Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Geobiology ; 16(3): 237-251, 2018 05.
Article in English | MEDLINE | ID: mdl-29569335

ABSTRACT

Fossil derivatives of isorenieratene, an accessory pigment in brown-colored green sulfur bacteria, are often used as tracers for photic zone anoxia through Earth's history, but their diagenetic behavior is still incompletely understood. Here, we assess the preservation of isorenieratene derivatives in organic-rich shales (1.5-8.4 wt.% TOC) from two Lower Jurassic anoxic systems (Bächental oil shale, Tyrol, Austria; Posidonia Shale, Baden-Württemberg, Germany). Bitumens and kerogens were investigated using catalytic hydropyrolysis (HyPy), closed-system hydrous pyrolysis (in gold capsules), gas chromatography-mass spectrometry (GC-MS) and gas chromatography combustion isotope ratio-mass spectrometry (GC-C-IRMS). Petrography and biomarkers indicate a syngenetic relationship between bitumens and kerogens. All bitumens contain abundant isorenieratane, diverse complex aromatized isorenieratene derivatives, and a pseudohomologous series of 2,3,6-trimethyl aryl isoprenoids. In contrast, HyPy and mild closed-system hydrous pyrolysis of the kerogens yielded only minor amounts of these compounds. Given the overall low maturity of the organic matter (below oil window), it appears that isorenieratene and its abundant derivatives from the bitumen had not been incorporated into the kerogens. Accordingly, sulfur cross-linking, the key mechanism for sequestration of functionalized lipids into kerogens in anoxic systems, was not effective in the Jurassic environments studied. We explain this by (i) early cyclization/aromatization and (ii) hydrogenation reactions that have prevented effective sulfurization. In addition, (iii) sulfide was locally removed via anoxygenic photosynthesis and efficiently trapped by the reaction with sedimentary iron, as further indicated by elevated iron contents (4.0-8.7 wt.%) and the presence of abundant pyrite aggregates in the rock matrix. Although the combined processes have hampered the kerogen incorporation of isorenieratene and its derivatives, they may have promoted the long-term preservation of these biomarkers in the bitumen fraction via early defunctionalization. This particular taphonomy of aromatic carotenoids has to be considered in studies of anoxic iron-rich environments (e.g., the Proterozoic ocean).


Subject(s)
Carotenoids/metabolism , Chlorobi/chemistry , Fossils , Geologic Sediments/chemistry , Iron/metabolism , Phenols/metabolism , Pigments, Biological/metabolism , Austria , Germany , Hypoxia , Spectrum Analysis
2.
Oncogene ; 35(24): 3114-24, 2016 06 16.
Article in English | MEDLINE | ID: mdl-26522723

ABSTRACT

Epidermal homeostasis depends on the coordinated control of keratinocyte cell cycle. Differentiation and the alteration of this balance can result in neoplastic development. Here we report on a novel DLX3-dependent network that constrains epidermal hyperplasia and squamous tumorigenesis. By integrating genetic and transcriptomic approaches, we demonstrate that DLX3 operates through a p53-regulated network. DLX3 and p53 physically interact on the p21 promoter to enhance p21 expression. Elevating DLX3 in keratinocytes produces a G1-S blockade associated with p53 signature transcriptional profiles. In contrast, DLX3 loss promotes a mitogenic phenotype associated with constitutive activation of ERK. DLX3 expression is lost in human skin cancers and is extinguished during progression of experimentally induced mouse squamous cell carcinoma (SCC). Reinstatement of DLX3 function is sufficient to attenuate the migration of SCC cells, leading to decreased wound closure. Our data establish the DLX3-p53 interplay as a major regulatory axis in epidermal differentiation and suggest that DLX3 is a modulator of skin carcinogenesis.


