Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 197
Filter
1.
Oncogene ; 36(34): 4843-4858, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28414310

ABSTRACT

High CD44 expression is associated with enhanced malignant potential in esophageal squamous cell carcinoma (ESCC), among the deadliest of all human carcinomas. Although alterations in autophagy and CD44 expression are associated with poor patient outcomes in various cancer types, the relationship between autophagy and cells with high CD44 expression remains incompletely understood. In transformed oesophageal keratinocytes, CD44Low-CD24High (CD44L) cells give rise to CD44High-CD24-/Low (CD44H) cells via epithelial-mesenchymal transition (EMT) in response to transforming growth factor (TGF)-ß. We couple patient samples and xenotransplantation studies with this tractable in vitro system of CD44L to CD44H cell conversion to investigate the functional role of autophagy in generation of cells with high CD44 expression. We report that high expression of the autophagy marker cleaved LC3 expression correlates with poor clinical outcome in ESCC. In ESCC xenograft tumours, pharmacological autophagy inhibition with chloroquine derivatives depletes cells with high CD44 expression while promoting oxidative stress. Autophagic flux impairment during EMT-mediated CD44L to CD44H cell conversion in vitro induces mitochondrial dysfunction, oxidative stress and cell death. During CD44H cell generation, transformed keratinocytes display evidence of mitophagy, including mitochondrial fragmentation, decreased mitochondrial content and mitochondrial translocation of Parkin, essential in mitophagy. RNA interference-mediated Parkin depletion attenuates CD44H cell generation. These data suggest that autophagy facilitates EMT-mediated CD44H generation via modulation of redox homeostasis and Parkin-dependent mitochondrial clearance. This is the first report to implicate mitophagy in regulation of tumour cells with high CD44 expression, representing a potential novel therapeutic avenue in cancers where EMT and CD44H cells have been implicated, including ESCC.


Subject(s)
Autophagy/physiology , Hyaluronan Receptors/metabolism , Mitochondria/physiology , Oxidative Stress/physiology , Ubiquitin-Protein Ligases/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/physiology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Humans , Keratinocytes/metabolism , Keratinocytes/physiology , Mitochondria/metabolism , Oxidation-Reduction , RNA Interference/physiology , Transforming Growth Factor beta/metabolism
2.
Oncogene ; 34(18): 2347-59, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-24931169

ABSTRACT

Notch activity regulates tumor biology in a context-dependent and complex manner. Notch may act as an oncogene or a tumor-suppressor gene even within the same tumor type. Recently, Notch signaling has been implicated in cellular senescence. Yet, it remains unclear as to how cellular senescence checkpoint functions may interact with Notch-mediated oncogenic and tumor-suppressor activities. Herein, we used genetically engineered human esophageal keratinocytes and esophageal squamous cell carcinoma cells to delineate the functional consequences of Notch activation and inhibition along with pharmacological intervention and RNA interference experiments. When expressed in a tetracycline-inducible manner, the ectopically expressed activated form of Notch1 (ICN1) displayed oncogene-like characteristics inducing cellular senescence corroborated by the induction of G0/G1 cell-cycle arrest, Rb dephosphorylation, flat and enlarged cell morphology and senescence-associated ß-galactosidase activity. Notch-induced senescence involves canonical CSL/RBPJ-dependent transcriptional activity and the p16(INK4A)-Rb pathway. Loss of p16(INK4A) or the presence of human papilloma virus (HPV) E6/E7 oncogene products not only prevented ICN1 from inducing senescence but permitted ICN1 to facilitate anchorage-independent colony formation and xenograft tumor growth with increased cell proliferation and reduced squamous-cell differentiation. Moreover, Notch1 appears to mediate replicative senescence as well as transforming growth factor-ß-induced cellular senescence in non-transformed cells and that HPV E6/E7 targets Notch1 for inactivation to prevent senescence, revealing a tumor-suppressor attribute of endogenous Notch1. In aggregate, cellular senescence checkpoint functions may influence dichotomous Notch activities in the neoplastic context.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Receptor, Notch1/metabolism , Retinoblastoma Protein/metabolism , Signal Transduction , Cell Cycle Checkpoints , Cell Transformation, Viral , Cells, Cultured , Cellular Senescence , Esophageal Squamous Cell Carcinoma , Esophagus/cytology , Esophagus/metabolism , Humans , Keratinocytes/metabolism , Phosphorylation , Transforming Growth Factor beta/metabolism , Viral Proteins/metabolism
3.
Oncogene ; 33(4): 411-20, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-23318423

