of IV were observed within single families. Therefore, the relationship between granular layer abnormalities and IV is complex and requires the study of more affected families. One interpretation of the data is that IV is a multigenic disorder in which one of the genes alters profilaggrin expression. We propose this clinical and histologic phenotype as useful for identifying the gene(s) involved and also for determining whether it represents a modifier or a major locus of the disorder.


Subject(s)
Ichthyosis Vulgaris/metabolism , Ichthyosis Vulgaris/pathology , Keratins/deficiency , Case-Control Studies , Cells, Cultured , Female , Filaggrin Proteins , Gene Expression , Humans , Ichthyosis Vulgaris/classification , Ichthyosis Vulgaris/genetics , Intermediate Filament Proteins/genetics , Keratinocytes/metabolism , Keratinocytes/pathology , Male , Microscopy, Electron , Pedigree , Phenotype , Protein Precursors/genetics
14.
J Cell Sci ; 115(Pt 13): 2639-50, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12077355

ABSTRACT

In the past, keratins have been established as structural proteins. Indeed, mutations in keratin 10 (K10) and other epidermal keratins lead to severe skin fragility syndromes. Here, we present adult K10-/- mice, which reveal a novel connection between the regulation of cell proliferation and K10. Unlike most keratin mutant mice, the epidermis of adult K10-/- mice showed no cytolysis but displayed hyperproliferation of basal keratinocytes and an increased cell size. BrdU labelling revealed a shortened transition time for keratinocytes migrating outwards and DAPI staining of epidermal sheets uncovered an impaired organization of epidermal proliferation units. These remarkable changes were accompanied by the induction of c-Myc, cyclin D1, 14-3-3sigma and of wound healing keratins K6 and K16. The phosphorylation of Rb remained unaltered. In line with the downregulation of K10 in squamous cell carcinomas and its absence in proliferating cells in vivo, our data suggest that the tissue-restricted expression of some members of the keratin gene family not only serves structural functions. Our results imply that the altered composition of the suprabasal cytoskeleton is able to alter the proliferation state of basal cells through the induction of c-Myc. A previous model based on transfection of K10 in immortalized human keratinocytes suggested a direct involvement of K10 in cell cycle control. While those experiments were performed in human cultured keratinocytes, our data establish, that in vivo, K10 acts by an indirect control mechanism in trans.


Subject(s)
Biomarkers, Tumor , Cell Division/genetics , Epidermis/metabolism , Exonucleases/metabolism , Keratins/deficiency , Neoplasm Proteins , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins , Skin Diseases, Genetic/metabolism , 14-3-3 Proteins , Animals , Cell Differentiation/genetics , Cytoskeleton/genetics , Cytoskeleton/metabolism , Cytoskeleton/pathology , Epidermis/pathology , Epidermis/physiopathology , Exonucleases/genetics , Exoribonucleases , Gene Expression Regulation/genetics , Hyperkeratosis, Epidermolytic/genetics , Hyperkeratosis, Epidermolytic/metabolism , Hyperkeratosis, Epidermolytic/physiopathology , Hypertrophy/genetics , Hypertrophy/metabolism , Hypertrophy/physiopathology , Keratin-10 , Keratin-6 , Keratins/biosynthesis , Keratins/genetics , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-myc/genetics , Skin Diseases, Genetic/genetics , Skin Diseases, Genetic/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation/genetics
15.
Cell Microbiol ; 4(2): 73-86, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11896764

ABSTRACT

A highly transmissible strain of Burkholderia cepacia from genomovar III carries the cable pilin gene, expresses the 22 kDa adhesin (cblA +ve/Adh +ve), binds to cytokeratin 13 (CK13) and is invasive. CK13 is expressed abundantly in the airway epithelia of cystic fibrosis (CF) patients. We have now investigated whether binding of cblA +ve/Adh +ve B. cepacia to CK13 potentiates bacterial invasion and epithelial damage using bronchial epithelial cell cultures differentiated into either squamous (CK13-enriched) or mucociliary (CK13-deficient) epithelia. Three different B. cepacia isolates (cblA +ve/Adh +ve, cblA +ve/Adh -ve and cblA -ve/Adh -ve) showed minimal binding to mucociliary cultures, and did not invade or cause cell damage. In contrast, the cblA +ve/Adh +ve isolate, but not others, bound to CK13-expressing cells in squamous cultures, caused cytotoxicity and stimulated IL-8 release within 2 h. By 24 h, this isolate invaded and migrated across the squamous culture, causing moderate to severe epithelial damage. A specific antiadhesin antibody, which blocked the initial binding of the cblA +ve/Adh +ve isolate to CK13, significantly inhibited all the pathologic effects. Transmission electron microscopy of squamous cultures incubated with the cblA +ve/Adh +ve isolate, revealed bacteria on the surface surrounded by filopodia by 2 h, and within the cells in membrane-bound vesicles by 24 h. Bacteria were also observed free in the cytoplasm, surrounded by intermediate filaments containing CK13. These findings suggest that binding of B. cepacia to CK13 is an important initial event and that it promotes bacterial invasion and epithelial damage.


