Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Life Sci Alliance ; 6(4)2023 04.
Article in English | MEDLINE | ID: mdl-36732018

ABSTRACT

The initial dissemination of cancer cells from many primary tumors implies intravasation to lymphatic nodes or blood vessels. To investigate the mechanisms involved, we analyzed the expression of small non-coding RNAs in cutaneous squamous cell carcinoma (cSCC), a prevalent tumor that mainly spreads to lymph nodes. We report the reduced expression of small nucleolar RNAs in primary cSCCs that metastasized when compared to non-metastasizing cSCCs, and the progressive loss of DKC1 (dyskerin, which stabilizes the small nucleolar RNAs) along the metastasis. DKC1 depletion in cSCC cells triggered lipid metabolism by altering the mevalonate pathway and the acquisition of metastatic traits. Treatment of DKC1-depleted cells with simvastatin, an inhibitor of the mevalonate pathway, blocked the expression of proteins involved in the epithelial-to-mesenchymal transition. Consistently, the expression of the enzyme 3-hydroxy-3-methylglutaryl-CoA synthase 1 was associated with pathological features of high metastatic risk in cSCC patients. Our data underpin the relevance of the mevalonate metabolism in metastatic dissemination and pave the possible incorporation of therapeutic approaches among the antineoplastic drugs used in routine patient care.


Subject(s)
Carcinoma, Squamous Cell , Skin Neoplasms , Humans , Carcinoma, Squamous Cell/metabolism , Skin Neoplasms/pathology , Mevalonic Acid , Phenotype , Simvastatin/pharmacology , Nuclear Proteins , Cell Cycle Proteins
2.
Sci Adv ; 6(43)2020 10.
Article in English | MEDLINE | ID: mdl-33097530

ABSTRACT

Ewing sarcoma (EwS) is an aggressive tumor that affects adolescents and young adults. EwS is defined by a chromosomal translocation, EWSR1-FLI1 being the most common, that causes genome reprogramming through remodeling of enhancers. Here, we describe an unexpected function of RING1B, which is highly expressed in EwS. While retaining its repressive activity at Polycomb developmental regulated genes, RING1B colocalizes with EWSR1-FLI1 at active enhancers. We demonstrate that RING1B is necessary for the expression of key EWSR1-FLI1 targets by facilitating oncogene recruitment to their enhancers. Knockdown of RING1B impairs growth of tumor xenografts and expression of genes regulated by EWSR1-FLI1 bound enhancers. Pharmacological inhibition of AURKB with AZD1152 increases H2Aub levels causing down-regulation of RING1B/EWSR1-FLI1 common targets. Our findings demonstrate that RING1B is a critical modulator of EWSR1-FLI1-induced chromatin remodeling, and its inhibition is a potential therapeutic strategy for the treatment of these tumors.


Subject(s)
Chromatin , Sarcoma, Ewing , Adolescent , Carcinogenesis , Cell Line, Tumor , Cell Transformation, Neoplastic , Chromatin/genetics , Chromatin Assembly and Disassembly , Gene Expression Regulation, Neoplastic , Humans , Oncogene Proteins, Fusion/genetics , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/drug therapy , Sarcoma, Ewing/genetics , Young Adult
3.
PLoS One ; 15(5): e0232146, 2020.
Article in English | MEDLINE | ID: mdl-32413042