Subject(s)
Carcinoma, Squamous Cell/pathology , Homeodomain Proteins/metabolism , Skin Neoplasms/pathology , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Carcinogenesis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Cycle Checkpoints , Cell Differentiation/physiology , Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Progression , Female , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Male , Mice , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transfection , Tumor Suppressor Protein p53/genetics
3.
Cell Death Dis ; 6: e1647, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25695600

ABSTRACT

Epidermal keratinocyte differentiation on the body surface is a carefully choreographed process that leads to assembly of a barrier that is essential for life. Perturbation of keratinocyte differentiation leads to disease. Activator protein 1 (AP1) transcription factors are key controllers of this process. We have shown that inhibiting AP1 transcription factor activity in the suprabasal murine epidermis, by expression of dominant-negative c-jun (TAM67), produces a phenotype type that resembles human keratoderma. However, little is understood regarding the structural and molecular changes that drive this phenotype. In the present study we show that TAM67-positive epidermis displays altered cornified envelope, filaggrin-type keratohyalin granule, keratin filament, desmosome formation and lamellar body secretion leading to reduced barrier integrity. To understand the molecular changes underlying this process, we performed proteomic and RNA array analysis. Proteomic study of the corneocyte cross-linked proteome reveals a reduction in incorporation of cutaneous keratins, filaggrin, filaggrin2, late cornified envelope precursor proteins, hair keratins and hair keratin-associated proteins. This is coupled with increased incorporation of desmosome linker, small proline-rich, S100, transglutaminase and inflammation-associated proteins. Incorporation of most cutaneous keratins (Krt1, Krt5 and Krt10) is reduced, but incorporation of hyperproliferation-associated epidermal keratins (Krt6a, Krt6b and Krt16) is increased. RNA array analysis reveals reduced expression of mRNA encoding differentiation-associated cutaneous keratins, hair keratins and associated proteins, late cornified envelope precursors and filaggrin-related proteins; and increased expression of mRNA encoding small proline-rich proteins, protease inhibitors (serpins), S100 proteins, defensins and hyperproliferation-associated keratins. These findings suggest that AP1 factor inactivation in the suprabasal epidermal layers reduces expression of AP1 factor-responsive genes expressed in late differentiation and is associated with a compensatory increase in expression of early differentiation genes.


Subject(s)
Activating Transcription Factor 1/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Activating Transcription Factor 1/genetics , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Epidermal Cells , Epidermis/ultrastructure , Female , Filaggrin Proteins , Keratinocytes/ultrastructure , Keratins/metabolism , Mice , Microscopy, Electron , Peptide Fragments/genetics , Peptide Fragments/metabolism , Proteomics , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism
4.
Geobiology ; 12(5): 406-23, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25039581

ABSTRACT

Symbiont-bearing and non-symbiotic marine bivalves were used as model organisms to establish biosignatures for the detection of distinctive symbioses in ancient bivalves. For this purpose, the isotopic composition of lipids (δ13C) and bulk organic shell matrix (δ13C, δ34S, δ15N) from shells of several thiotrophic, phototrophic, or non-symbiotic bivalves were compared (phototrophic: Fragum fragum, Fragum unedo, Tridacna maxima; thiotrophic: Codakia tigerina, Fimbria fimbriata, Anodontia sp.; non-symbiotic: Tapes dorsatus, Vasticardium vertebratum, Scutarcopagia sp.). ∆13C values of bulk organic shell matrices, most likely representing mainly original shell protein/chitin biomass, were depleted in thio- and phototrophic bivalves compared to non-symbiotic bivalves. As the bulk organic shell matrix also showed a major depletion of δ15N (down to -2.2 ‰) for thiotrophic bivalves, combined δ13C and δ15N values are useful to differentiate between thio-, phototrophic, and non-symbiotic lifestyles. However, the use of these isotopic signatures for the study of ancient bivalves is limited by the preservation of the bulk organic shell matrix in fossils. Substantial alteration was clearly shown by detailed microscopic analyses of fossil (late Pleistocene) T. maxima and Trachycardium lacunosum shell, demonstrating a severe loss of quantity and quality of bulk organic shell matrix with time. Likewise, the composition and δ13C-values of lipids from empty shells indicated that a large part of these compounds derived from prokaryotic decomposers. The use of lipids from ancient shells for the reconstruction of the bivalve's life style therefore appears to be restricted.


