Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 87(11): 1301-7, 2002 Nov 18.
Article in English | MEDLINE | ID: mdl-12439721

ABSTRACT

Differential diagnosis between keratoacanthomas and well differentiated squamous cell carcinomas based on clinical and histomorphological data is problematic. Recent findings of cellular atypia in a large proportion of keratoacanthomas indicated that these potentially 'self-healing' cutaneous neoplasms had the potential for malignant progression. Another malignancy-associated criterion is enhanced angiogenesis with increased microvessel density. To provide further diagnostic markers for keratoacanthomas we examined microvessel density on paraffin sections of 13 keratoacanthomas in comparison with 10 normal skin biopsies and 16 late-stage skin squamous cell carcinomas by counting and by computer-assisted image analysis of CD31-immunostained vessels. A significant increase of microvessel density in 'hot spots' was observed in keratoacanthomas as compared to normal skin. Furthermore, when keratoacanthomas were subdivided into tumours with and without malignancy-associated atypic areas, only those with atypia (n=6) were significantly better vascularised than normal skin and had a mean microvessel density in the range of late-stage squamous cell carcinomas. Both keratoacanthoma subtypes revealed comparable levels of inflammatory cell infiltration, tumour cell proliferation and vascular endothelial growth factor expression (mRNA and protein). Thus, in addition to malignancy-associated cellular atypia, increased microvessel density may serve as further diagnostic parameter to discriminate keratoacanthomas with a potential to progress to malignancy.


Subject(s)
Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/diagnosis , Cell Transformation, Neoplastic , Keratoacanthoma/diagnosis , Neovascularization, Pathologic , Skin Diseases/diagnosis , Skin Neoplasms/blood supply , Skin Neoplasms/diagnosis , Biopsy , Carcinoma, Squamous Cell/pathology , Diagnosis, Differential , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Keratoacanthoma/pathology , Microcirculation , Skin Diseases/pathology , Skin Neoplasms/pathology
2.
Invest Radiol ; 37(4): 193-8, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11923641

ABSTRACT

RATIONALE: Malignant tumors often show an increased uptake and metabolism of plasma proteins, especially albumin. OBJECTIVES: Determine whether the accumulation of low loaded Gd-albumin improves visualization of malignant tumors by MRI. METHODS: Twelve nude mice with heterotransplanted squamous cell carcinomas were studied. The signal intensity of tumor, blood, liver, kidney and muscle tissue was studied in MR images after application of Gd-albumin during a period of 144 hours. MRI results were histologically correlated after simultaneously injection of Gd- and fluorescein-labeled albumins in 9 nude mice. RESULTS: Although liver and kidney had a maximum increase in signal intensity within 30 minutes, tumors showed a delayed 51% increase in the 24 hours after application. Histologic and fluorescence evaluation demonstrated albumin localization in tumors predominantly in stroma and necroses. CONCLUSIONS: Gd-albumin is efficiently accumulated in SCC transplants. MRI with low loaded Gd-albumin may offer relevant opportunities for recognizing tumors sensitive to a therapy with cyostic drug-labeled albumins.


Subject(s)
Albumins/pharmacokinetics , Carcinoma, Squamous Cell/metabolism , Gadolinium DTPA/pharmacokinetics , Magnetic Resonance Imaging/methods , Animals , Carcinoma, Squamous Cell/pathology , Contrast Media , Fluorescein/pharmacokinetics , Kidney/metabolism , Liver/metabolism , Mice , Mice, Nude , Muscles/metabolism , Neoplasm Transplantation , Transplantation, Heterologous
3.
Am J Pathol ; 159(4): 1567-79, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11583982

ABSTRACT

Tumor microenvironment is crucial for cancer growth and progression as evidenced by reports on the significance of tumor angiogenesis and stromal cells. Using the HaCaT/HaCaT-ras human skin carcinogenesis model, we studied tumor progression from benign tumors to highly malignant squamous cell carcinomas. Progression of tumorigenic HaCaT-ras clones to more aggressive and eventually metastatic phenotypes was reproducibly achieved by their in vivo growth as subcutaneous tumors in nude mice. Their enhanced malignant phenotype was stably maintained in recultured tumor cells that represented, identified by chromosomal analysis, a distinct subpopulation of the parental line. Additional mutagenic effects were apparent in genetic alterations involving chromosomes 11 and 2, and in amplification and overexpression of the H-ras oncogene. Importantly, in vitro clonal selection of benign and malignant cell lines never resulted in late-stage malignant clones, indicating the importance of the in vivo environment in promoting an enhanced malignant phenotype. Independently of their H-ras status, all in vivo-progressed tumor cell lines (five of five) exhibited a constitutive and stable expression of the hematopoietic growth factors granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor, which may function as autocrine/paracrine mediators of tumor progression in vivo. Thus, malignant progression favored by the in vivo microenvironment requires both clonal selection of subpopulations adapted to in vivo growth and mutational events leading to stable functional alterations.