ABSTRACT

Overexpression of the NEDD9/HEF1/Cas-L scaffolding protein is frequent, and drives invasion and metastasis in breast, head and neck, colorectal, melanoma, lung and other types of cancer. We have examined the consequences of genetic ablation of Nedd9 in the MMTV-HER2/ERBB2/neu mouse mammary tumor model. Unexpectedly, we found that only a limited effect on metastasis in MMTV-neu;Nedd9(-/-) mice compared with MMTV-neu;Nedd9(+/+) mice, but instead a dramatic reduction in tumor incidence (18 versus 80%), and a significantly increased latency until tumor appearance. Orthotopic reinjection and tail-vein injection of cells arising from tumors, coupled with in vivo analysis, indicated tumors arising in MMTV-neu;Nedd9(-/-) mice had undergone mutational selection that overcame the initial requirement for Nedd9. To better understand the defects in early tumor growth, we compared mammary progenitor cell pools from MMTV-neu;Nedd9(-/-) versus MMTV-neu;Nedd9(+/+) mice. The MMTV-neu;Nedd9(-/-) genotype selectively reduced both the number and colony-forming potential of mammary luminal epithelial progenitor cells, while not affecting basal epithelial progenitors. MMTV-neu;Nedd9(-/-) mammospheres had striking defects in morphology and cell polarity. All of these defects were seen predominantly in the context of the HER2/neu oncogene, and were not associated with randomization of the plane of mitotic division, but rather with depressed expression the cell attachment protein FAK, accompanied by increased sensitivity to small molecule inhibitors of FAK and SRC. Surprisingly, in spite of these significant differences, only minimal changes were observed in the gene expression profile of Nedd9(-/-) mice, indicating critical Nedd9-dependent differences in cell growth properties were mediated via post-transcriptional regulation of cell signaling. Coupled with emerging data indicating a role for NEDD9 in progenitor cell populations during the morphogenesis of other tissues, these results indicate a functional requirement for NEDD9 in the growth of mammary cancer progenitor cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinogenesis/metabolism , Mammary Neoplasms, Experimental/metabolism , Neoplasm Invasiveness/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Carcinogenesis/genetics , Female , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse , Mice , Mice, Knockout , Neoplasm Invasiveness/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
4.
Oncogene ; 33(5): 539-49, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-23334327

ABSTRACT

Aurora kinase A (AURKA) localizes to centrosomes and mitotic spindles where it mediates mitotic progression and chromosomal stability. Overexpression of AURKA is common in cancer, resulting in acquisition of alternate non-mitotic functions. In the current study, we identified a novel role for AURKA in regulating ovarian cancer cell dissemination and evaluated the efficacy of an AURKA-selective small molecule inhibitor, alisertib (MLN8237), as a single agent and combined with paclitaxel using an orthotopic xenograft model of epithelial ovarian cancer (EOC). Ovarian carcinoma cell lines were used to evaluate the effects of AURKA inhibition and overexpression on migration and adhesion. Pharmacological or RNA interference-mediated inhibition of AURKA significantly reduced ovarian carcinoma cell migration and adhesion and the activation-associated phosphorylation of the cytoskeletal regulatory protein SRC at tyrosine 416 (pSRC(Y416)). Conversely, enforced expression of AURKA resulted in increased migration, adhesion and activation of SRC in cultured cells. In vivo tumor growth and dissemination were inhibited by alisertib treatment as a single agent. Moreover, combination of alisertib with paclitaxel, an agent commonly used in treatment of EOC, resulted in more potent inhibition of tumor growth and dissemination compared with either drug alone. Taken together, these findings support a role for AURKA in EOC dissemination by regulating migration and adhesion. They also point to the potential utility of combining AURKA inhibitors with taxanes as a therapeutic strategy for the treatment of EOC patients.