Subject(s)
Bivalvia/metabolism , Fossils , Light , Sulfur/metabolism , Animal Shells/chemistry , Animals , Carbon Isotopes/analysis , Lipids/analysis , Nitrogen Isotopes/analysis , Species Specificity , Sulfur Isotopes/analysis , Symbiosis
5.
FASEB J ; 15(13): 2533-5, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11641260

ABSTRACT

The epidermis, our first line of defense from ultraviolet (UV) light, bears the majority of photodamage, which results in skin thinning, wrinkling, keratosis, and malignancy. Hypothesizing that skin has specific mechanisms to protect itself and the organism from UV damage, we used DNA arrays to follow UV-caused gene expression changes in epidermal keratinocytes. Of the 6,800 genes examined, UV regulates the expression of at least 198. Three waves of changes in gene expression can be distinguished, 0.5-2, 4-8, and 16-24 h after illumination. The first contains transcription factors, signal transducing, and cytoskeletal proteins that change cell phenotype from a normal, fast-growing cell to an activated, paused cell. The second contains secreted growth factors, cytokines, and chemokines; keratinocytes, having changed their own physiology, alert the surrounding tissues to the UV damage. The third wave contains components of the cornified envelope, as keratinocytes enhance the epidermal protective covering and, simultaneously, terminally differentiate and die, removing a carcinogenic threat. UV also induces the expression of mitochondrial proteins that provide additional energy, and the enzymes that synthesize raw materials for DNA repair. Using a novel skin organ culture model, we demonstrated that the UV-induced changes detected in keratinocyte cultures also occur in human epidermis in vivo.


Subject(s)
Gene Expression Profiling , Oligonucleotide Array Sequence Analysis/methods , Ultraviolet Rays , Chemokines/genetics , Cytokines/genetics , DNA Repair , Epidermal Cells , Epidermis/metabolism , Epidermis/radiation effects , Gene Expression Regulation/radiation effects , Growth Substances/genetics , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Keratinocytes/radiation effects , Mitochondria/metabolism , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/radiation effects , Signal Transduction/genetics , Transcription Factor AP-1/genetics , Transcription, Genetic/radiation effects , Tumor Suppressor Protein p53/genetics
6.
J Invest Dermatol ; 116(5): 633-40, 2001 May.
Article in English | MEDLINE | ID: mdl-11348449

ABSTRACT

In wound healing and many pathologic conditions, keratinocytes become activated: they turn into migratory, hyperproliferative cells that produce and secrete extracellular matrix components and signaling polypeptides. At the same time, their cytoskeleton is also altered by the production of specific keratin proteins. These changes are orchestrated by growth factors, chemokines, and cytokines produced by keratinocytes and other cutaneous cell types. The responding intracellular signaling pathways activate transcription factors that regulate expression of keratin genes. Analysis of these processes led us to propose the existence of a keratinocyte activation cycle, in which the cells first become activated by the release of IL-1. Subsequently, they maintain the activated state by autocrine production of proinflammatory and proliferative signals. Keratins K6 and K16 are markers of the active state. Signals from the lymphocytes, in the form of Interferon-gamma, induce the expression of K17 and make keratinocytes contractile. This enables the keratinocytes to shrink the provisional fibronectin-rich basement membrane. Signals from the fibroblasts, in the form of TGF-beta, induce the expression of K5 and K14, revert the keratinocytes to the healthy basal phenotype, and thus complete the activation cycle.


Subject(s)
Keratinocytes/physiology , Keratins/physiology , Humans , Interferon-gamma/physiology , Interleukin-1/physiology , Phenotype , Transforming Growth Factor beta/physiology
7.
J Invest Dermatol ; 116(2): 330-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11180011