ABSTRACT

Actinic keratosis is a common skin disease that may progress to invasive squamous cell carcinoma if left untreated. Ingenol mebutate has demonstrated efficacy in field treatment of actinic keratosis. However, molecular mechanisms on ingenol mebutate response are not yet fully understood. In this study, we evaluated the gene expression profiles of actinic keratosis lesions before and after treatment with ingenol mebutate using microarray technology. Actinic keratoses on face/scalp of 15 immunocompetent patients were identified and evaluated after treatment with topical ingenol mebutate gel 0.015%, applied once daily for 3 consecutive days. Diagnostic and clearance of lesions was determined by clinical, dermoscopic, and reflectance confocal microscopy criteria. Lesional and non-lesional skin biopsies were subjected to gene expression analysis profiled by Affymetrix microarray. Differentially expressed genes were identified, and enrichment analyses were performed using STRING database. At 8 weeks post-treatment, 60% of patients responded to ingenol mebutate therapy, achieving complete clearance in 40% of cases. A total of 128 differentially expressed genes were identified following treatment, and downregulated genes (114 of 128) revealed changes in pathways important to epidermal development, keratinocyte differentiation and cornification. In responder patients, 388 downregulated genes (of 450 differentially expressed genes) were also involved in development/differentiation of the epidermis, and immune system-related pathways, such as cytokine and interleukin signaling. Cluster analysis revealed two relevant clusters showing upregulated profile patterns in pre-treatment actinic keratoses of responders, as compared to non-responders. Again, differentially expressed genes were mainly associated with cornification, keratinization and keratinocyte differentiation. Overall, the present study provides insight into the gene expression profile of actinic keratoses after treatment with ingenol mebutate, as well as identification of genetic signatures that could predict treatment response.


Subject(s)
Diterpenes/pharmacology , Gene Expression Regulation/drug effects , Keratosis, Actinic/genetics , Aged , Aged, 80 and over , Diterpenes/therapeutic use , Gels , Humans , Male , Oligonucleotide Array Sequence Analysis
4.
J Med Genet ; 57(9): 601-604, 2020 09.
Article in English | MEDLINE | ID: mdl-31937562

ABSTRACT

Background: Keratinocytic epidermal naevi (KENs) are congenital benign skin mosaic lesions that share common mutations with some subsets of urothelial carcinomas. Moreover, several patients with extensive KEN who also developed urothelial carcinomas at young ages have been reported. Thus, patients with extensive KEN may harbour mosaic urothelial oncogenic mutations that would favour the early development of urothelial carcinomas. Methods: We selected five patients with extensive KEN involving the lower part of the back and performed a molecular characterisation of urothelial and cutaneous samples using a next-generation sequencing (NGS) custom panel targeting candidate oncogenic genes. Results: Mosaic pathogenic mutations were detected in KEN in all patients. In four out of five patients, mosaic pathogenic mutations in FGFR2 or HRAS were also detected in samples from the urothelial tract. Moreover, we report a patient who developed urothelial carcinomas at age 29 and harboured an HRAS G12S mutation both in skin and urothelial tumour samples. Conclusions: We conclude that patients with extensive KEN involving the lower part of the back frequently harbour oncogenic mutations in the urothelium that may induce the development of carcinomas. NGS panels can be considered as highly sensitive tools to identify this subgroup of patients, which might permit adoption of screening measures to detect malignant transformation at early stages.


Subject(s)
Nevus/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Urothelium/metabolism , Adult , Carcinogenesis/genetics , Female , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Male , Middle Aged , Mutation/genetics , Nevus/complications , Nevus/pathology , Skin Neoplasms/complications , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Urothelium/pathology , Young Adult
6.
Genes Chromosomes Cancer ; 58(3): 164-174, 2019 03.
Article in English | MEDLINE | ID: mdl-30474248

ABSTRACT

Although most cutaneous squamous cell carcinomas (cSCCs) develop from actinic keratoses (AKs), the key events in this evolution remain unclear. We have combined the results of different genomic and expression array platforms on matched concomitant samples of sun-exposed skin (SES), AK, and cSCC from 10 immunocompetent patients. Gene expression analysis and copy number alterations were assessed using GeneChip Human Gene 2.0 ST Array (Affymetrix, Santa Clara, CA) and CytoScan HD Cytogenetics Solution (Affymetrix) platforms, respectively. Integration of transcriptome and genome results was evaluated using the DR-Integrator tool. Additional studies (qPCR, immunohistochemistry, and Western blot) were performed for selected genes. FOSL1 and BNC1 encode transcription factors whose expression was increased in cSCC in the expression array and the qPCR. By immunohistochemistry, FOSL1 showed an intense staining at the invasive front of cSCC samples and BNC1 expression varied from a nuclear (SES) to a cytoplasmic location (cSCC). Western blot analyses confirmed the enhancement of FOSL1 and BNC1. In addition, the smallest overlapping regions (SORIs) of genomic imbalance involving at least three of the samples were selected. One of the SORIs was a deletion in the p24.1 band of chromosome 3, shared by seven of the cSCCs. A strong correlation in the integration analysis was found for NEK10, a gene contained in the previously mentioned SORI. Loss of NEK10 expression in cSCC was confirmed by immunohistochemistry and Western blot analyses. In addition, functional studies in NEK10 depleted cells were performed. In conclusion, we identified FOSL1 and BNC1, which could act as tumor drivers, and NEK10, which could function as a tumor suppressor, to be differentially expressed during cSCC development.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Skin Neoplasms/genetics , Transcriptome , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Humans , Male , NIMA-Related Kinases/genetics , NIMA-Related Kinases/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Skin Neoplasms/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Acta Derm Venereol ; 99(1): 89-94, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30176040