Subject(s)
Adhesins, Bacterial/metabolism , Burkholderia cepacia/metabolism , Membrane Proteins/metabolism , Respiratory Mucosa/metabolism , Adhesins, Bacterial/immunology , Antibodies, Bacterial/pharmacology , Bacterial Adhesion/drug effects , Bronchi , Burkholderia cepacia/isolation & purification , Cell Differentiation , Cells, Cultured , Fimbriae Proteins , Humans , Interleukin-8/analysis , Keratins/deficiency , Keratins/genetics , Keratins/metabolism , Microscopy, Electron , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Time Factors
16.
Hepatology ; 34(6): 1174-83, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11732007

ABSTRACT

Simple epithelial tissues such as liver and pancreas express keratins 8 (K8) and 18 (K18) as their major intermediate filament proteins. K8 and K18 null mice and transgenic mice that express mutant K18 (K18C) manifest several hepatocyte abnormalities and demonstrate that K8/18 are important in maintaining liver tissue and cell integrity, although other potential functions remain uncharacterized. Here, we report an additional abnormal liver phenotype, which is similar in K8 null, K18 null, and K18C mouse models. Liver histologic examination showed large polynuclear areas that lacked cell membranes, desmosomal structures, and filamentous actin. Similar, but less prominent, areas were observed in the pancreas. The parenchyma outside the polynuclear areas displayed irregular sinusoidal structures and markedly enlarged nuclei. Most K8 null hepatocytes were positive for the proliferating cell nuclear antigen (PCNA) with a doubled DNA content in comparison with the predominantly PCNA-negative wild-type hepatocytes. The distribution of the 14-3-3zeta protein was also altered in K8 null mice. Taken together, our results indicate that absence of keratin filaments causes disturbances in cell-cycle regulation, driving cells into the S-G2 phase and causing aberrant cytokinesis. These effects could stem from disturbed functions of K8/18-dependent cell-cycle regulators, such as the signaling integrator, 14-3-3.


Subject(s)
Keratins/physiology , Liver/pathology , Actins/deficiency , Animals , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Desmosomes/pathology , Keratins/deficiency , Keratins/genetics , Mice , Mice, Knockout/genetics , Mice, Transgenic/genetics , Mutation/physiology , Pancreas/pathology
17.
J Invest Dermatol ; 115(4): 708-13, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10998148

ABSTRACT

Point mutations in the suprabasal cytokeratins 1 (K1) or 10 (K10) in humans have been shown to be the cause of the congenital ichthyosis epidermolytic hyperkeratosis. Recently, a K10 deficient mouse model was established serving as a model for epidermolytic hyperkeratosis. Homozygotes suffered from severe skin fragility and died shortly after birth. Heterozygotes developed hyperkeratosis with age. To see whether phenotypic abnormalities in the mouse model were associated with changes in skin barrier function and skin water content we studied basal transepidermal water loss and capacity for barrier repair after experimental barrier disruption as well as stratum corneum hydration. Also, we determined the activities of acid and neutral sphingomyelinase key enzymes of the tumor necrosis factor and interleukin-1 signal transduction pathways generating the ceramides most important for epidermal permeability barrier homeostasis. Neonatal homozygotes showed an 8-fold increase in basal transepidermal water loss compared with wild type controls. Adult heterozygotes exhibited delayed barrier repair after experimental barrier disruption. Stratum corneum hydration was reduced in homozygous and heterozygous mice. Acid sphingomyelinase activity, which is localized in the epidermal lamellar bodies and generates ceramides for extracellular lipid lamellae in the stratum corneum permeability barrier, was reduced in homozygous as well as heterozygous animals. Neutral sphingomyelinase activity, which has a different location and generates ceramides involved in cell signaling, was increased. The reduction in acid sphingomyelinase activity may explain the recently described decreased ratio of ceramides to total lipids in K10 deficient mice. In summary, our results demonstrate the crucial role of the keratin filament for permeability barrier function and stratum corneum hydration.


Subject(s)
Body Water/metabolism , Cell Membrane Permeability/physiology , Keratins/deficiency , Skin/cytology , Sphingomyelin Phosphodiesterase/metabolism , Animals , Animals, Newborn , Disease Models, Animal , Heterozygote , Homeostasis , Homozygote , Humans , Hyperkeratosis, Epidermolytic , Mice , Skin/enzymology , Skin/metabolism
18.
Br J Dermatol ; 143(3): 621-7, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10971341

