Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(3)2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35163299

ABSTRACT

NMSC (non-melanoma skin cancer) is a common tumor in the Caucasian population, accounting for 90% of skin cancers. Among them, squamous cell carcinomas (SCCs) can metastasize and, due to its high incidence, constitute a severe health problem. It has been suggested that cutaneous SCCs with more risk to metastasize express high levels of nuclear IKKα. However, the molecular mechanisms that lead to this enhanced aggressiveness are largely unknown. To understand in depth the influence of nuclear IKKα in skin SCC progression, we have generated murine PDVC57 skin carcinoma cells expressing exogenous IKKα either in the nucleus or in the cytoplasm to further distinguish the tumor properties of IKKα in both localizations. Our results show that IKKα promotes changes in both subcellular compartments, resembling EMT (epithelial-mesenchymal transition), which are more pronounced when IKKα is in the nucleus of these tumor cells. These EMT-related changes include a shift toward a migratory phenotype and induction of the expression of proteins involved in cell matrix degradation, cell survival and resistance to apoptosis. Additionally, we have found that apigenin, a flavonoid with anti-cancer properties, inhibits the expression of IKKα and attenuates most of the pro-tumoral EMT changes induced by IKKα in mouse tumor keratinocytes. Nevertheless, we have found that apigenin only inhibits the expression of the IKKα protein when it is localized in the cytoplasm.


Subject(s)
Apigenin/pharmacology , I-kappa B Kinase/metabolism , Skin Neoplasms/metabolism , Animals , Apigenin/metabolism , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Nucleus/metabolism , Cytoplasm/metabolism , Epithelial-Mesenchymal Transition/physiology , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , I-kappa B Kinase/genetics , Keratinocytes/metabolism , Mice , Signal Transduction/genetics , Skin/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/pathology
2.
Int J Oral Sci ; 12(1): 1, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31900382

ABSTRACT

Odontogenic tumours are a heterogeneous group of lesions that develop in the oral cavity region and are characterized by the formation of tumoural structures that differentiate as teeth. Due to the diversity of their histopathological characteristics and clinical behaviour, the classification of these tumours is still under debate. Alterations in morphogenesis pathways such as the Hedgehog, MAPK and WNT/ß-catenin pathways are implicated in the formation of odontogenic lesions, but the molecular bases of many of these lesions are still unknown. In this study, we used genetically modified mice to study the role of IKKß (a fundamental regulator of NF-κB activity and many other proteins) in oral epithelial cells and odontogenic tissues. Transgenic mice overexpressing IKKß in oral epithelial cells show a significant increase in immune cells in both the oral epithelia and oral submucosa. They also show changes in the expression of several proteins and miRNAs that are important for cancer development. Interestingly, we found that overactivity of IKKß in oral epithelia and odontogenic tissues, in conjunction with the loss of tumour suppressor proteins (p53, or p16 and p19), leads to the appearance of odontogenic tumours that can be classified as ameloblastic odontomas, sometimes accompanied by foci of secondary ameloblastic carcinomas. These tumours show NF-κB activation and increased ß-catenin activity. These findings may help to elucidate the molecular determinants of odontogenic tumourigenesis and the role of IKKß in the homoeostasis and tumoural transformation of oral and odontogenic epithelia.


Subject(s)
Epithelial Cells/metabolism , Genes, Tumor Suppressor , I-kappa B Kinase/biosynthesis , Mouth Mucosa/pathology , Odontogenic Tumors/pathology , Odontoma/pathology , RNA, Messenger/genetics , Animals , Blotting, Western , Epithelial Cells/pathology , Flow Cytometry , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Mice , Mice, Transgenic , Mouth Mucosa/metabolism , Odontogenic Tumors/metabolism , Odontoma/metabolism , Real-Time Polymerase Chain Reaction
3.
Comput Struct Biotechnol J ; 17: 251-262, 2019.
Article in English | MEDLINE | ID: mdl-30867890