ABSTRACT

Keratinocytes respond to injury by releasing the proinflammatory cytokine interleukin-1, which serves as the initial "alarm signal" to surrounding cells. Among the consequences of interleukin-1 release is the production of additional cytokines and their receptors by keratinocytes and other cells in the skin. Here we describe an additional effect of interleukin-1 on keratinocytes, namely the alteration in the keratinocyte cytoskeleton in the form of the induction of keratin 6 expression. Keratin 6 is a marker of hyperproliferative, activated keratinocytes, found in wound healing, psoriasis, and other inflammatory disorders. Skin biopsies in organ culture treated with interleukin-1 express keratin 6 in all suprabasal layers of the epidermis, throughout the tissue. In cultured epidermal keratinocytes, the induction of keratin 6 is time and concentration dependent. Importantly, only confluent keratinocytes respond to interleukin-1, subconfluent cultures do not. In the cells starved of growth factors, epidermal growth factor or tumor necrosis factor-alpha, if added simultaneously with interleukin-1, they synergistically augment the effects of interleukin-1. Using DNA-mediated cell transfection, we analyzed the molecular mechanisms regulating the keratin 6 induction by interleukin-1, and found that the induction occurs at the transcriptional level. We used a series of deletions and point mutations to identify the interleukin-1 responsive DNA element in the keratin 6 promoter, and determined that it contains a complex of C/EBP binding sites. The transcription factor C/EBPbeta binds this element in vitro, and the binding is augmented by pretreatment of the cells with interleukin-1. The interleukin-1 responsive element is clearly distinct from the epidermal growth factor responsive one, which means that the proinflammatory and proliferative signals independently regulate the expression of keratin 6. Thus, interleukin-1 initiates keratinocyte activation not only by triggering additional signaling events, but also by inducing directly the synthesis of keratin 6 in epidermal keratinocytes, and thus changing the composition of their cytoskeleton.


Subject(s)
Interleukin-1/pharmacology , Keratinocytes/metabolism , Keratins/genetics , Base Sequence , Chromosome Mapping , Culture Media/pharmacology , Gene Expression Regulation/drug effects , Genes , HeLa Cells , Humans , Promoter Regions, Genetic , Skin/metabolism , Transcription, Genetic/drug effects
8.
J Biol Chem ; 275(41): 32077-88, 2000 Oct 13.
Article in English | MEDLINE | ID: mdl-10887174

ABSTRACT

Epidermal keratinocytes respond to injury by becoming activated, i.e. hyperproliferative, migratory, and proinflammatory. These processes are regulated by growth factors and cytokines. One of the markers of activated keratinocytes is keratin K6. We used a novel organ culture system to show that tumor necrosis factor alpha (TNFalpha) induces the expression of K6 protein and mRNA in human skin. Multiple isoforms of K6 are encoded by distinct genes and have distinct patterns of expression. By having shown previously that proliferative signals, such as epidermal growth factor (EGF), induce expression of the cytoskeletal protein keratin K6b, we here demonstrate that the same isoform, K6b, is also induced by TNFalpha, a proinflammatory cytokine. Specifically, TNFalpha induces the transcription of the K6b gene promoter. By using co-transfection, specific inhibitors, and antisense oligonucleotides, we have identified NFkappaB and C/EBPbeta as the transcription factors that convey the TNFalpha signal. Both transcription factors are necessary for the induction of K6b by TNFalpha and act as a complex, although only C/EBPbeta binds the K6b promoter DNA. By using transfection, site-directed mutagenesis, and footprinting, we have mapped the site that responds to TNFalpha, NFkappaB, and C/EBPbeta. This site is separate from the one responsive to EGF and AP1. Our results show that the proinflammatory (TNFalpha) and the proliferative (EGF) signals in epidermis separately and independently regulate the expression of the same K6b keratin isoform. Thus, the cytoskeletal responses in epidermal cells can be precisely tuned by separate proliferative and inflammatory signals to fit the nature of the injuries that caused them.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Keratinocytes/metabolism , Keratins/biosynthesis , NF-kappa B/metabolism , Transcriptional Activation/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Base Sequence , Binding Sites , Cell Division , DNA Footprinting , Epidermal Cells , Epidermal Growth Factor/pharmacology , Epidermis/drug effects , Epidermis/metabolism , Epidermis/pathology , Fluorescent Antibody Technique , HeLa Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Keratinocytes/drug effects , Keratins/genetics , Molecular Sequence Data , Mutation , Oligonucleotides, Antisense/pharmacology , Promoter Regions, Genetic/genetics , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Response Elements/genetics , Transcription Factor AP-1/metabolism , Transfection
9.
Mol Cell Biol ; 20(12): 4328-39, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10825196