Subject(s)
Autocrine Communication/physiology , Carcinoma/physiopathology , Granulocyte Colony-Stimulating Factor/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Mutagenesis/physiology , Skin Neoplasms/physiopathology , Carcinoma/genetics , Carcinoma/pathology , Clone Cells/physiology , Cytogenetic Analysis , Disease Progression , Gene Amplification , Gene Expression , Genes, ras , Humans , Oncogenes/genetics , Phenotype , Selection, Genetic , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transfection , Tumor Cells, Cultured
4.
J Invest Dermatol ; 116(5): 816-20, 2001 May.
Article in English | MEDLINE | ID: mdl-11348477

ABSTRACT

Organotypic cocultures of keratinocytes and fibroblasts generate a normal epidermis irrespective of the species and tissue origin of fibroblasts. The use of mouse fibroblasts and human keratinocytes facilitates the identification of the origin of compounds involved in epidermal tissue reconstitution and growth regulation. Moreover, the functional significance for the keratinocyte phenotype of genetically modified fibroblasts from transgenic or knockout mice, even those exhibiting an embryonic lethal phenotype, can be studied in such heterologous in vitro tissue equivalents. Here we communicate results of such studies revealing the antagonistic function of mouse fibroblasts defective in the AP-1 constituents c-Jun and JunB, respectively, on human keratinocyte growth and differentiation. Furthermore, the hematopoietic growth factor granulocyte macrophage-colony stimulating factor has been identified as a novel regulator of keratinocyte growth and differentiation. As will be reported in detail elsewhere both granulocyte macrophage-colony stimulating factor and keratinocyte growth factor have been identified as major mediators of fibroblast-keratinocyte interactions and their expression is induced via AP-1 by interleukin-1 released by the epithelial cells. Thus, these heterologous cocultures provide a novel promising tool for elucidating molecular mechanisms of epithelial-mesenchymal interactions and their consequences on epithelial cell proliferation and differentiation.


Subject(s)
Keratinocytes/cytology , Adult , Animals , Cell Differentiation/physiology , Cell Division/physiology , Embryo, Mammalian , Fibroblasts/metabolism , Humans , Mice , Mice, Knockout/genetics , Organ Culture Techniques , Proto-Oncogene Proteins c-jun/deficiency , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/physiology
5.
Gene Ther ; 8(3): 232-8, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11313795

ABSTRACT

The potential of gene therapy to treat premalignant disease or recurrent cancer has not been investigated. The goal of the present investigation was to explore the efficacy of pro-drug-mediated, suicide gene therapy as a strategy to treat incipient neoplasia in stratified squamous epithelium. To test this strategy, a tissue model of premalignancy was generated by mixing normal human keratinocytes (NHK) that express the bacterial cytosine deaminase gene (CD) with premalignant keratinocytes which have been genetically marked with the bacterial gene for beta-galactosidase (II-4-beta-gal) in skin-like organotypic cultures. Preliminary studies in monolayer cultures demonstrated that CD-transduced NHK (NHK/CD) efficiently expressed the transgene and deaminated the pro-drug 5-fluorocytosine (5FC) to the toxic product 5-fluorouracil (5FU). The capacity of NHK/CD to kill II-4-beta-gal cells through bystander effect was assayed in both submerged culture and in the organotypic model of premalignancy. In submerged cultures, it was found that CD-mediated killing of II-4-beta-gal cells did not require cell-cell contact and that the LD(50) of 5FC for efficient bystander killing of II-4-beta-gal was 0.5 mM. When this concentration of pro-drug was used in organotypic cultures, a significant number of dysplastic II-4-beta-gal cells were eliminated from the tissue. Bystander killing of II-4-beta-gal cells was related to the number of NHK/CD present. These findings demonstrated that potentially malignant keratinocytes could be eliminated from a dysplastic tissue through activation of pro-drug and killing of adjacent cells through the bystander effect. By establishing an in vitro model to eliminate premalignant cells using suicide gene therapy, these studies provide a new approach for the treatment of incipient cancer as it develops, thereby preventing invasive disease.


Subject(s)
Carcinoma in Situ/therapy , Genetic Therapy/methods , Mouth Neoplasms/therapy , Precancerous Conditions/therapy , Antimetabolites/pharmacology , Cell Culture Techniques , Cell Death/drug effects , Cytosine Deaminase , Dose-Response Relationship, Drug , Flucytosine/pharmacology , Gene Expression , Humans , Infant, Newborn , Keratinocytes/drug effects , Keratinocytes/enzymology , Male , Nucleoside Deaminases/genetics , Prodrugs/pharmacology
6.
J Cell Biol ; 152(4): 777-84, 2001 Feb 19.
Article in English | MEDLINE | ID: mdl-11266468