ABSTRACT

Lung cancer is the leading worldwide cause of cancer mortality, however, neither curative treatments nor substantial prolonged survival has been achieved, highlighting the need for investigating new proteins responsible for its development and progression. IKKα is an essential protein for cell survival and differentiation, which expression is enhanced in human non-small cell lung cancer (NSCLC) and correlates with poor patient survival, appearing as a relevant molecule in lung cancer progression. However, there are not conclusive results about its role in this type of cancer. We have recently found that IKKα performs different functions and activates different signaling pathways depending on its nuclear or cytoplasmic localization in tumor epidermal cells. In this work, we have studied the involvement of IKKα in lung cancer progression through the generation of lung cancer cell lines expressing exogenous IKKα either in the nucleus or in the cytoplasm. We demonstrate that IKKα signaling promotes increased cell malignancy of NSCLC cells as well as lung tumor progression and metastasis in either subcellular localization, through activation of common protumoral proteins, such as Erk, p38 and mTor. But, additionally, we found that depending on its subcellular localization, IKKα has non-overlapping roles in the activation of other different pathways known for their key implication in lung cancer progression: while cytoplasmic IKKα increases EGFR and NF-κB activities in lung tumor cells, nuclear IKKα causes lung tumor progression through c-Myc, Smad2/3 and Snail activation. These results suggest that IKKα may be a promising target for intervention in human NSCLC.

4.
Aging (Albany NY) ; 11(1): 127-159, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30631004

ABSTRACT

CYLD is a deubiquitinating enzyme known for its role as a tumor suppressor whose mutation leads to skin appendages tumors and other cancers. In this manuscript we report that the tumor suppressor CYLD, similarly to other renowned tumor suppressor genes, protects from premature aging and cancer. We have generated transgenic mice expressing the mutant CYLDC/S protein, lacking its deubiquitinase function, under the control of the keratin 5 promoter, the K5-CYLDC/S mice. These mice express the transgene in different organs, including those considered to be more susceptible to aging, such as skin and thymus. Our results show that K5-CYLDC/S mice exhibit epidermal, hair follicle, and sebaceous gland alterations; and, importantly, they show signs of premature aging from an early age. Typically, 3-month-old K5-CYLDC/S mice exhibit a phenotype characterized by alopecia and kyphosis, and, the histological examination reveals that transgenic mice show signs of accelerated aging in numerous organs such as skin, thymus, pancreas, liver and lung. Additionally, they spontaneously develop tumors of diverse origin. Over-activation of the NF-κB pathway, along with hyperactivation of Akt, JNK and c-Myc, and chronic inflammation, appear as the mechanisms responsible for the premature aging of the K5-CYLDC/S mice.


Subject(s)
Aging, Premature/genetics , Deubiquitinating Enzyme CYLD/metabolism , Hair Follicle/growth & development , Neoplasms/etiology , Animals , Cell Line , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Deubiquitinating Enzyme CYLD/genetics , Gene Expression Regulation/physiology , Humans , Keratinocytes/physiology , Mice , Mice, Transgenic , Mutation , NF-kappa B , Promoter Regions, Genetic , Skin Aging/genetics , Thymus Gland/physiology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
5.
Sci Rep ; 8(1): 13038, 2018 08 29.
Article in English | MEDLINE | ID: mdl-30158566

ABSTRACT

Although Ras genes are frequently mutated in human tumors, these mutations are uncommon in breast cancer. However, many breast tumors show evidences of Ras pathway activation. In this manuscript, we have analyzed and characterized mouse mammary tumors generated by random Sleeping Beauty transposon mutagenesis and identify ERAS -a member of the RAS family silenced in adult tissues- as a new gene involved in progression and malignancy of breast cancer. Forced expression of ERAS in human non-transformed mammary gland cells induces a process of epithelial-to-mesenchymal transition and an increase in stem cells markers; these changes are mediated by miR-200c downregulation. ERAS expression in human tumorigenic mammary cells leads to the generation of larger and less differentiated tumors in xenotransplant experiments. Immunohistochemical, RT-qPCR and bioinformatics analysis of human samples show that ERAS is aberrantly expressed in 8-10% of breast tumors and this expression is associated with distant metastasis and reduced metastasis-free survival. In summary, our results reveal that inappropriate activation of ERAS may be important in the development of a subset of breast tumors. These findings open the possibility of new specific treatments for this subset of ERAS-expressing tumors.