ABSTRACT

Glucocorticoids (GCs), important regulators of epidermal growth, differentiation, and homeostasis, are used extensively in the treatment of skin diseases. Using keratin gene expression as a paradigm of epidermal physiology and pathology, we have developed a model system to study the molecular mechanism of GCs action in skin. Here we describe a novel mechanism of suppression of transcription by the glucocorticoid receptor (GR) that represents an example of customizing a device for transcriptional regulation to target a specific group of genes within the target tissue, in our case, epidermis. We have shown that GCs repress the expression of the basal-cell-specific keratins K5 and K14 and disease-associated keratins K6, K16, and K17 but not the differentiation-specific keratins K3 and K10 or the simple epithelium-specific keratins K8, K18, and K19. We have identified the negative recognition elements (nGREs) in all five regulated keratin gene promoters. Detailed footprinting revealed that the function of nGREs is to instruct the GR to bind as four monomers. Furthermore, using cotransfection and antisense technology we have found that, unlike SRC-1 and GRIP-1, which are not involved in the GR complex that suppresses keratin genes, histone acetyltransferase and CBP are. In addition, we have found that GR, independently from GREs, blocks the induction of keratin gene expression by AP1. We conclude that GR suppresses keratin gene expression through two independent mechanisms: directly, through interactions of keratin nGREs with four GR monomers, as well as indirectly, by blocking the AP1 induction of keratin gene expression.


Subject(s)
Glucocorticoids/pharmacology , Keratinocytes/metabolism , Keratins/genetics , Receptors, Glucocorticoid/metabolism , Skin/metabolism , Gene Expression Regulation/drug effects , Glucocorticoids/genetics , Glucocorticoids/metabolism , HeLa Cells , Humans , Keratins/metabolism , Promoter Regions, Genetic/genetics , Receptors, Glucocorticoid/genetics , Skin Diseases/drug therapy , Skin Diseases/genetics , Skin Diseases/metabolism
10.
J Dermatol Sci ; 17(3): 167-81, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9697045

ABSTRACT

In the area of biology, many laboratories around the world are dissecting and characterizing signal transduction mechanisms and transcription factors responsive to various growth factors and cytokines, in various cell types. However, because of the differences in systems used, it is not clear whether these systems coexist, whether they interact meaningfully, and what their relative roles are. Epidermal keratinocytes are the perfect cell type in which to integrate this knowledge, because in these cells these mechanisms are known to be relevant. Keratinocytes both produce and respond to growth factors and cytokines, especially in pathological conditions and during wound healing, when the physiology of keratinocytes is altered in a way specified by the presence of a subset growth factors and cytokines. In fact, growth factors and cytokines cause the major changes in gene expression and keratinocyte behavior in various cutaneous diseases. In some cases, such as in wound healing, these responses are highly beneficial; in others, such as in psoriasis, they are pathological. It is not clear at present which are operating in which conditions, which are important for the healing process and which are harmful. Growth factors and cytokines affect keratinocytes sometimes simultaneously, at other times individually. In this manuscript we describe the signal transduction pathways responsible for the effects of interferons, the EGF/TGF alpha family and the TNF alpha/IL-1 family of signaling molecules. We also describe the important transcription factors known to be functional in epidermis, with particular emphasis on those factors that are activated by growth factors and cytokines. Finally, we describe what is known about transcriptional regulation of keratin genes, especially those specifically expressed in pathological processes in the epidermis. We expect that the enhanced understanding of the pathways regulating gene expression in keratinocytes will identify the pharmacological targets, the signal transducing proteins and the corresponding transcription factors, used by growth factors and cytokines. This research will led to development of compounds precisely aimed at those targets, allowing us to isolate and inhibit the harmful side effects of growth factors and cytokines. Such compounds should lead to highly specific and therefore more effective treatments of the cutaneous disorders in which these pathways play significant roles.