ABSTRACT

Epidermolysis bullosa simplex (EBS) is a blistering skin disease caused in most cases by mis-sense mutations in genes encoding the basal epidermal keratin (K) 5 and K14. The inheritance is usually autosomal dominant and the mutant keratin proteins appear to exert a dominant negative effect on the keratin intermediate filament cytoskeleton in basal keratinocytes. We report a child with a homozygous K14 mutation resulting in the complete absence of K14 protein in the epidermis; remarkably, he only had mild to moderate disease. Electron microscopy of a skin biopsy showed a marked reduction in numbers of keratin intermediate filaments in the basal keratinocytes. Immunofluorescence microscopy using monoclonal antibody LL001 against K14 showed no staining, suggesting a functional knockout of K14. Sequence analysis of genomic DNA revealed a homozygous mutation in codon 31 of K14 that resulted in a premature stop codon further downstream in exon 1. The child's mother, who is unaffected by the disease, is heterozygous for the mutation. The consanguineous father was unaffected and unavailable for testing. The resulting mRNA is predicted to encode a protein of 116 amino acids, of which the first 30 are identical to the normal K14 sequence, and the remaining 86 residues are mis-sense sequence. Four previously reported cases of autosomal recessive EBS with functional knockout of K14 were severely affected by blistering, in contrast to our patient in whom the predicted protein has only the first 30 amino acids of K14 and is therefore the closest to a true knockout of K14 protein yet identified.


Subject(s)
Epidermolysis Bullosa Simplex/genetics , Genes, Recessive , Keratins/genetics , Mutation, Missense/genetics , Consanguinity , Epidermolysis Bullosa Simplex/metabolism , Epidermolysis Bullosa Simplex/pathology , Homozygote , Humans , Infant , Keratin-14 , Keratins/deficiency , Male , Microscopy, Electron , Microscopy, Fluorescence , Pedigree , Polymerase Chain Reaction , Sequence Analysis, DNA
19.
J Cell Biol ; 150(4): 921-8, 2000 Aug 21.
Article in English | MEDLINE | ID: mdl-10953016

ABSTRACT

Mammalian genomes feature multiple genes encoding highly related keratin 6 (K6) isoforms. These type II keratins show a complex regulation with constitutive and inducible components in several stratified epithelia, including the oral mucosa and skin. Two functional genes, K6alpha and K6beta, exist in a head-to-tail tandem array in mouse genomes. We inactivated these two genes simultaneously via targeting and homologous recombination. K6 null mice are viable and initially indistinguishable from their littermates. Starting at two to three days after birth, they show a growth delay associated with reduced milk intake and the presence of white plaques in the posterior region of dorsal tongue and upper palate. These regions are subjected to greater mechanical stress during suckling. Morphological analyses implicate the filiform papillae as being particularly sensitive to trauma in K6alpha/K6beta null mice, and establish the complete absence of keratin filaments in their anterior compartment. All null mice die about a week after birth. These studies demonstrate an essential structural role for K6 isoforms in the oral mucosa, and implicate filiform papillae as being the major stress bearing structures in dorsal tongue epithelium.


Subject(s)
Keratins/physiology , Mouth Mucosa/ultrastructure , Animals , Crosses, Genetic , Female , Keratins/deficiency , Keratins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , Mouth Mucosa/pathology , Mouth Mucosa/physiology , Phosphoglycerate Kinase/genetics , Phosphoglycerate Kinase/metabolism , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/physiology , Thymidine Kinase/genetics , Thymidine Kinase/metabolism , Tongue/abnormalities
20.
J Cell Biol ; 149(1): 17-22, 2000 Apr 03.
Article in English | MEDLINE | ID: mdl-10747083

ABSTRACT

Tumor necrosis factor (TNF) is a cytokine produced by macrophages and T lymphocytes that acts through two distinct receptors, TNFR1 (60 kD, CD120a) and TNFR2 (80 kD, CD120b), to affect cellular proliferation, differentiation, survival, and cell death. In addition to its proinflammatory actions in mucosal tissue, TNF is important for liver regeneration. Keratin 8 (K8) and keratin 18 (K18) form intermediate filaments characteristic of liver and other single cell layered, internal epithelia and their derivative cancers. K8-deficient (K8(-)) mice, which escape embryonic lethality, develop inflammatory colorectal hyperplasia, mild liver abnormalities, and tolerate hepatectomy poorly. We show that normal and malignant epithelial cells deficient in K8 and K18 are approximately 100 times more sensitive to TNF-induced death. K8 and K18 both bind the cytoplasmic domain of TNFR2 and moderate TNF-induced, Jun NH(2)-terminal kinase (JNK) intracellular signaling and NFkappaB activation. Furthermore, K8(-) and K18(-) mice are much more sensitive to TNF dependent, apoptotic liver damage induced by the injection of concanavalin A. This moderation of the effects of TNF may be the fundamental function of K8 and K18 common to liver regeneration, inflammatory bowel disease, hepatotoxin sensitivity, and the diagnostic, persistent expression of these keratins in many carcinomas.


Subject(s)
Apoptosis/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Keratins/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Line , Chemical and Drug Induced Liver Injury/pathology , Concanavalin A/toxicity , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , JNK Mitogen-Activated Protein Kinases , Keratins/deficiency , Keratins/genetics , Liver/cytology , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Binding , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/drug effects , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...