Subject(s)
Breast Neoplasms/physiopathology , Oncogene Protein p21(ras)/metabolism , Animals , Breast Neoplasms/pathology , Carcinogenesis , Cell Differentiation , Cells, Cultured , Epithelial Cells/physiology , Epithelial-Mesenchymal Transition , Humans , Mice , Neoplasm Transplantation , Neoplasms, Experimental/pathology , Neoplasms, Experimental/physiopathology , Oncogene Protein p21(ras)/genetics , Transplantation, Heterologous
7.
Mol Cancer Res ; 15(9): 1255-1264, 2017 09.
Article in English | MEDLINE | ID: mdl-28584022

ABSTRACT

IKKß (encoded by IKBKB) is a protein kinase that regulates the activity of numerous proteins important in several signaling pathways, such as the NF-κB pathway. IKKß exerts a protumorigenic role in several animal models of lung, hepatic, intestinal, and oral cancer. In addition, genomic and proteomic studies of human tumors also indicate that IKBKB gene is amplified or overexpressed in multiple tumor types. Here, the relevance of IKKß in skin cancer was determined by performing carcinogenesis studies in animal models overexpressing IKKß in the basal skin layer. IKKß overexpression resulted in a striking resistance to skin cancer development and an increased expression of several tumor suppressor proteins, such as p53, p16, and p19. Mechanistically, this skin tumor-protective role of IKKß is independent of p53, but dependent on the activity of the Ink4a/Arf locus. Interestingly, in the absence of p16 and p19, IKKß-increased expression favors the appearance of cutaneous spindle cell-like squamous cell carcinomas, which are highly aggressive tumors. These results reveal that IKKß activity prevents skin tumor development, and shed light on the complex nature of IKKß effects on cancer progression, as IKKß can both promote and prevent carcinogenesis depending on the cell type or molecular context.Implications: The ability of IKKß to promote or prevent carcinogenesis suggests the need for further evaluation when targeting this protein. Mol Cancer Res; 15(9); 1255-64. ©2017 AACR.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , I-kappa B Kinase/biosynthesis , Skin Neoplasms/metabolism , Tumor Suppressor Protein p14ARF/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Epigenesis, Genetic , I-kappa B Kinase/genetics , Mice , Mice, Transgenic , Mutation , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Cancer Res ; 77(6): 1357-1368, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28108518

ABSTRACT

RAS genes are mutated in 20% of human tumors, but these mutations are very rare in breast cancer. Here, we used a mouse model to generate tumors upon activation of a mutagenic T2Onc2 transposon via expression of a transposase driven by the keratin K5 promoter in a p53+/- background. These animals mainly developed mammary tumors, most of which had transposon insertions in one of two RASGAP genes, neurofibromin1 (Nf1) and RAS p21 protein activator (Rasa1). Immunohistochemical analysis of a collection of human breast tumors confirmed that low expression of RASA1 is frequent in basal (triple-negative) and estrogen receptor negative tumors. Bioinformatic analysis of human breast tumors in The Cancer Genome Atlas database showed that although RASA1 mutations are rare, allelic loss is frequent, particularly in basal tumors (80%) and in association with TP53 mutation. Inactivation of RASA1 in MCF10A cells resulted in the appearance of a malignant phenotype in the context of mutated p53. Our results suggest that alterations in the Ras pathway due to the loss of negative regulators of RAS may be a common event in basal breast cancer. Cancer Res; 77(6); 1357-68. ©2017 AACR.