ABSTRACT

Although desmoplasia has been associated with poor prognoses in cutaneous squamous cell carcinoma, little attention has been paid to the patterns of fibrosis. This study aimed to examine the different stromal fibrotic patterns as markers of metastatic risk. We performed a multicenter retrospective study that included 102 cutaneous squamous cell carcinomas (52 non-metastatic and 50 metastatic carcinomas). Clinical and histopathological data were registered. The fibrotic reaction pattern was classified as mature, intermediate or immature depending on the presence of keloid-like collagen and myxoid stroma. The immature pattern (areas characterized by myxoid changes with no inflammation) was observed in 18 samples and its presence was significantly associated with immunosuppression, budding, desmoplasia, perineural invasion, anatomic level, tumoural depth and metastatic risk in the multivariate analysis. Our findings suggest that the presence of an immature myxoid fibrotic pattern, which can be easily identified by routine hematoxylin-eosin staining, is strongly associated with metastatic risk.


Subject(s)
Carcinoma, Squamous Cell/secondary , Skin Neoplasms/pathology , Stromal Cells/pathology , Aged , Aged, 80 and over , Biopsy , Coloring Agents , Eosine Yellowish-(YS) , Female , Fibrosis , Hematoxylin , Humans , Male , Phenotype , Prognosis , Retrospective Studies , Risk Assessment , Risk Factors , Spain , Staining and Labeling/methods , Tumor Microenvironment
8.
Am J Dermatopathol ; 40(9): 647-654, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29742559

ABSTRACT

Programmed cell death ligand 1 (PD-L1) expression by tumor cells plays an important role in the inhibition of T cell-mediated immune response in cancer. PD-L1 expression by tumor cells has been linked to poor prognosis in a wide variety of cancers. However, PD-L1 expression in cutaneous squamous cell carcinoma (cSCC) has been scarcely studied, and its role as a prognosis biomarker remains controversial. The association of PD-L1 expression and the metastatic risk in a series of cSCC was assessed. PD-L1 and CD8 immunostainings of full excision sections of 99 primary tumors and 24 lymphatic metastases were semiquantitatively evaluated. Primary cSCCs were grouped according to the development of lymphatic metastatic spread [metastasizing squamous cell carcinoma (MSCC)] (n = 48) or the absence of progression [nonmetastasizing squamous cell carcinoma (NMSCC)] (n = 51). PD-L1-positive expression (cut off ≥1%) was found in 26% NMSCCs and in 50% MSCCs (P = 0.02). PD-L1 association with an increased metastatic risk was confirmed in the multivariate analysis (P < 0.05), along with the following features: recurrence, poor differentiation, and perineural invasion. Ninety percent of the metastases of PD-L1-positive tumors were also positive for PD-L1, displaying a trend toward a higher PD-L1 expression when compared with their primary tumors (P = 0.058). No significant differences in the peritumoral inflammatory infiltrate or in the expression of CD8 were found between metastasizing and nonmetastasizing primary tumors. Our results suggest that PD-L1 may play a relevant role in metastatic spread and may be a candidate prognostic biomarker in cSCC.