ABSTRACT

The plasminogen (Plg)/plasminogen activator (PA) system plays a key role in cancer progression, presumably via mediating extracellular matrix degradation and tumor cell migration. Consequently, urokinase-type PA (uPA)/plasmin antagonists are currently being developed for suppression of tumor growth and angiogenesis. Paradoxically, however, high levels of PA inhibitor 1 (PAI-1) are predictive of a poor prognosis for survival of patients with cancer. We demonstrated previously that PAI-1 promoted tumor angiogenesis, but by an unresolved mechanism. We anticipated that PAI-1 facilitated endothelial cell migration via its known interaction with vitronectin (VN) and integrins. However, using adenoviral gene transfer of PAI-1 mutants, we observed that PAI-1 promoted tumor angiogenesis, not by interacting with VN, but rather by inhibiting proteolytic activity, suggesting that excessive plasmin proteolysis prevents assembly of tumor vessels. Single deficiency of uPA, tissue-type PA (tPA), uPA receptor, or VN, as well as combined deficiencies of uPA and tPA did not impair tumor angiogenesis, whereas lack of Plg reduced it. Overall, these data indicate that plasmin proteolysis, even though essential, must be tightly controlled during tumor angiogenesis, probably to allow vessel stabilization and maturation. These data provide insights into the clinical paradox whereby PAI-1 promotes tumor progression and warrant against the uncontrolled use of uPA/plasmin antagonists as tumor angiogenesis inhibitors.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Endopeptidases/metabolism , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Plasminogen Activator Inhibitor 1/pharmacology , Vitronectin/metabolism , Animals , Endothelium, Vascular/drug effects , Fibrinolysin/metabolism , Keratinocytes/pathology , Mice , Mice, Mutant Strains , Muscle Neoplasms/blood supply , Neoplasm Invasiveness , Neoplasms, Experimental/blood supply , Neovascularization, Pathologic/etiology , Plasminogen Activator Inhibitor 1/metabolism , Protein Binding , Vitronectin/genetics
7.
J Invest Dermatol ; 116(1): 85-92, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11168802

ABSTRACT

There is accumulating evidence of the critical role of tumor stroma in carcinoma development and progression. We have studied the significance of stromal components in regulating matrix metalloproteinases in different stages of human skin carcinogenesis using the HaCaT keratinocyte transformation model. Expression of matrix metalloproteinase 1 and matrix metalloproteinase 13 was analyzed in nontumorigenic HaCaT cells and their c-Ha-ras-transformed tumorigenic clones, benign A-5 and malignant A-5RT3, in response to different matrices and cocultured fibroblasts as well as in transplants in nude mice. When cultured on a collagen type I gel, expression of matrix metalloproteinase 1 mRNA was induced in A-5 and A-5RT3 but less in HaCaT cells, whereas matrix metalloproteinase 13 was only induced in A-5 cells. Induction of matrix metalloproteinase 1 by collagen was also observed in two other malignant HaCaT-ras clones as well as in 2/2 primary squamous cell carcinoma lines. In organotypic cocultures with skin fibroblasts, matrix metalloproteinase 1 mRNA and protein was further strongly upregulated in A-5RT3 cells but less in HaCaT and A-5 cells. Importantly, matrix metalloproteinase 1 was also upregulated in fibroblasts when cocultured with A-5RT3 cells. In vivo, A-5RT3 transplants and subcutaneous tumors expressed matrix metalloproteinase 1 mRNA consistently, preferentially at the tumor front to the mouse stroma. In contrast, matrix metalloproteinase 1 expression was absent in the transplants of A-5 cells and HaCaT cells. Thus, our results demonstrate the specific induction of matrix metalloproteinase 1 in malignant keratinocytes by fibroblasts, supposedly through paracrine-acting factors, and a reciprocally enhanced expression in fibroblasts. This further substantiates the important role of tumor stroma in regulating the expression of matrix metalloproteinase 1, a major matrix-degrading proteinase implicated in tumor invasion.


Subject(s)
Keratinocytes/metabolism , Matrix Metalloproteinase 1/genetics , Skin Neoplasms/genetics , Skin/cytology , Stromal Cells/metabolism , Cell Line, Transformed , Coculture Techniques , Collagen/pharmacology , Collagenases/genetics , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression/drug effects , Gene Expression Regulation , Humans , Matrix Metalloproteinase 13 , Skin Diseases/genetics
8.
Cell ; 103(5): 745-55, 2000 Nov 22.
Article in English | MEDLINE | ID: mdl-11114331

ABSTRACT

Interactions between mesenchymal and epithelial cells are responsible for organogenesis and tissue homeostasis. This mutual cross-talk involves cell surface proteins and soluble factors, which are mostly the result of regulated transcription. To elucidate dimer-specific functions of the AP-1 family of transcription factors, we reconstituted skin by combining primary human keratinocytes and mouse wild-type, c-jun(-/-), and junB(-/-) fibroblasts. We have discovered an antagonistic function of these AP-1 subunits in the fibroblast-mediated paracrine control of keratinocyte proliferation and differentiation, and traced this effect to the IL-1-dependent regulation of KGF and GM-CSF. These data suggest that the relative activation state of these AP-1 subunits in a non-cell-autonomous, transregulatory fashion directs regeneration of the epidermis and maintenance of tissue homeostasis in skin.