Subject(s)
Biomarkers, Tumor/metabolism , DNA Transposable Elements/genetics , Transposases/physiology , Triple Negative Breast Neoplasms/pathology , Tumor Suppressor Protein p53/physiology , p120 GTPase Activating Protein/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Cells, Cultured , Female , Humans , Mice , Mice, Transgenic , Neoplasm Staging , Prognosis , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , p120 GTPase Activating Protein/genetics
10.
Oncotarget ; 7(20): 29531-47, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27121058

ABSTRACT

Nonmelanoma skin cancers (NMSC) are the most common human malignancies. IKKα is an essential protein for skin development and is also involved in the genesis and progression of NMSC, through mechanisms not fully understood. While different studies show that IKKα protects against skin cancer, others indicate that it promotes NMSC. To resolve this controversy we have generated two models of transgenic mice expressing the IKKα protein in the nucleus (N-IKKα mice) or the cytoplasm (C-IKKα mice) of keratinocytes. Chemical skin carcinogenesis experiments show that tumors developed by both types of transgenic mice exhibit histological and molecular characteristics that make them more prone to progression and invasion than those developed by Control mice. However, the mechanisms through which IKKα promotes skin tumors are different depending on its subcellular localization; while IKKα of cytoplasmic localization increases EGFR, MMP-9 and VEGF-A activities in tumors, nuclear IKKα causes tumor progression through regulation of c-Myc, Maspin and Integrin-α6 expression. Additionally, we have found that N-IKKα skin tumors mimic the characteristics associated to aggressive human skin tumors with high risk to metastasize. Our results show that IKKα has different non-overlapping roles in the nucleus or cytoplasm of keratinocytes, and provide new targets for intervention in human NMSC progression.


Subject(s)
Cell Nucleus/enzymology , Cytoplasm/enzymology , I-kappa B Kinase/metabolism , Keratinocytes/pathology , Skin Neoplasms/pathology , Animals , Carcinoma, Basal Cell/enzymology , Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Humans , Keratinocytes/enzymology , Mice , Mice, Transgenic , Skin Neoplasms/enzymology
11.
Oncotarget ; 7(15): 20902-18, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26959115

ABSTRACT

p53 is a protein that causes cell cycle arrest, apoptosis or senescence, being crucial in the process of tumor suppression in several cell types. Different in vitro and animal models have been designed for the study of p53 role in skin cancer. These models have revealed opposing results, as in some experimental settings it appears that p53 protects against skin cancer, but in others, the opposite conclusion emerges. We have generated cohorts of mice with efficient p53 deletion restricted to stratified epithelia and control littermates expressing wild type p53 and studied their sensitivity to both chemically-induced and spontaneous tumoral transformation, as well as the tumor types originated in each experimental group. Our results indicate that the absence of p53 in stratified epithelia leads to the appearance, in two-stage skin carcinogenesis experiments, of a higher number of tumors that grow faster and become malignant more frequently than tumors arisen in mice with wild type p53 genotype. In addition, the histological diversity of the tumor type is greater in mice with epidermal p53 loss, indicating the tumor suppressive role of p53 in different epidermal cell types. Aging mice with p53 inactivation in stratified epithelia developed spontaneous carcinomas in skin and other epithelia. Overall, these results highlight the truly protective nature of p53 functions in the development of cancer in skin and in other stratified epithelia.


Subject(s)
Carcinogenesis/pathology , Carcinoma, Squamous Cell/pathology , Epidermis/pathology , Skin Neoplasms/pathology , Tumor Suppressor Protein p53/physiology , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Apoptosis , Carcinogenesis/metabolism , Carcinogens/toxicity , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/metabolism , Cell Proliferation , Cells, Cultured , Epidermis/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Skin Neoplasms/etiology , Skin Neoplasms/metabolism
12.
ScientificWorldJournal ; 2014: 193803, 2014.
Article in English | MEDLINE | ID: mdl-24523631