Subject(s)
B7-H1 Antigen/analysis , Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/secondary , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Aged , Aged, 80 and over , Biopsy , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation , Chi-Square Distribution , Female , Humans , Immunohistochemistry , Logistic Models , Lymphatic Metastasis , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Male , Multivariate Analysis , Neoplasm Invasiveness , Retrospective Studies , Risk Factors , Spain , Up-Regulation
9.
Carcinogenesis ; 39(3): 503-513, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29394319

ABSTRACT

Cutaneous squamous cell carcinoma (cSCC) is the second most common malignancy in humans and approximately 5% metastasize, usually to regional lymph nodes. Epigenetic regulation of gene expression may allow tumoral cells to acquire new functions in order to escape from the primary tumor. The aim of this study was to investigate the expression and function of proteins of the Polycomb family of epigenetic regulators in the metastatic process of cSCC. A higher expression of RING1B and EZH2 was detected by immunohistochemistry in a series of primary cSCC tumors that metastasized (MSCCs) when compared with non-metastasizing cSCCs (non-MSCCs). Stable downregulation of RING1B and EZH2 in cSCC cells results in enhanced expression of inflammatory cytokines and activation of the NF-κB signaling pathway. Accordingly, non-MSCCs display higher levels of membranous pS176-inhibitor of NF-kB kinase, and their stroma is enriched in neutrophils and eosinophils when compared with MSCCs. In vitro, hematopoietic cells exhibit a substantial migratory response to supernatants from Polycomb-depleted cSCC cells. Altogether, these data indicate that RING1B and EZH2 repress the innate inflammatory cSCC function and impair tumor immunosurveillance and suggest that patients with high-risk cSCCs could benefit from clinical therapies addressed to harness the immune response.


Subject(s)
Carcinoma, Squamous Cell/immunology , Enhancer of Zeste Homolog 2 Protein/immunology , Polycomb Repressive Complex 1/immunology , Skin Neoplasms/immunology , Tumor Escape/immunology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic/immunology , Female , Humans , Immunologic Surveillance/immunology , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Male , Neoplasm Invasiveness/immunology , Neoplasm Invasiveness/pathology , Polycomb Repressive Complex 1/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
10.
Mol Cancer ; 15(1): 53, 2016 07 25.
Article in English | MEDLINE | ID: mdl-27457246

ABSTRACT

BACKGROUND: Cutaneous squamous cell carcinoma (cSCC) is the second most common skin cancer and frequently progresses from an actinic keratosis (AK), a sun-induced keratinocyte intraepithelial neoplasia (KIN). Epigenetic mechanisms involved in the phenomenon of progression from AK to cSCC remain to be elicited. METHODS: Expression of microRNAs in sun-exposed skin, AK and cSCC was analysed by Agilent microarrays. DNA methylation of miR-204 promoter was determined by bisulphite treatment and pyrosequencing. Identification of miR-204 targets and pathways was accomplished in HaCat cells. Immunofluorescence and immunohistochemistry were used to analyze STAT3 activation and PTPN11 expression in human biopsies. RESULTS: cSCCs display a marked downregulation of miR-204 expression when compared to AK. DNA methylation of miR-204 promoter was identified as one of the repressive mechanisms that accounts for miR-204 silencing in cSCC. In HaCaT cells miR-204 inhibits STAT3 and favours the MAPK signaling pathway, likely acting through PTPN11, a nuclear tyrosine phosphatase that is a direct miR-204 target. In non-peritumoral AK lesions, activated STAT3, as detected by pY705-STAT3 immunofluorescence, is retained in the membrane and cytoplasm compartments, whereas AK lesions adjacent to cSCCs display activated STAT3 in the nuclei. CONCLUSIONS: Our data suggest that miR-204 may act as a "rheostat" that controls the signalling towards the MAPK pathway or the STAT3 pathway in the progression from AK to cSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Gene Expression Profiling/methods , Keratosis, Actinic/genetics , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis/methods , Skin Neoplasms/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , DNA Methylation , Disease Progression , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Keratosis, Actinic/metabolism , Promoter Regions, Genetic , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Sequence Analysis, DNA , Skin Neoplasms/metabolism
11.
Oncotarget ; 7(29): 46283-46300, 2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27317769