Subject(s)
Aurora Kinase A/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Azepines/pharmacology , Carcinoma, Ovarian Epithelial , Cell Adhesion , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement , Female , Humans , Mice , Mitosis/drug effects , Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms, Glandular and Epithelial/enzymology , Ovarian Neoplasms/enzymology , Paclitaxel/pharmacology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , RNA Interference , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays , src-Family Kinases/metabolism
5.
Oncogenesis ; 2: e59, 2013 Aug 05.
Article in English | MEDLINE | ID: mdl-23917221

ABSTRACT

Periostin (POSTN), a matricellular protein, has been reported to be important in supporting tumor cell dissemination. However, the molecular mechanisms underlying POSTN function within the tumor microenvironment are poorly understood. In this study, we observe that the inducible knockdown of POSTN decreases esophageal squamous cell carcinoma (ESCC) tumor growth in vivo and demonstrate that POSTN cooperates with a conformational missense p53 mutation to enhance invasion. Pathway analyses reveal that invasive esophageal cells expressing POSTN and p53(R175H) mutation display activation of signal transducer and activator of transcription 1 (STAT1) target genes, suggesting that the induction of STAT1 and STAT1-related genes could foster a permissive microenvironment that facilitates invasion of esophageal epithelial cells into the extracellular matrix. Genetic knockdown of STAT1 in transformed esophageal epithelial cells underscores the importance of STAT1 in promoting invasion. Furthermore, we find that STAT1 is activated in ESCC xenograft tumors, but this activation is attenuated with inducible knockdown of POSTN in ESCC tumors. Overall, these results highlight the novel molecular mechanisms supporting the capacity of POSTN in mediating tumor invasion during ESCC development and have implications of therapeutic strategies targeting the tumor microenvironment.

6.
Cell Death Differ ; 19(7): 1152-61, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22322860

ABSTRACT

Inactivation of the retinoblastoma protein (pRb) by phosphorylation triggers uncontrolled cell proliferation. Accordingly, activation of cyclin-dependent kinase (CDK)/cyclin complexes or downregulation of CDK inhibitors appears as a common event in human cancer. Here we show that Pin1 (protein interacting with NIMA (never in mitosis A)-1), a peptidylprolyl isomerase involved in the control of protein phosphorylation, is an essential mediator for inactivation of the pRb. Our results indicate that Pin1 controls cell proliferation by altering pRb phosphorylation without affecting CDK and protein phosphatase 1 and 2 activity. We demonstrated that Pin1 regulates tumor cell proliferation through direct interaction with the spacer domain of the pRb protein, and allows the interaction between CDK/cyclin complexes and pRb in mid/late G1. Phosphorylation of pRb Ser 608/612 is the crucial motif for Pin1 binding. We propose that Pin1 selectively boosts the switch from hypo- to hyper-phosphorylation of pRb in tumor cells. In addition, we demonstrate that the CDK pathway is responsible for the interaction of Pin1 and pRb. Prospectively, our findings therefore suggest that the synergism among CDK and Pin1 inhibitors holds great promise for targeted pharmacological treatment of cancer patients, with the possibility of reaching high effectiveness at tolerated doses.


Subject(s)
Peptidylprolyl Isomerase/metabolism , Retinoblastoma Protein/metabolism , Cell Line , Cell Proliferation , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , G1 Phase , Humans , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/antagonists & inhibitors , Peptidylprolyl Isomerase/genetics , Phosphorylation , Protein Binding , Protein Phosphatase 1/metabolism , Protein Phosphatase 2/metabolism , RNA Interference , RNA, Small Interfering/metabolism
7.
Breast Cancer Res Treat ; 104(2): 129-44, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17393302

ABSTRACT

Breast tumors with prominent plasma cell (PC) infiltrates often have a more favorable natural course that may plausibly be mediated by anti-tumor activity of the elicited antibodies. These breast tumor-associated PCs are typically IgG dominant in contrast to normal breast PCs, which are mainly IgA. It is our hypothesis that this PC infiltration represents a host immune response that is driven by one or more tumor antigens. Previously, we and others showed that medullary carcinoma (MC) had a focused repertoire and features suggestive of a protein antigen driven response. Infrequently, non-MC, not otherwise specified (NOS) breast tumors may exhibit heavy PC infiltrations, also of IgG isotype. In this first characterization of this favorable prognosis NOS subgroup, IgG heavy chain (Hc) and light chain (Lc) variable (V) regions from three PC-infiltrated NOS tumors were randomly cloned and sequenced. We found biased (V) gene usage by the infiltrating PCs and somatic hypermutation in the rearranged Ig Hc and Lc V regions that were compatible with antigenic selection of the progenitor B cells. The antibody response of NOS infiltrated breast cancer is repertoire-focused, with 13-68% of isolates being clonally reiterated in the samples. Each NOS patient used distinct Hc V-D-J and Lc V-J rearrangements, with her own immune response "footprint," but the overall pattern of gene usage followed that typical of exogenous antigen-induced immune responses. The data are consistent with the hypothesis that PC infiltrates infrequently arising in NOS tumors, as previously inferred for MC, are in response to one or more breast cancer-associated protein tumor antigens.