Subject(s)
Cytokines/metabolism , Epidermis/metabolism , Mesoderm/metabolism , Proto-Oncogene Proteins c-jun/physiology , Skin/metabolism , Animals , Cell Differentiation , Cell Division , Cells, Cultured , Coculture Techniques , Collagen/metabolism , Enzyme-Linked Immunosorbent Assay , Estrogen Antagonists/pharmacology , Fibroblast Growth Factor 7 , Fibroblast Growth Factors/metabolism , Fibroblasts/metabolism , Fluorescent Antibody Technique, Indirect , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Keratinocytes/metabolism , Mice , Models, Biological , Mutation , RNA, Messenger/metabolism , Rats , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tamoxifen/pharmacology , Transcription Factor AP-1/metabolism
9.
Blood ; 96(12): 3857-65, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11090070

ABSTRACT

The 5-lipoxygenase (5-LO) pathway in human CD34(+) hematopoietic progenitor cells, which were induced to differentiate into dendritic cells (DCs) by cytokines in vitro and in DCs of lymphoid tissues in situ, was examined. Extracts prepared from HPCs contained low levels of 5-LO or 5-LO-activating protein. Granulocyte-macrophage colony-stimulating factor (GM-CSF) plus tumor necrosis factor-alpha (TNF-alpha) promoted DC differentiation and induced a strong rise in 5-LO and FLAP expression. Fluorescence-activated cell sorter (FACS) analyses identified a major DC population coexpressing human leukocyte antigen (HLA)-DR/CD80 and monocytic or Langerhans cell markers. Transforming growth factor-beta1 (TGF-beta-1), added to support DC maturation, strongly promoted the appearance of CD1a(+)/Lag(+) Langerhans-type cells as well as mature CD83(+) DCs. TGF-beta-1 further increased 5-LO and FLAP expression, recruited additional cells into the 5-LO(+) DC population, and promoted production of 5-hydroxyeicosatetraenoic acid and leukotriene B(4) in response to calcium (Ca(++)) ionophore A23187. These in vitro findings were corroborated by 5-LO expression in distinct DC phenotypes in vivo. Scattered 5-LO and FLAP in situ hybridization signals were recorded in cells of paracortical T-lymphocyte-rich areas and germinal centers (GCs) of lymph nodes (LNs) and tonsil and in cells of mucosae overlying the Waldeyer tonsillar ring. 5-LO protein localized to both CD1a(+) immature DCs and to CD83(+) mature interdigitating DCs of T-lymphocyte-rich areas of LNs and tonsil. As DCs have the unique ability to initiate naive lymphocyte activation, our data support the hypothesis that leukotrienes act at proximal steps of adaptive immune responses. (Blood. 2000;96:3857-3865)


Subject(s)
Antigens, CD34/analysis , Arachidonate 5-Lipoxygenase/metabolism , Dendritic Cells/enzymology , Arachidonate 5-Lipoxygenase/biosynthesis , Arachidonate 5-Lipoxygenase/genetics , Arachidonic Acid/pharmacology , Calcimycin/pharmacology , Cell Differentiation/drug effects , Cell Lineage , Chemotactic Factors/biosynthesis , Cytokines/pharmacology , Dendritic Cells/cytology , Fetal Blood/cytology , Flow Cytometry , HLA-DR Antigens , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Hydroxyeicosatetraenoic Acids/biosynthesis , Immunohistochemistry , Immunophenotyping , Ionophores/pharmacology , Leukotriene B4/biosynthesis , Lymph Nodes/enzymology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Macrophages/enzymology , Palatine Tonsil/enzymology
10.
Biol Chem ; 381(5-6): 497-507, 2000.
Article in English | MEDLINE | ID: mdl-10937882

ABSTRACT

In order to investigate the correlations between constitutive proteinase expression and the degree of tumorigenicity of cancer cells we have studied a model system of three keratinocyte cell lines. RT-PCR studies showed that the cell lines express the genes of matrix metalloproteinase-2, -3, -7, -9, -10 and -11, indicating that they are able to synthesize the corresponding enzymes. Actual MMP synthesis was proven by zymography and Western blotting. In conditioned media gelatinolytic activities or immunoreactive forms of MMP-2, -3, -7, -9, -10 and -11 were detected. The signal intensities showed that MMP secretion increases in the order HaCaT < A5 < or = II-4RT, whereas only MMP-11 is secreted by all cell lines in equal amounts. Intracellularly, enhanced levels of one or both of the tumorigenic variants were only found for MMP-3, -9 and -10, suggesting special functions of these intracellular MMP pools for the tumorigenic cell lines. For MMP-11 exclusive expression in stromal fibroblasts of tumor tissues is widely accepted; however, our results and three other recent reports demonstrate that this concept is not generally valid. In conclusion, the three keratinocyte cell lines investigated here represent an excellent model for studying constitutive expression and secretion of MMPs in correlation to the degree of in vivo tumorigenicity.