ABSTRACT

Inhibition of gene expression through siRNAs is a tool increasingly used for the study of gene function in model systems, including transgenic mice. To achieve perdurable effects, the stable expression of siRNAs by an integrated transgenic construct is necessary. For transgenic siRNA expression, promoters transcribed by either RNApol II or III (such as U6 or H1 promoters) can be used. Relatively large amounts of small RNAs synthesis are achieved when using RNApol III promoters, which can be advantageous in knockdown experiments. To study the feasibility of H1 promoter-driven RNAi-expressing constructs for protein knockdown in transgenic mice, we chose IKK1 as the target gene. Our results indicate that constructs containing the H1 promoter are sensitive to the presence of prokaryotic sequences and to transgene position effects, similar to RNApol II promoters-driven constructs. We observed variable expression levels of transgenic siRNA among different tissues and animals and a reduction of up to 80% in IKK1 expression. Furthermore, IKK1 knockdown led to hair follicle alterations. In summary, we show that constructs directed by the H1 promoter can be used for knockdown of genes of interest in different organs and for the generation of animal models complementary to knockout and overexpression models.


Subject(s)
Gene Knockdown Techniques , I-kappa B Kinase/genetics , Promoter Regions, Genetic , RNA Interference , Animals , Cell Line , Gene Expression Regulation , Gene Order , Gene Silencing , Gene Targeting , Humans , Mice , Mice, Transgenic , Phenotype , RNA, Small Interfering
13.
J Invest Dermatol ; 133(7): 1870-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23426135

ABSTRACT

CYLD is a tumor-suppressor gene mutated in the skin appendage tumors cylindromas, trichoepitheliomas, and spiradenomas. We have performed in vivo metastasis assays in nude mice and found that the loss of the deubiquitinase function of CYLD in squamous cell carcinoma (SCC) cells greatly enhances the lung metastatic capability of these cells. These metastases showed several characteristics that make them distinguishable from those carrying a functional CYLD, such as robust angiogenesis, increased expression of tumor malignancy markers of SCCs, and a decrease in the expression of the suppressor of metastasis Maspin. Restoration of Maspin expression in the epidermal SCC cells defective in CYLD deubiquitination function significantly reduces their ability to form metastases, thereby suggesting that the decrease in the levels of Maspin expression plays an important role in the acquisition of metastatic potential of these cells. In addition, we have characterized Maspin downregulation in cylindromas, trichoepitheliomas, and spiradenomas carrying functional inactivating mutations of CYLD, also providing an evidence of the correlation between impaired CYLD function and Maspin decreased expression in vivo in human tumors.


Subject(s)
Carcinoma, Squamous Cell/secondary , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cysteine Endopeptidases/genetics , Lung Neoplasms/secondary , Mutation/genetics , Skin Neoplasms/pathology , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cysteine Endopeptidases/metabolism , Deubiquitinating Enzyme CYLD , Disease Models, Animal , Keratin-13/genetics , Keratin-13/metabolism , Keratin-8/genetics , Keratin-8/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Nude , Neovascularization, Pathologic/genetics , Serpins/genetics , Serpins/metabolism , Skin/metabolism , Skin/pathology , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism
14.
J Invest Dermatol ; 133(1): 239-48, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22832494

ABSTRACT

Nonmelanoma skin cancer (NMSC) is by far the most frequent type of cancer in humans. NMSC includes several types of malignancies with different clinical outcomes, the most frequent being basal and squamous cell carcinomas. We have used the Sleeping Beauty transposon/transposase system to identify somatic mutations associated with NMSC. Transgenic mice bearing multiple copies of a mutagenic Sleeping Beauty transposon T2Onc2 and expressing the SB11 transposase under the transcriptional control of regulatory elements from the keratin K5 promoter were treated with TPA, either in wild-type or Ha-ras mutated backgrounds. After several weeks of treatment, mice with transposition developed more malignant tumors with decreased latency compared with control mice. Transposon/transposase animals also developed basal cell carcinomas. Genetic analysis of the transposon integration sites in the tumors identified several genes recurrently mutated in different tumor samples, which may represent novel candidate cancer genes. We observed alterations in the expression levels of some of these genes in human tumors. Our results show that inactivating mutations in Notch1 and Nsd1, among others, may have an important role in skin carcinogenesis.