ABSTRACT

Ewing sarcoma (ES) is an aggressive tumor defined by EWSR1 gene fusions that behave as an oncogene. Here we demonstrate that RING1B is highly expressed in primary ES tumors, and its expression is independent of the fusion oncogene. RING1B-depleted ES cells display an expression profile enriched in genes functionally involved in hematological development but RING1B depletion does not induce cellular differentiation. In ES cells, RING1B directly binds the SCN8A sodium channel promoter and its depletion results in enhanced Nav1.6 expression and function. The signaling pathway most significantly modulated by RING1B is NF-κB. RING1B depletion results in enhanced p105/p50 expression, which sensitizes ES cells to apoptosis by FGFR/SHP2/STAT3 blockade. Reduced NaV1.6 function protects ES cells from apoptotic cell death by maintaining low NF-κB levels. Our findings identify RING1B as a trait of the cell-of-origin and provide a potential targetable vulnerability.


Subject(s)
Bone Neoplasms/metabolism , NAV1.6 Voltage-Gated Sodium Channel/metabolism , NF-kappa B/metabolism , Polycomb Repressive Complex 1/metabolism , Sarcoma, Ewing/metabolism , Signal Transduction , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Humans , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Signal Transduction/physiology , Tumor Cells, Cultured
12.
J Urol ; 193(2): 699-705, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25063494

ABSTRACT

PURPOSE: Epithelial-to-mesenchymal transition is a phenomenon in epithelial tumors that involves loss of intercellular adhesion, mesenchymal phenotype acquisition and enhanced migratory potential. While the epithelial-to-mesenchymal transition process has been extensively linked to metastatic progression of squamous cell carcinoma, studies of the role of epithelial-to-mesenchymal transition in squamous cell carcinoma containing high risk human papillomaviruses are scarce. Moreover, to our knowledge epithelial-to-mesenchymal transition involvement in human penile squamous cell carcinoma, which can arise through transforming HPV infections or independently of HPV, has not been investigated. We evaluated the presence of epithelial-to-mesenchymal transition markers and their relationship to HPV in penile squamous cell carcinoma. MATERIALS AND METHODS: We assessed the expression of E-cadherin, vimentin and the epithelial-to-mesenchymal transition related transcription factors Twist, Zeb1 and Snail by immunohistochemical staining in 64 penile squamous cell carcinoma cases. HPV was detected by polymerase chain reaction amplification. RESULTS: Simultaneous loss of membranous E-cadherin expression and vimentin over expression were noted in 43.5% of penile squamous cell carcinoma cases. HPV was significantly associated with loss of membranous E-cadherin but not with epithelial-to-mesenchymal transition. Recurrence and mortality rates were significantly higher in cases showing epithelial-to-mesenchymal transition. CONCLUSIONS: Our findings indicate that in penile squamous cell carcinoma epithelial-to-mesenchymal transition is associated with poor prognosis but not with the presence of HPV.


Subject(s)
Carcinoma, Squamous Cell/pathology , Epithelial-Mesenchymal Transition , Penile Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/virology , Humans , Male , Middle Aged , Papillomavirus Infections/complications , Penile Neoplasms/virology
13.
Oncotarget ; 5(8): 2065-76, 2014 Apr 30.
Article in English | MEDLINE | ID: mdl-24742605

ABSTRACT

In early stages of metastasis malignant cells must acquire phenotypic changes to enhance their migratory behavior and their ability to breach the matrix surrounding tumors and blood vessel walls. Epigenetic regulation of gene expression allows the acquisition of these features that, once tumoral cells have escape from the primary tumor, can be reverted. Here we report that the expression of the Polycomb epigenetic repressor Ring1B is enhanced in tumoral cells that invade the stroma in human ductal breast carcinoma and its expression is coincident with that of Fak in these tumors. Ring1B knockdown in breast cancer cell lines revealed that Ring1B is required to sustain Fak expression in basal conditions as well as in Tgfß-treated cells. Functionally, endogenous Ring1B is required for cell migration and invasion in vitro and for in vivo invasion of the mammary fat pad by tumoral cells. Finally we identify p63 as a target of Ring1B to regulate Fak expression: Ring1B depletion results in enhanced p63 expression, which in turns represses Fak expression. Importantly, Fak downregulation upon Ring1B depletion is dependent on p63 expression. Our findings provide new insights in the biology of the breast carcinoma and open new avenues for breast cancer prognosis and therapy.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Focal Adhesion Kinase 1/metabolism , Membrane Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Animals , Blotting, Western , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Cells, Cultured , Chromatin Immunoprecipitation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Immunohistochemistry , Mice , Mice, Nude , Neoplasm Invasiveness , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis
14.
J Dermatol Sci ; 72(2): 93-102, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23928229