Subject(s)
Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Immunoglobulin G/immunology , Plasma Cells/immunology , Base Sequence , Breast Neoplasms/metabolism , Female , Humans , Immunoglobulin G/metabolism , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Isotypes/immunology , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Immunophenotyping , Lymphocytes/immunology , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid
8.
Curr Protoc Pharmacol ; Chapter 14: Unit 14.2, 2007 Jun.
Article in English | MEDLINE | ID: mdl-21948161

ABSTRACT

This unit provides two models of oral carcinogenesis (hamster cheek pouch and rat tongue), as well as more simple procedures to induce squamous cell carcinomas (SCCs) in mouse skin. The most significant usage of these models has been their application in studying the molecular pathology of the genesis and multi-step progression of tobacco-associated SCC. Nevertheless, these protocols have been used frequently as in vivo bioassays to demonstrate the chemopreventive and, to a lesser extent, the chemotherapeutic effects of numerous compounds. Several examples, using four different carcinogenesis protocols, are presented with detailed steps on how to elicit squamous lesions and when and how to apply test compounds that could have either cancer chemopreventive or chemotherapeutic effects.


Subject(s)
Carcinoma, Squamous Cell/chemically induced , Disease Models, Animal , Head and Neck Neoplasms/chemically induced , Animals , Benzo(a)pyrene/toxicity , Cricetinae , Female , Male , Mesocricetus , Mice , Rats , Rats, Inbred F344 , Skin Neoplasms/chemically induced , Squamous Cell Carcinoma of Head and Neck
9.
Acta Odontol Latinoam ; 15(1-2): 29-37, 2002.
Article in English | MEDLINE | ID: mdl-15208940

ABSTRACT

Furin is a proprotein convertase that activates many cancer development-related substrates such as growth factors, growth factor-receptors, adhesion molecules, and matrix degrading enzymes. Furin expression was studied in sections from tissue microarrays (TMA) and conventional paraffin blocks in a collection of squamous cell carcinomas (SCC) from three different sites. A total of 118 SCCs from the oral cavity, lung and esophagus as well as 34 precursor lesions (intraepithelial neoplasia) from the oral and bronchial mucosae were studied by immunohistochemistry. Furin expression was notably higher in most precursor lesions than in normal epithelia. Tumors from either the TMAs or the conventional blocks showed significant differences when compared to the mostly negative normal epithelia. High levels of furin expression were observed in approximately 50% SCCs of three different sites as well as in precursor lesions of the oral and bronchial mucosae. In addition another 30% showed low furin expression that was localized in all tumor cells including those in a basaloid position. Normal epithelia sometimes showed low level expression but the normal basal cells were always negative. These results show that furin is up-regulated in SCCs from three different organs and validates its use as a tumor marker in both invasive and pre-invasive neoplasia.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Furin/biosynthesis , Leukoplakia, Oral/enzymology , Mouth Neoplasms/enzymology , Neoplasm Proteins/biosynthesis , Biomarkers, Tumor , Esophageal Neoplasms/enzymology , Humans , Immunohistochemistry , Lung Neoplasms/enzymology , Protein Array Analysis , Up-Regulation
10.
Acta odontol. latinoam ; 15(1-2): 29-37, 2002.
Article in Spanish | LILACS-Express | LILACS, BINACIS | ID: biblio-1157645

ABSTRACT

Furin is a proprotein convertase that activates many cancer development-related substrates such as growth factors, growth factor-receptors, adhesion molecules, and matrix degrading enzymes. Furin expression was studied in sections from tissue microarrays (TMA) and conventional paraffin blocks in a collection of squamous cell carcinomas (SCC) from three different sites. A total of 118 SCCs from the oral cavity, lung and esophagus as well as 34 precursor lesions (intraepithelial neoplasia) from the oral and bronchial mucosae were studied by immunohistochemistry. Furin expression was notably higher in most precursor lesions than in normal epithelia. Tumors from either the TMAs or the conventional blocks showed significant differences when compared to the mostly negative normal epithelia. High levels of furin expression were observed in approximately 50


SCCs of three different sites as well as in precursor lesions of the oral and bronchial mucosae. In addition another 30


showed low furin expression that was localized in all tumor cells including those in a basaloid position. Normal epithelia sometimes showed low level expression but the normal basal cells were always negative. These results show that furin is up-regulated in SCCs from three different organs and validates its use as a tumor marker in both invasive and pre-invasive neoplasia.