Subject(s)
Carcinoma, Basal Cell/genetics , Carcinoma, Squamous Cell/genetics , Cell Transformation, Neoplastic/genetics , DNA Transposable Elements/genetics , Mutation , Skin Neoplasms/genetics , Animals , Carcinogens/toxicity , Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/pathology , Carrier Proteins/genetics , DNA Mutational Analysis/methods , Genes, ras/genetics , Histone Methyltransferases , Histone-Lysine N-Methyltransferase , Humans , Intracellular Signaling Peptides and Proteins/genetics , Keratin-15 , Keratin-5/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Nuclear Proteins/genetics , Promoter Regions, Genetic , Receptor, Notch1/genetics , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate/toxicity , Transposases/genetics
15.
Mol Cancer Res ; 9(10): 1329-38, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21821676

ABSTRACT

Alterations in nuclear factor kappaB (NFκB) signaling have been related with several diseases and importantly also with cancer. Different animal models with increased or diminished NFκB signaling have shown that NFκB subunits and their regulators are relevant to the pathophysiology of different organs and tissues. In particular, both the deletion of the regulatory subunit ß of the kinase of the inhibitor of NFκB (IKKß) and its overexpression in epidermis lead to the development of skin inflammatory diseases not associated with tumoral lesions. In this work, we have studied the consequences of IKKß overexpression in other organs and tissues. We found that elevated IKKß levels led to altered development and functionality of exocrine glands (i.e., mammary glands) in transgenic female mice. In oral epithelia, increased IKKß expression produced lichenoid inflammation with abundant granulocytes, macrophages, and B cells, among other inflammatory cells. This inflammatory phenotype was associated with high incidence of tumoral lesions in oral epithelia, contrary to what was found in skin. Moreover, IKKß also increased the malignant progression of both spontaneous and experimentally induced oral tumors. These results highlight the importance of IKKß in epithelial and glandular homeostasis as well as in oral tumorigenesis and open the possibility that IKKß activity might be implicated in the development of oral cancer in humans.


Subject(s)
Cell Transformation, Neoplastic/metabolism , I-kappa B Kinase/biosynthesis , Mouth Mucosa/pathology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Immunohistochemistry , Male , Mice , Mice, Transgenic , Mouth Mucosa/metabolism , Phenotype , Signal Transduction , Transfection
16.
PLoS One ; 6(7): e21984, 2011.
Article in English | MEDLINE | ID: mdl-21755017

ABSTRACT

Non-melanoma skin cancer is the most frequent type of cancer in humans. In this study we demonstrate that elevated IKKα expression in murine epidermis increases the malignancy potential of skin tumors. We describe the generation of transgenic mice overexpressing IKKα in the basal, proliferative layer of the epidermis and in the outer root sheath of hair follicles. The epidermis of K5-IKKα transgenic animals shows several alterations such as hyperproliferation, mislocalized expression of integrin-α6 and downregulation of the tumor suppressor maspin. Treatment of the back skin of mice with the mitogenic agent 12-O-tetradecanoylphorbol-13-acetate causes in transgenic mice the appearance of different preneoplastic changes such as epidermal atypia with loss of cell polarity and altered epidermal tissue architecture, while in wild type littermates this treatment only leads to the development of benign epidermal hyperplasia. Moreover, in skin carcinogenesis assays, transgenic mice carrying active Ha-ras (K5-IKKα-Tg.AC mice) develop invasive tumors, instead of the benign papillomas arising in wild type-Tg-AC mice also bearing an active Ha-ras. Therefore we provide evidence for a tumor promoter role of IKKα in skin cancer, similarly to what occurs in other neoplasias, including hepatocarcinomas and breast, prostate and colorectal cancer. The altered expression of cyclin D1, maspin and integrin-α6 in skin of transgenic mice provides, at least in part, the molecular bases for the increased malignant potential found in the K5-IKKα skin tumors.