ABSTRACT

BACKGROUND: Cutaneous squamous cell carcinoma (cSCC) is the second most common malignancy in humans and approximately 5% metastasize, usually to regional lymph nodes. Epithelial to mesenchymal transition (EMT) is a process involving loss of intercellular adhesion, acquisition of a mesenchymal phenotype and enhanced migratory potential; epithelial markers, such as E-cadherin, are down-regulated and mesenchymal proteins (Vimentin), increased. OBJECTIVE: To investigate the expression of EMT markers in metastatic SCC (MSCC) and their corresponding metastases, and to correlate them with clinico-pathological factors associated with an increased risk of metastasis. METHODS: We performed a retrospective study that included 146 cSCC samples (51 primary non-metastatic, 56 primary metastatic, 39 lymphatic metastases). Immunohistochemistry for E-cadherin, Vimentin, Snail, beta-catenin, Twist, Zeb1 and Podoplanin was performed. RESULTS: Loss of membranous E-cadherin was observed in 77% cSCCs, with no differences between MSCC and non-MSCC. Among the transcriptional factors controlling EMT, no significant Snail1 expression was detected. Twist, Zeb1, Vimentin, beta-catenin and Podoplanin were significantly overexpressed in MSCCs. Twist ectopic expression in SCC13 cells induced Zeb1, Vimentin and Podoplanin expression and E-cadherin delocalization. These changes resulted in a scattered migration pattern in vitro. Expression of EMT markers was decreased in the metastases when compared with the corresponding primary tumors. CONCLUSION: These results suggest that a partial EMT, characterized by the expression of Twist but without a total E-cadherin depletion, is involved in the acquisition of invasive traits by cSCC, but the process is downregulated in lymph node metastases.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Lymphatic Metastasis , Skin Neoplasms/metabolism , Antigens, CD , Cadherins/metabolism , Down-Regulation , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Membrane Glycoproteins/metabolism , Nuclear Proteins/metabolism , Phenotype , Retrospective Studies , Risk , Snail Family Transcription Factors , Transcription Factors/metabolism , Twist-Related Protein 1/metabolism , Vimentin/metabolism , Zinc Finger E-box-Binding Homeobox 1 , beta Catenin/metabolism
15.
J Gastrointest Cancer ; 42(2): 76-84, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21279552

ABSTRACT

INTRODUCTION: Deregulated expression/activation of transcription factors is a key event in the establishment and progression of human cancer. Furthermore, most oncogenic signaling pathways converge on sets of transcription factors that ultimately control gene expression patterns resulting in cancer development, progression, and metastasis. METHODS: Ductal pancreatic adenocarcinoma (PDA) is the main type of pancreatic cancer and the fourth leading cause of cancer mortality in the Western world. The early stage of the disease is characterized by pancreatic intraepithelial neoplasia lesions bearing mutations in the K-RAS proto-oncogene, which progress to malignant PDA by accumulating additional mutations in the tumor suppressor gene CDKN2A (p16) and in SMAD4 and TP53 transcription factors. The involvement of other signaling pathways in PDA development and progression is an active area of research which may help to clarify the critical steps of this devastating disease. RESULTS: In this regard, several in vitro and in vivo data have demonstrated the contribution of the transcription factor c-Myc to pancreatic carcinogenesis although the molecular mechanisms are poorly understood. c-Myc is a proto-oncogene which has a pivotal function in growth control, differentiation and apoptosis and is known to act as a downstream transcriptional effector of many signaling pathways involved in these processes. It is regulated at multiple levels and its abnormal expression contributes to the genesis of many human tumors. CONCLUSIONS: This review focuses on the role of c-Myc in pancreatic embryonic development and homeostasis as well as its involvement on pancreatic tumorigenesis. Evidences showing that c-Myc function is highly dose and cell context dependent, together with its recently demonstrated ability to reprogram somatic cells towards a pluripotent stem cell-like state, indicate that the role of c-Myc in pancreas pathophysiology might have been previously underscored.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Genes, myc/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction/genetics , Animals , Carcinoma, Pancreatic Ductal/metabolism , Humans , Pancreatic Neoplasms/metabolism , Proto-Oncogene Mas
16.
Biomol Concepts ; 1(3-4): 225-38, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-25961999