Subject(s)
Gene Expression Regulation , Keratinocytes/physiology , Signal Transduction/physiology , Skin Physiological Phenomena , Transcription Factors/physiology , Humans , Keratins/genetics , Keratins/metabolism
11.
J Invest Dermatol ; 109(4): 566-72, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9326392

ABSTRACT

Retinoic acid and thyroid hormone are important regulators of epidermal growth, differentiation, and homeostasis. Retinoic acid is extensively used in the treatment of many epidermal disorders ranging from wrinkles to skin cancers. Retinoic acid and thyroid hormone directly control the transcription of differentiation-specific genes including keratins. Their effect is mediated through nuclear receptors RAR and T3R. We have previously identified the response element in the K14 gene, K14RARE/TRE, to which these receptors bind, and found that it consists of a cluster of five half-sites with variable spacing and orientation. To determine whether this specific structure is found in other keratin genes, we have mapped and analyzed the RARE/TRE elements in three additional epidermal keratin genes: K5, K6, and K17. We used three different approaches to identify these elements: co-transfection of promoter deletion constructs, gel-shift assays, and site-specific mutagenesis. We localized the RARE/TRE elements relatively close to the TATA box in all three promoters. All three RARE/TRE elements have a similar structural organization: they consist of clusters of 3-6 half-sites with variable spacing and orientation. This means that the clustered structure of the RARE/TREs is a common characteristic for keratin genes. RARE and TRE in the K5 promoter are adjacent to each other whereas in the K17 promoter they overlap. All three keratin REs bind specifically both RAR and T3R in gel-shift assays. Interestingly, addition of ligand to the receptor changes the binding pattern ofthe T3R from homodimer to monomer, reflecting the change in regulation from induction to inhibition.


Subject(s)
Genes, Regulator , Keratins/genetics , Multigene Family , Promoter Regions, Genetic/genetics , Receptors, Retinoic Acid/genetics , Receptors, Thyroid Hormone/genetics , Base Sequence , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/metabolism
13.
Gene Expr ; 6(6): 361-70, 1997.
Article in English | MEDLINE | ID: mdl-9495317

ABSTRACT

The expression of keratins K5 and K14 is restricted to the basal layers of the healthy epidermis, whereas the expression of K6 and K17 is induced in response to proliferative and inflammatory signals, respectively. The control of keratin expression occurs primarily at the transcriptional level. We studied the effects of transcription factors of the AP-1 and NF-kappaB families on the expression of those four keratin genes. We chose AP-1 and NF-kappaB proteins because they are activated by many extracellular signals, including those in hyperproliferative and inflammatory processes. DNA constructs expressing the transcription factors were, in various combinations, cotransfected with constructs containing keratin gene promoters and the CAT reporter gene into HeLa cells or keratinocytes. We found that the K5 and K14 promoters, which are coexpressed in vivo, are regulated in parallel by the cotransfected genes. Both were activated by the c-Fos and c-Jun components of AP-1, but not by Fra1. On the other hand, the NF-kappaB proteins, especially p65, suppressed these two promoters. The K17 promoter was specifically activated by c-Jun, whereas the other transcription factors tested had no significant effect. In contrast, the K6 promoter was very strongly activated by all AP-1 proteins, especially by the c-Fos + c-Jun and Fra1 + c-Jun combinations. It was also strongly activated by the p65 NF-kappaB protein. AP-1 and NF-kappaB acted synergistically in activating the K6 promoter, although the AP-1 and the NF-kappaB responsive sites could be separated physically. These results suggest that the interplay of AP-1 and NF-kappaB proteins regulates epidermal gene expression and that the activation of these transcription factors by extracellular signaling molecules brings about the differential expression of keratin genes in epidermal differentiation, cutaneous diseases, and wound healing.


Subject(s)
Gene Expression Regulation/genetics , Keratins/genetics , NF-kappa B/physiology , Transcription Factor AP-1/physiology , Transcription, Genetic/genetics , Base Sequence , Cells, Cultured , Epidermal Cells , Genes/genetics , HeLa Cells , Humans , Keratinocytes , Molecular Sequence Data , NF-kappa B/genetics , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/physiology , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/physiology , Sequence Deletion , Transcription Factor RelA , Transfection
14.
J Invest Dermatol ; 107(4): 569-75, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8823363