Subject(s)
Epidermis/enzymology , Epidermis/pathology , I-kappa B Kinase/metabolism , Skin Neoplasms/pathology , Animals , Biomarkers, Tumor/metabolism , Cattle , Cell Proliferation/drug effects , Epidermis/drug effects , Immunohistochemistry , Integrin alpha6/metabolism , Keratinocytes/drug effects , Keratinocytes/enzymology , Keratinocytes/pathology , Mice , Mice, Transgenic , NF-kappa B/metabolism , Rabbits , Serpins/metabolism , Signal Transduction/drug effects , Skin Neoplasms/enzymology , Tetradecanoylphorbol Acetate/pharmacology
17.
J Invest Dermatol ; 130(6): 1598-610, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20200541

ABSTRACT

IKKbeta is a subunit of the IkappaB kinase (IKK) complex required for NF-kappaB activation in response to pro-inflammatory signals. NF-kappaB regulates the expression of many genes involved in inflammation, immunity, and apoptosis, and also controls cell proliferation and differentiation in different tissues; however, its function in skin physiopathology remains controversial. In this study we report the alterations caused by increased IKKbeta activity in skin basal cells of transgenic mice. These animals suffered chronic inflammation with abundant macrophages and other CD45(+) infiltrating cells in the skin, which resulted in epidermal basal cell injury and degeneration of hair follicles. They showed histological features characteristic of interface dermatitis (ID). This phenotype is accompanied by an increased production of inflammatory cytokines by transgenic keratinocytes. Accordingly, transcriptome studies show upregulation of genes associated with inflammatory responses. The inflammatory phenotype observed as a consequence of IKKbeta overexpression is independent of T and B lymphocytes, as it also arises in mice lacking these cell types. In summary, our data indicate the importance of IKKbeta in the development of ID and in the homeostasis of stratified epithelia. Our results also support the idea that IKKbeta might be a valid therapeutic target for the treatment of skin inflammatory diseases.


Subject(s)
Dermatitis/metabolism , Dermatitis/physiopathology , I-kappa B Kinase/metabolism , Immune System/immunology , Immune System/physiopathology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Disease Models, Animal , Mice , Mice, Transgenic , NF-kappa B/metabolism , Phenotype , Signal Transduction/physiology , Skin/immunology , Skin/metabolism , Skin/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
18.
J Cell Sci ; 122(Pt 14): 2491-503, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19549684

ABSTRACT

Atypical PKC (PKC iota) is a key organizer of cellular asymmetry. Sequential extractions of intestinal cells showed a pool of enzymatically active PKC iota and the chaperone Hsp70.1 attached to the apical cytoskeleton. Pull-down experiments using purified and recombinant proteins showed a complex of Hsp70 and atypical PKC on filamentous keratins. Transgenic animals overexpressing keratin 8 displayed delocalization of Hsp70 and atypical PKC. Two different keratin-null mouse models, as well as keratin-8 knockdown cells in tissue culture, also showed redistribution of Hsp70 and a sharp decrease in the active form of atypical PKC, which was also reduced by Hsp70 knockdown. An in-vitro turn motif rephosphorylation assay indicated that PKC iota is dephosphorylated by prolonged activity. The Triton-soluble fraction could rephosphorylate PKC iota only when supplemented with the cytoskeletal pellet or filamentous highly purified keratins, a function abolished by immunodepletion of Hsp70 but rescued by recombinant Hsp70. We conclude that both filamentous keratins and Hsp70 are required for the rescue rephosphorylation of mature atypical PKC, regulating the subcellular distribution and steady-state levels of active PKC iota.