Subject(s)
Keratinocytes/enzymology , Matrix Metalloproteinases/metabolism , Base Sequence , Blotting, Western , Cell Extracts , Cell Line , Cell Line, Transformed , Culture Media, Conditioned , DNA Primers , Gelatin/metabolism , Humans , Matrix Metalloproteinases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
J Invest Dermatol ; 114(6): 1075-84, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10844548

ABSTRACT

Balanced keratinocyte proliferation and differentiation resulting in regular tissue organization strictly depend on dermal support. Organotypic cultures represent biologically relevant in vitro models to study the molecular mechanism of the underlying dermal-epidermal interactions. To mimic the state of resting fibroblasts in the dermis, postmitotic (irradiated) fibroblasts were incorporated in the collagen matrix, where they typically support epidermal proliferation and tissue organization. In coculture with keratinocytes, fibroblasts exhibit an enhanced expression of keratinocyte growth factor and the interleukin-1 receptor (type I), which further increase with culture time. In cocultured keratinocytes, keratinocyte growth factor receptor as well as RNA expression and protein release of interleukin-1alpha and interleukin-1beta are upregulated. We hypothesized that the modulated cytokine expression represents a basic mechanism for keratinocyte growth regulation. The functional significance of this double paracrine pathway, i.e., induction of keratinocyte growth factor expression in fibroblasts by keratinocytes via release of interleukin-1, was confirmed by interfering with both signaling elements: (i) interleukin-1-neutralizing antibodies and interleukin-1 receptor antagonist significantly inhibited keratinocyte growth factor release, keratinocyte proliferation, and tissue formation comparable to the effect produced by keratinocyte-growth-factor-blocking antibodies; (ii) addition of keratinocyte growth factor to cocultures with inactivated interleukin-1 pathway completely reverted growth inhibition; (iii) in organotypic cocultures with subthreshold fibroblast numbers both interleukin-1 and keratinocyte growth factor restored the impaired epidermal morphogenesis. Thus, epidermal tissue regeneration in organotypic cocultures is mainly regulated by keratinocyte-derived interleukin-1 signaling, which induces keratinocyte growth factor expression in cocultured fibroblasts. This demonstrates a novel role for interleukin-1 in skin homeostasis substantiating data from wound healing studies in vivo.


Subject(s)
Fibroblast Growth Factors , Fibroblasts/metabolism , Growth Substances/biosynthesis , Interleukin-1/pharmacology , Keratinocytes/cytology , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Coculture Techniques , Culture Media, Serum-Free , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Growth Substances/pharmacology , Humans , Mitosis , Time Factors
12.
J Pathol ; 190(5): 579-88, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10727984

ABSTRACT

The three mammalian isoforms of transforming growth factor-beta (TGF-beta1, -beta2, and -beta3) are potent regulators of cell growth, differentiation, and extracellular matrix deposition. To study their role in skin carcinogenesis, normal human keratinocytes, early (31) and late (310) passage immortalized keratinocytes (HaCaT cells), and five HaCaT-ras clones exhibiting benign (A-5, I-7), malignant (II-4, A-5 RT1), and highly aggressive (A-5 RT3) tumourigenic phenotypes were examined for the expression of TGF-beta isoforms, by immunohistochemistry. This was performed under in vivo conditions, in surface transplants and subcutaneously growing tumours in nude mice. Generally, all tissues that formed keratinized epithelia demonstrated an immunostaining pattern similar to normal human skin. TGF-beta1 was localized to the upper differentiated layers, the stratum granulosum and corneum, in a perimembranous pattern, whereas TGF-beta2 and, weaker, TGF-beta3 immunostaining was present in all suprabasal layers of normal keratinizing epithelia. In contrast, non-keratinizing transplants of non-tumourigenic or highly aggressive cells showed little to no immunoreactivity for TGF-beta1. Whereas TGF-beta2 expression was moderate in the upper layers of non-tumourigenic epithelia, large tumour cells of the malignant HaCaT-ras clones, particularly at the invasion front, were strongly positive for TGF-beta2. TGF-beta3 immunostaining was most pronounced in the stroma of malignant tumours, implying its paracrine induction by the malignant tumour transplants. These results suggest differential functions for each TGF-beta isoform in epidermal carcinogenesis, such that TGF-beta1 is associated with the more differentiated state, TGF-beta2 with highly malignant and invading cells, and TGF-beta3 with tumour stroma formation and angiogenesis. Furthermore, the expression of TGF-betas by both early- and late-stage tumours implies that the isoforms may have distinct functions at different stages of malignancy, supporting their dual role in skin carcinogenesis.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Keratinocytes/metabolism , Skin Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Humans , Immunoenzyme Techniques , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Protein Isoforms/metabolism , Skin Neoplasms/pathology , Tumor Cells, Cultured
13.
Br J Cancer ; 82(3): 591-600, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10682671