ABSTRACT

Keratin K17, the myoepithelial keratin, is expressed in psoriasis but is not present in healthy skin. Psoriasis is associated with production of gamma interferon (IFN gamma), which induces the expression of keratin K17 by activating transcription factor STAT1. Our hypothesis states that the induction of K17 is specific for the inflammatory reactions associated with high levels of IFN gamma and activation of STAT1. One of the corollaries of the hypothesis is that the STAT1-activating cytokines should induce the expression of keratin K17, whereas those cytokines that work through other mechanisms should not. Furthermore, because the STAT activation pathway is dependent upon protein phosphorylation events, phosphorylation inhibitors should attenuate the induction of keratin K17, whereas protein phosphatase inhibitors should augment it. To test this hypothesis, we analyzed lesional samples of inflammatory diseases using immunofluorescence, transfected keratinocytes with K17 gene promoter DNAs in the presence of various cytokines, and followed nuclear translocation of STAT1 in keratinocytes using specific antibodies. Confirming the hypothesis, we found that K17 is induced in psoriasis and dermatitis caused by delayed type hypersensitivity, which are associated with high levels of IFN gamma, but not in samples of atopic dermatitis, which is not. Two cytokines, interleukin-6 and leukemia inhibitory factor, which can induce phosphorylation of STAT1, can also induce K17 expression, whereas interleukin-3, interleukin-4, interleukin-10, and granulocyte macrophage colony stimulating factor have no effect on K17 expression. As expected, staurosporine and genistein inhibited, whereas okadaic acid augmented, the induction of K17 by IFN gamma. Our data indicate that in inflammatory skin diseases, lymphocytes, through the cytokines they produce, differently regulate not only each other, but also keratin gene expression in epidermis one of their target tissues.


Subject(s)
Dermatitis/metabolism , Epidermis/metabolism , Keratins/metabolism , Cytokines/pharmacology , DNA-Binding Proteins/physiology , Dermatitis, Atopic/metabolism , Enzyme Inhibitors/pharmacology , Growth Inhibitors/pharmacology , Humans , Interferon-gamma/pharmacology , Interleukin-10/pharmacology , Interleukin-4/pharmacology , Interleukin-6/pharmacology , Leukemia Inhibitory Factor , Lymphokines/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Promoter Regions, Genetic/drug effects , Protein Kinase Inhibitors , Psoriasis/metabolism , STAT1 Transcription Factor , Trans-Activators/physiology , Transcription, Genetic/drug effects
15.
Genomics ; 34(1): 134-8, 1996 May 15.
Article in English | MEDLINE | ID: mdl-8661035

ABSTRACT

Mapping studies of functional keratin genes in the human genome have localized most of the acidic keratin genes to chromosome 17q12-q21 and the basic keratin genes to chromosome 12q11-q13. Within the acidic keratin locus two clusters were identified, one containing the genes for K15 and K19, the other the genes for K14, K16, and K17. The relative positions and the distance between the two clusters have not been determined previously. In this paper we describe our analysis of P1 clones containing multiple acidic keratin genes, which were studied using restriction analysis and Southern blot hybridization with PCR-amplified probes specific for functional human keratin genes 15, 17, and 19. Our results show that the two clusters are very closely linked to each other, within a 55-kb region in the human genome. The genes are organized 5' to 3' in the following order: 5'-K19-K15-K17-K16-K14. Between K15 and K17 at least one additional, unidentified keratin gene is present.


Subject(s)
Genetic Linkage , Keratins/genetics , Multigene Family , Base Sequence , Blotting, Southern , Chromosomes, Human, Pair 17 , Humans , In Situ Hybridization , Molecular Sequence Data , Restriction Mapping
16.
Exp Cell Res ; 224(1): 96-102, 1996 Apr 10.
Article in English | MEDLINE | ID: mdl-8612697

ABSTRACT

Epidermal keratinocytes are subject to a large variety of signals that modulate their differentiation in health and their activation in disease. Hormones and vitamins, which act via nuclear receptors, affect the differentiation process, whereas growth factors and cytokines, which act via cell surface receptors, affect keratinocyte activation and related events. Using expression of keratin genes as markers for keratinocyte phenotype, we examined the interaction between the nuclear receptor and cell surface receptor pathways. We expected to find dominance of one of the pathways. Surprisingly, we found that the two pathways are codominant. Specifically, while EGF induces expression of K6 and K16 keratin genes, retinoic acid suppresses their expression, and when both mediators are present simultaneously, the level of expression is intermediate, a product of both signals. Similar codominant effects were found on other keratin genes using interferon gamma, TGF beta, and thyroid hormone signaling molecules. These codominant effects are specific only for genes that are regulated by both pathways. Our results suggest that a judicious combination of hormones, vitamins, growth factors, and cytokines may be used to target specific expression of appropriate genes in the treatment of human epidermal diseases.