ABSTRACT

In multicellular organisms differentiated cells must maintain their cellular memory, which will be faithfully inherited and maintained by their progeny. In addition, these specialized cells are exposed to specific environmental and cell-intrinsic signals and will have to appropriately respond to them. Some of these stimuli lead to changes in a subset of genes or to a genome-wide reprogramming of the cells that will remain after stimuli removal and, in some instances, will be inherited by the daughter cells. The molecular substrate that integrates cellular memory and plasticity is the chromatin, a complex of DNA and histones unique to eukaryotes. The nucleosome is the fundamental unit of the chromatin and nucleosomal organization defines different chromatin conformations. Chromatin regulators affect chromatin conformation and accessibility by covalently modifying the DNA or the histones, substituting histone variants, remodeling the nucleosome position or modulating chromatin looping and folding. These regulators frequently act in multiprotein complexes and highly specific interplays among chromatin marks and different chromatin regulators allow a remarkable array of possibilities. Therefore, chromatin regulator nets act to propagate the conformation of different chromatin regions through DNA replication and mitosis, and to remodel the chromatin fiber to regulate the accessibility of the DNA to transcription factors and to the transcription and repair machineries. Here, the state-of-the-art of the best-known chromatin regulators is reviewed.

17.
J Pathol ; 219(2): 205-13, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19585519

ABSTRACT

Chronic pancreatitis and pancreatic ductal adenocarcinoma (PDAC) are associated with major changes in cell differentiation. These changes may be at the basis of the increased risk for PDAC among patients with chronic pancreatitis. Polycomb proteins are epigenetic silencers expressed in adult stem cells; up-regulation of Polycomb proteins has been reported to occur in a variety of solid tumours such as colon and breast cancer. We hypothesized that Polycomb might play a role in preneoplastic states in the pancreas and in tumour development/progression. To test these ideas, we determined the expression of PRC1 complex proteins (Bmi1 and Ring1b) during pancreatic development and in pancreatic tissue from mouse models of disease: acute and chronic pancreatic injury, duct ligation, and in K-Ras(G12V) conditional knock-in and caerulein-treated K-Ras(G12V) mice. The study was extended to human pancreatic tissue samples. To obtain mechanistic insights, Bmi1 expression in cells undergoing in vitro exocrine cell metaplasia and the effects of Bmi1 depletion in an acinar cancer cell line were studied. We found that Bmi1 and Ring1B are expressed in pancreatic exocrine precursor cells during early development and in ductal and islet cells-but not acinar cells-in the adult pancreas. Bmi1 expression was induced in acinar cells during acute injury, in acinar-ductal metaplastic lesions, as well as in pancreatic intraepithelial neoplasia (PanIN) and PDAC. In contrast, Ring1B expression was only significantly and persistently up-regulated in high-grade PanINs and in PDAC. Bmi1 knockdown in cultured acinar tumour cells led to changes in the expression of various digestive enzymes. Our results suggest that Bmi1 and Ring1B are modulated in pancreatic diseases and could contribute differently to tumour development.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Nuclear Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatitis, Chronic/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Acute Disease , Animals , Cells, Cultured , Disease Models, Animal , Humans , Male , Metaplasia/metabolism , Mice , Mice, Inbred C57BL , Pancreas/metabolism , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/pathology , Pancreatitis/metabolism , Polycomb Repressive Complex 1 , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Rats , Rats, Wistar , Transcription Factors/metabolism
18.
Pancreatology ; 8(4-5): 462-9, 2008.
Article in English | MEDLINE | ID: mdl-18724072