ABSTRACT

Angiogenesis is a crucial event in carcinogenesis and its onset has been associated with premalignant tumour stages. In order to elucidate the significance of angiogenesis in different stages of epithelial skin tumours, we analysed the vessel density in ten normal skin samples, 14 actinic keratosis (AK), 12 hypertrophic AKs, and in nine early- and 16 late-stage squamous cell carcinomas (SCCs). Mean vascular density was quantitated by counting the number of CD 31-immunostained blood vessels and by morphometric assessment of stained vessel area by computer-assisted image analysis. The results from both methods were well correlated. Mean vascular density was similar in normal dermis and in AK, and only slightly elevated in hypertrophic AKs and early SCC stages (tumour thickness < 2 mm). Only late-stage SCCs infiltrating the subcutis exhibited a significant increase in vascularization. Vessel density was independent of tumour localization, degree of proliferation and inflammatory cell infiltration. Furthermore, tumour vascularization was not correlated with the expression of vascular endothelial growth factor, a major angiogenic factor, as revealed by in situ hybridization and immunohistochemistry. The restriction of enhanced vascularization to increased tumour thickness may be a major reason for the rather low metastatic spread of cutaneous SCCs.


Subject(s)
Carcinoma, Squamous Cell/blood supply , Neovascularization, Pathologic , Skin Neoplasms/blood supply , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Cell Division , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Humans , Immunohistochemistry , Lymphokines/genetics , Lymphokines/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Precancerous Conditions/blood supply , Precancerous Conditions/immunology , Precancerous Conditions/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Skin/blood supply , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
14.
Oncogene ; 19(2): 248-57, 2000 Jan 13.
Article in English | MEDLINE | ID: mdl-10645003

ABSTRACT

Collagenase-3 (MMP-13) is characterized by an exceptionally wide substrate specificity and restricted expression. MMP-13 is specifically expressed by transformed human keratinocytes in squamous cell carcinomas in vivo and its expression correlates with their invasion capacity. Here, we show, that interferon-gamma (IFN-gamma) markedly inhibits expression of MMP-13 by human cutaneous SCC cells (UT-SCC-7) and by ras-transformed human epidermal keratinocytes (A-5 cells) at the transcriptional level. In addition, IFN-gamma inhibits collagenase-1 (MMP-1) expression in these cells. IFN-gamma abolished the enhancement of MMP-13 and MMP-1 expression by transforming growth factor-beta (TGF-beta) and tumor necrosis factor-alpha (TNF-alpha), and inhibited invasion of A-5 cells through type I collagen. IFN-gamma also rapidly and transiently activates extracellular signal-regulated kinase 1,2 (ERK1,2) and blocking ERK1,2 pathway (Raf/MEK1,2/ERK1,2) by specific MEK1,2 inhibitor PD98059 partially (by 50%) prevents Ser-727 phosphorylation of STAT1 and suppression of MMP-13 expression by IFN-gamma. Furthermore, Ser-727 phosphorylation of STAT1 by ERK1,2, or independently of ERK1,2 activation is associated with marked reduction in MMP-13 expression. These observations identify a novel role for IFN-gamma as a potent inhibitor of collagenolytic activity and invasion of transformed squamous epithelial cells, and show that inhibition of MMP-13 expression by IFN-gamma involves activation of ERK1,2 and STAT1.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Collagenases/biosynthesis , DNA-Binding Proteins/metabolism , Interferon-gamma/pharmacology , Keratinocytes/enzymology , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/biosynthesis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Trans-Activators/metabolism , Cell Line, Transformed , Cell Survival/drug effects , Collagenases/genetics , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Matrix Metalloproteinase 13 , Matrix Metalloproteinases/genetics , Mitogen-Activated Protein Kinase 3 , Organ Specificity/genetics , Recombinant Proteins , STAT1 Transcription Factor , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Transforming Growth Factor beta/antagonists & inhibitors , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/antagonists & inhibitors
15.
J Cell Sci ; 113 Pt 2: 227-35, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10633074

ABSTRACT

Collagenase-3 (MMP-13) is a human matrix metalloproteinase specifically expressed by transformed squamous epithelial cells, i.e. squamous cell carcinoma (SCC) cells in culture and in vivo. Here, we have elucidated the signaling pathways regulating MMP-13 expression in transformed human epidermal keratinocytes, i.e. ras-transformed HaCaT cell line A-5 and cutaneous SCC cell line (UT-SCC-7). Treatment with tumor necrosis factor-(alpha) (TNF-(alpha) resulted in activation of extracellular signal-regulated kinase (ERK)1,2, Jun N-terminal kinase and p38 mitogen-activated protein kinase (MAPK) in both cell lines. In addition, transforming growth factor-(beta) (TGF-(beta) activated p38 MAPK in both cell lines, and ERK2 in A-5 cells. Selective inhibition of p38 activity with SB 203580 abolished the enhancement of MMP-13, as well as collagenase-1 (MMP-1) and 92-kDa gelatinase (MMP-9) expression by TNF-(alpha) and TGF-(beta). Blocking the ERK1, 2 pathway by PD 98059 had no effect on the induction of MMP-13 expression by TNF-(alpha) or TGF-(beta), but potently suppressed MMP-1 and MMP-9 production. Inhibition of p38 activity by SB 203580 also suppressed collagenolytic activity produced by both cell lines and inhibited invasion of TNF-(alpha) or TGF-(beta) stimulated A-5 cells through type I collagen and reconstituted basement membrane (Matrigel). These results show that activation of p38 MAPK pathway plays a crucial role in the invasive phenotype of transformed squamous epithelial cells, suggesting p38 MAPK as a target to specifically inhibit their invasion.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Collagenases/genetics , Keratinocytes/enzymology , Matrix Metalloproteinase 1/genetics , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Cell Line, Transformed , Collagenases/metabolism , Enzyme Activation/drug effects , Gene Expression/drug effects , Genes, fos/drug effects , Genes, jun/drug effects , Humans , JNK Mitogen-Activated Protein Kinases , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 13 , Matrix Metalloproteinase 9/biosynthesis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Transforming Growth Factor beta/pharmacology , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases
16.
Int J Cancer ; 83(6): 780-9, 1999 Dec 10.
Article in English | MEDLINE | ID: mdl-10597195