Subject(s)
Gene Expression Regulation , Keratins/biosynthesis , Receptors, Cell Surface/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Epidermal Growth Factor/pharmacology , HeLa Cells , Humans , Interferon-gamma/pharmacology , Keratinocytes , Keratins/genetics , Promoter Regions, Genetic , Recombinant Proteins/biosynthesis , Transforming Growth Factor beta/pharmacology , Tretinoin/pharmacology
19.
DNA Cell Biol ; 15(1): 65-74, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8561898

ABSTRACT

Keratin K17, while not present in healthy skin, is expressed under various pathological conditions, including psoriasis and cutaneous allergic reactions. The regulatory circuits involved in transcription of the human keratin K17 gene are poorly understood. To begin an analysis of the molecular mechanisms that regulate K17 gene transcription, we have studied the interactions between the nuclear proteins and the promoter region of the human K17 gene. That promoter region comprised 450 bp upstream from the translation initiation site. For these studies, we used electrophoretic mobility-shift assays, computer analysis, site-directed mutagenesis, and DNA-mediated cell transfection. In addition to the previously characterized interferon-gamma-responsive elements, we identified eight protein binding sites in the promoter. Five of them bind the known transcription factors NF1, AP2, and Sp1 and three others bind still unidentified proteins. Using site-directed mutagenesis, we have demonstrated the importance of the protein binding sites for the promoter function involved in both constitutive and interferon-induced expression of the K17 keratin gene.


Subject(s)
Keratins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Transcriptional Activation , Antineoplastic Agents/pharmacology , Base Sequence , Binding Sites/genetics , HeLa Cells , Humans , Interferon-gamma/pharmacology , Keratins/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins/genetics , Sequence Analysis , Transcription Factors/genetics , Transcription Factors/metabolism
20.
J Biol Chem ; 271(3): 1416-23, 1996 Jan 19.
Article in English | MEDLINE | ID: mdl-8576132

ABSTRACT

Expression of keratin proteins, markers of epidermal differentiation and pathology, is uniquely regulated by the nuclear receptors for retinoic acid (RAR) and thyroid hormone (T3R) and their ligands: it is constitutively activated by unliganded T3R, but it is suppressed by ligand-occupied T3R or RAR. This regulation was studied using gel mobility shift assays with purified receptors and transient transfection assays with vectors expressing various receptor mutants. Regulation of keratin gene expression by RAR and T3R occurs through direct binding of these receptors to receptor response elements of the keratin gene promoters. The DNA binding "C" domain of these receptors is essential for both ligand-dependent and -independent regulation. However, the NH2-terminal "A/B" domain of T3R is not required for either mode of regulation of keratin gene expression. Furthermore, v-ErbA, an oncogenic derivative of cT3R, also activates keratin gene expression. In contrast to the previously described mechanism of gene regulation by T3R, heterodimerization with the retinoid X receptor is not essential for activation of keratin gene expression by unliganded T3R. These findings indicate that the mechanism of regulation of keratin genes by RAR and T3R differs significantly from the mechanisms described for other genes modulated by these receptors.


Subject(s)
Gene Expression Regulation , Keratins/biosynthesis , Keratins/genetics , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/metabolism , Base Sequence , Cell Nucleus/metabolism , Cloning, Molecular , DNA-Binding Proteins/metabolism , Escherichia coli , HeLa Cells , Humans , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides , Oncogene Proteins v-erbA/metabolism , Promoter Regions, Genetic , Protein Biosynthesis , Receptors, Retinoic Acid/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Retinoic Acid Receptor alpha , Retinoid X Receptors , Sequence Deletion , Transcription Factors/metabolism , Transfection , Retinoic Acid Receptor gamma
SELECTION OF CITATIONS
SEARCH DETAIL