ABSTRACT

Pancreatic cancer has a very poor prognosis, in part due to its diagnosis at late stages of the disease and to limited response to chemotherapy and radiotherapy. The vast majority of pancreatic cancers are classified as pancreatic ductal adenocarcinomas (PDACs). Despite advances in knowledge on the cellular origin of PDAC or the involvement of signal transduction pathways therein, many questions remain unanswered. In this review, we summarize recent findings and current hypotheses regarding these two questions. Since pancreatitis is a risk factor for human PDAC, and the latter proceeds with an intense fibrotic reaction, we also analyze the role of the stroma in PDAC progression. An improved understanding of these key aspects for PDAC ontogeny will open new avenues for tumor prevention, early detection, and improved therapy.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , Signal Transduction , Humans
19.
J Biol Chem ; 282(17): 12851-9, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17322296

ABSTRACT

Poly(ADP-ribose) polymerase 1 (PARP-1) is a nuclear enzyme that is involved in modulating chromatin structure, regulation of gene expression, and sensing DNA damage. Here, we report that PARP-1 enzymatic activity is inhibited by macroH2A, a vertebrate histone H2A variant that is enriched on facultative heterochromatin. MacroH2A family members have a large C-terminal non-histone domain (NHD) and H2A-like histone domain. MacroH2A1.2 and PARP-1 interact in vivo and in vitro via the NHD. The NHD of each macroH2A family member was sufficient to inhibit PARP-1 enzymatic activity in vitro. The NHD of macroH2A1.2 was a mixed inhibitor of PARP-1 catalytic activity, with affects on both catalytic activity and the substrate binding affinity of PARP-1. Depletion of PARP-1 by RNA interference caused reactivation of a reporter gene on the inactive X chromosome, demonstrating that PARP-1 participates in the maintenance of silencing. These results suggest that one function of macroH2A in gene silencing is to inhibit PARP-1 enzymatic activity, and this may affect PARP-1 association with chromatin.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Chromatin/enzymology , Gene Silencing/physiology , Histones/metabolism , Poly(ADP-ribose) Polymerases/metabolism , X Chromosome Inactivation/physiology , Cell Line , Chromatin/genetics , Histones/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Protein Binding/physiology , Protein Structure, Tertiary/physiology
20.
Mol Cell Biol ; 25(24): 11047-58, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16314526

ABSTRACT

Polycomb group (PcG) proteins are epigenetic chromatin modifiers involved in heritable gene repression. Two main PcG complexes have been characterized. Polycomb repressive complex 2 (PRC2) is thought to be involved in the initiation of gene silencing, whereas Polycomb repressive complex 1 (PRC1) is implicated in the stable maintenance of gene repression. Here, we investigate the kinetic properties of the binding of one of the PRC1 core components, BMI1, with PcG bodies. PcG bodies are unique nuclear structures located on regions of pericentric heterochromatin, found to be the site of accumulation of PcG complexes in different cell lines. We report the presence of at least two kinetically different pools of BMI1, a highly dynamic and a less dynamic fraction, which may reflect BMI1 pools with different binding capacities to these stable heterochromatin domains. Interestingly, PRC2 members EED and EZH2 appear to be essential for BMI1 recruitment to the PcG bodies. Furthermore, we demonstrate that the maintenance DNA methyltransferase DNMT1 is necessary for proper PcG body assembly independent of DNMT-associated histone deacetylase activity. Together, these results provide new insights in the mechanism for regulation of chromatin silencing by PcG proteins and suggest a highly regulated recruitment of PRC1 to chromatin.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA-Binding Proteins/metabolism , Gene Silencing , Heterochromatin/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Cells, Cultured , DNA/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , Enhancer of Zeste Homolog 2 Protein , Heterochromatin/chemistry , Humans , Nuclear Proteins/analysis , Polycomb Repressive Complex 1 , Polycomb Repressive Complex 2 , Proto-Oncogene Proteins/analysis , Repressor Proteins/analysis , S Phase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...