Subject(s)
Enterocytes/enzymology , HSP70 Heat-Shock Proteins/metabolism , Intermediate Filaments/enzymology , Isoenzymes/metabolism , Keratins/metabolism , Protein Kinase C/metabolism , Animals , Caco-2 Cells , HSP70 Heat-Shock Proteins/genetics , Humans , Isoenzymes/genetics , Keratin-18/metabolism , Keratin-19/metabolism , Keratin-8/metabolism , Keratins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphorylation , Protein Kinase C/genetics , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference
19.
Mol Biol Cell ; 16(9): 4096-107, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15987737

ABSTRACT

Ezrin connects the apical F-actin scaffold to membrane proteins in the apical brush border of intestinal epithelial cells. Yet, the mechanisms that recruit ezrin to the apical domain remain obscure. Using stable CACO-2 transfectants expressing keratin 8 (K8) antisense RNA under a tetracycline-responsive element, we showed that the actin-ezrin scaffold cannot assemble in the absence of intermediate filaments (IFs). Overexpression of ezrin partially rescued this phenotype. Overexpression of K8 in mice also disrupted the assembly of the brush border, but ezrin distributed away from the apical membrane in spots along supernumerary IFs. In cytochalasin D-treated cells ezrin localized to a subapical compartment and coimmunoprecipitated with IFs. Overexpression of ezrin in undifferentiated cells showed a Triton-insoluble ezrin compartment negative for phospho-T567 (dormant) ezrin visualized as spots along IFs. Pulse-chase analysis showed that Triton-insoluble, newly synthesized ezrin transiently coimmunoprecipitates with IFs during the first 30 min of the chase. Dormant, but not active (p-T567), ezrin bound in vitro to isolated denatured keratins in Far-Western analysis and to native IFs in pull-down assays. We conclude that a transient association to IFs is an early step in the polarized assembly of apical ezrin in intestinal epithelial cells.


Subject(s)
Epithelial Cells/metabolism , Intermediate Filaments/metabolism , Intestinal Mucosa/metabolism , Phosphoproteins/metabolism , Animals , Caco-2 Cells , Cell Differentiation/physiology , Cell Line , Cytoskeletal Proteins , Enterocytes/cytology , Enterocytes/metabolism , Humans , Intestinal Mucosa/cytology , Keratin-8 , Keratins/biosynthesis , Keratins/genetics , Mice , Mice, Transgenic , Octoxynol
20.
Cancer Res ; 64(16): 5617-23, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15313899

ABSTRACT

Cannabinoids inhibit tumor angiogenesis in mice, but the mechanism of their antiangiogenic action is still unknown. Because the vascular endothelial growth factor (VEGF) pathway plays a critical role in tumor angiogenesis, here we studied whether cannabinoids affect it. As a first approach, cDNA array analysis showed that cannabinoid administration to mice bearing s.c. gliomas lowered the expression of various VEGF pathway-related genes. The use of other methods (ELISA, Western blotting, and confocal microscopy) provided additional evidence that cannabinoids depressed the VEGF pathway by decreasing the production of VEGF and the activation of VEGF receptor (VEGFR)-2, the most prominent VEGF receptor, in cultured glioma cells and in mouse gliomas. Cannabinoid-induced inhibition of VEGF production and VEGFR-2 activation was abrogated both in vitro and in vivo by pharmacological blockade of ceramide biosynthesis. These changes in the VEGF pathway were paralleled by changes in tumor size. Moreover, intratumoral administration of the cannabinoid Delta9-tetrahydrocannabinol to two patients with glioblastoma multiforme (grade IV astrocytoma) decreased VEGF levels and VEGFR-2 activation in the tumors. Because blockade of the VEGF pathway constitutes one of the most promising antitumoral approaches currently available, the present findings provide a novel pharmacological target for cannabinoid-based therapies.


Subject(s)
Cannabinoids/pharmacology , Glioma/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Astrocytoma/blood supply , Astrocytoma/drug therapy , Astrocytoma/genetics , Astrocytoma/metabolism , Cell Line, Tumor , Ceramides/metabolism , Gene Expression Profiling , Glioblastoma/blood supply , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Glioma/blood supply , Glioma/drug therapy , Glioma/genetics , Humans , Mice , Neovascularization, Pathologic/drug therapy , Second Messenger Systems , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...