ABSTRACT

Tumor progression is characterized by an increasing escape of tumor cells from the growth control of their microenvironment, often caused by aberrant expression of growth factors. In the human skin carcinoma model system, based on the HaCaT keratinocyte line, tumor progression to high-grade malignant cells is associated with constitutive expression and secretion of the hematopoietic growth factors G-CSF and GM-CSF in vitro and in vivo. All HaCaT keratinocyte variants express the G-CSF and the GM-CSF receptors at levels comparable to normal keratinocytes. Consequently, they exhibit a stimulation of cell proliferation and migration in culture when treated with these factors. Moreover, both proliferation and migration of the high-grade malignant cells were strongly inhibited by neutralizing antibodies to G-CSF and GM-CSF, respectively. This demonstrates the functional role of these factors in high-grade malignant HaCaT cells through an autocrine mechanism in vitro and implies their significance in tumor progression in vivo. In light of the increasing use of G-CSF and GM-CSF in adjuvant tumor therapy, our data, as well as those discussed for head-and-neck tumors and gliomas, warrant a careful re-evaluation of the clinical application of both factors.


Subject(s)
Granulocyte Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Animals , Cell Division , Cell Movement , Clone Cells , DNA Primers , Disease Progression , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Mice , Mice, Nude , RNA, Messenger/genetics , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/physiopathology , Transcription, Genetic , Transplantation, Heterologous , Tumor Cells, Cultured
17.
Am J Pathol ; 155(5): 1557-67, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10550313

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) and granulocyte macrophage colony-stimulating factor (GM-CSF) and/or their receptors are increasingly detected in solid human tumors, although little is known about their function in tumor growth and invasion. We analyzed RNA and protein expression of both factors and their receptors in 22 human gliomas (WHO grade II, III, and IV) and derived cell cultures. G-CSF, GM-CSF, and/or their receptors were expressed in all tumors and derived cell cultures, but coexpression of both factors and receptors was almost exclusively found in grade IV glioblastomas and thus correlated with advanced tumor stage. The functional significance of G-CSF and GM-CSF as regulators for glioma cells was demonstrated by 1) stimulation of proliferation and migration in tumor cells expressing one or both receptors by the corresponding factor; 2) inhibition of growth and migration of glioma cells expressing G-CSF, GM-CSF, and their receptors by neutralizing antibodies to both factors. These results indicate a significant role for both factors in the autocrine regulation of growth and migration in late-stage malignant gliomas and suggest a shift from paracrine to autocrine regulation with tumor progression. The implication of G-CSF and GM-CSF in glioblastoma growth regulation could make these factors further prognostic indicators and raises questions concerning their use in cancer therapy.


Subject(s)
Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/pathology , Glioma/pathology , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Autocrine Communication , Cell Differentiation/drug effects , Cell Division/drug effects , Central Nervous System Neoplasms/physiopathology , Glioma/physiopathology , Granulocyte Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Tumor Cells, Cultured
18.
Oncogene ; 18(41): 5638-45, 1999 Oct 07.
Article in English | MEDLINE | ID: mdl-10523843

ABSTRACT

UV-radiation is a major risk factor for non-melanoma skin cancer causing specific mutations in the p53 tumor suppressor gene and other genetic aberrations. We here propose that elevated temperature, as found in sunburn areas, may contribute to skin carcinogenesis as well. Continuous exposure of immortal human HaCaT skin keratinocytes (possessing UV-type p53 mutations) to 40 degrees C reproducibly resulted in tumorigenic conversion and tumorigenicity was stably maintained after recultivation of the tumors. Growth at 40 degrees C was correlated with the appearance of PARP, an enzyme activated by DNA strand breaks and the level corresponded to that seen after 5 Gy gamma-radiation. Concomitantly, comparative genomic hybridization (CGH) analyis demonstrated that chromosomal gains and losses were present in cells maintained at 40 degrees C while largely absent at 37 degrees C. Besides individual chromosomal aberrations, all tumor-derived cells showed gain of chromosomal material on 11q with the smallest common region being 11q13.2 to q14.1. Cyclin D1, a candidate gene of that region was overexpressed in all tumor-derived cells but cyclinD1/cdk4/cdk6 kinase activity was not increased. Thus, these data demonstrate that long-term thermal stress is a potential carcinogenic factor in this relevant skin cancer model, mediating its effect through induction of genetic instability which results in selection of tumorigenic cells characterized by gain of 11q.


Subject(s)
Cell Transformation, Neoplastic , Hot Temperature , Keratinocytes/pathology , Aneuploidy , Animals , Cell Line, Transformed , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 11/ultrastructure , Cyclin D1/biosynthesis , Cyclin D1/genetics , DNA Damage , Enzyme Induction , Gamma Rays , Humans , Mice , Nucleic Acid Hybridization , Poly(ADP-ribose) Polymerases/biosynthesis , Poly(ADP-ribose) Polymerases/genetics , Skin/cytology , Skin Neoplasms/etiology
19.
J Invest Dermatol ; 113(3): 384-91, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10469338

ABSTRACT

The role of cell interactions during early neoplastic progression in human skin is not well understood. We report that the fate and behavior of low-grade malignant cells in stratified epithelium is dependent on their interactions with neighboring cells and with extracellular matrix during the early events in neoplastic progression. We utilized an organotypic tissue model which mimics premalignancy to monitor malignant cells (II-4) genetically marked with beta-gal and grown in the context of either normal human keratinocytes or the immortalized cell line HaCaT. HaCaT cells were permissive for clonal expansion of II-4 cells at ratios of 4:1, 12:1, and 50:1 (HAC:II-4) when compared with coculture with normal human keratinocytes. This II-4 cell expansion was associated with the failure of neighboring HaCaT cells to induce differentiation and cell cycle withdrawal of II-4, as had been seen in the context of normal human keratinocytes. When 12:1 mixtures (NHK:II-4) were stripped of all suprabasal cells and regrown, all beta-gal cells were lost showing that these normal human keratinocyte-suppressed II-4 cells had been actively sorted to a suprabasal position where their clonal expansion was limited. These growth-suppressive effects of normal human keratinocytes were found to be conditional on direct cell-cell contact, as II-4 formed colonies when trypsinized from 12:1 (NHK:II-4) mixtures and grown at clonal density in submerged culture. The distribution and behavior of low-grade malignant cells was therefore dependent on the state of transformation of adjacent keratinocytes and on cell-matrix interactions. These results demonstrate that alterations in the cellular microenvironment are central to the induction of clonal expansion and early neoplastic progression in stratified epithelium.


Subject(s)
Cell Communication , Cell Transformation, Neoplastic , Keratinocytes/pathology , Skin Neoplasms/etiology , Cell Differentiation , Cell Division , Cells, Cultured , Humans
20.
Differentiation ; 64(5): 263-75, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10374263

ABSTRACT

Benign and malignant HaCaT-ras clones, derived from immortalized HaCaT cells were grown as nude mouse surface transplants rendering a human tumor progression model. Searching for malignancy-related alterations, the deposition, localization and mRNA of basement membrane and hemidesmosome components were analysed by immunofluorescence, in situ hybridization and electron microscopy. Initially, at 1 week epithelia of benign and malignant cells revealed a similarly low polarity and an enlarged 'activated basal' compartment, reflected by partial dislocation and extended pericellular staining of the hemidesmosome constituent integrin alpha 6 beta 4 seen by immunofluorescence. Whereas benign grafts eventually normalized, closely resembling grafts of HaCaT cells, malignant growth was correlated with a persisting epithelial activation state and continuing higher expression of alpha 6 (by immunofluorescence and in situ hybridization). The basement membrane components bullous pemphigoid antigen 1, laminin-5 and collagen IV exhibited a largely linear distribution at 1 week. However, in the malignant cell transplants initially minor basement membrane discontinuities became more severe at around 2 weeks, associated with close stromal cell contacts, angiogenesis and invasion. Most striking were basement membrane alterations seen by electron microscopy. At 1 week stretches of basement membrane had developed in malignant transplants, though to a much lesser extent than in benign specimens. With invasion these basement membrane structures mostly disappeared despite persistent although variable immunofluorescence, suggesting high turnover without ultrastructural assembly. The hemidesmosome structures were defective throughout, completely lacking anchoring plaques with keratin filaments, whereas they were still associated with basement membrane deposits. Thus, malignant HaCaT-ras transplants, while initially resembling regenerating wounds, revealed an increasing loss of tissue polarity and basement membrane structures, which seemed to be accelerated upon stromal cell contacts.


Subject(s)
Basement Membrane/ultrastructure , Desmosomes/ultrastructure , Genes, ras , Skin Neoplasms/pathology , Animals , Basement Membrane/chemistry , Cell Adhesion Molecules/analysis , Collagen/analysis , Desmosomes/chemistry , Female , Humans , Mice , Neoplasm Transplantation , Skin Neoplasms/chemistry , Skin Neoplasms/ultrastructure , Stromal Cells/physiology , Transplantation, Heterologous , Tumor Cells, Cultured , Kalinin
SELECTION OF CITATIONS
SEARCH DETAIL