11.
Acta odontol. latinoam ; 15(1-2): 29-37, 2002.
Article in English | BINACIS | ID: bin-39022

ABSTRACT

Furin is a proprotein convertase that activates many cancer development-related substrates such as growth factors, growth factor-receptors, adhesion molecules, and matrix degrading enzymes. Furin expression was studied in sections from tissue microarrays (TMA) and conventional paraffin blocks in a collection of squamous cell carcinomas (SCC) from three different sites. A total of 118 SCCs from the oral cavity, lung and esophagus as well as 34 precursor lesions (intraepithelial neoplasia) from the oral and bronchial mucosae were studied by immunohistochemistry. Furin expression was notably higher in most precursor lesions than in normal epithelia. Tumors from either the TMAs or the conventional blocks showed significant differences when compared to the mostly negative normal epithelia. High levels of furin expression were observed in approximately 50


SCCs of three different sites as well as in precursor lesions of the oral and bronchial mucosae. In addition another 30


showed low furin expression that was localized in all tumor cells including those in a basaloid position. Normal epithelia sometimes showed low level expression but the normal basal cells were always negative. These results show that furin is up-regulated in SCCs from three different organs and validates its use as a tumor marker in both invasive and pre-invasive neoplasia.

12.
Mol Carcinog ; 31(4): 224-32, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11536372

ABSTRACT

Pro-protein convertases (PCs) are proteases that recognize and cleave precursor proteins. Furin, a well-studied PC, is ubiquitously expressed, and it has been implicated in many physiological and pathological processes. Some substrates for furin, such as membrane type 1 (MT1) matrix metalloproteinase (MMP), an MMP that activates gelatinase, a collagen-degrading enzyme, are associated with the advanced malignant phenotype. This report examines the expression of furin in carcinoma cell lines of different invasive ability. The levels of furin mRNA and protein correlated with the aggressiveness of tumor cell lines derived from head and neck and lung cancers. Furin expression also was investigated in primary head and neck squamous cell carcinomas (HNSCCs). Furin mRNA was not detected in nonmetastasizing carcinomas. In contrast, furin mRNA was expressed in metastasizing HNSCCs. Immunohistochemistry and Western blot analysis confirmed these results at the protein level. Furin activity was investigated indirectly by evaluating the expression of the pro-form and the processed form of MT1-MMP. Metastasizing HNSCCs showed increased expression of MT1-MMP. Furthermore, pro-MT1-MMP expression was noted in most of the nonmetastasizing HNSCCs analyzed by Western blot, and it was absent in the metastasizing HNSCCs. This finding suggests a lower level of furin-mediated MT1-MMP activation in the less aggressive cancers. These observations indicate that furin plays a role in tumor progression. Its overexpression in more aggressive or metastasizing cancers resulted in increased MMP processing.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Head and Neck Neoplasms/enzymology , Subtilisins/biosynthesis , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/pathology , Female , Furin , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Proprotein Convertases , Serine Endopeptidases/biosynthesis , Subtilisins/metabolism , Tumor Cells, Cultured
13.
Proc Natl Acad Sci U S A ; 98(18): 10326-31, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11517338

ABSTRACT

Pro-protein convertases such as furin are expressed in many human tumor lines and primary tumors. Furin processes stromelysin-3, membrane type 1 matrix metalloproteinase (MMPs) involved in tumor cell invasiveness, as well as growth factors such as transforming growth factor beta1. Evaluation of furin expression in head and neck squamous cell carcinoma (HNSCC) cells exhibiting different invasive ability showed that furin overexpression correlated with their respective invasiveness. The use of a selective furin inhibitor, alpha 1-PDX (PDX) was studied in three furin-expressing invasive HNSCC cell lines. The effects of PDX transfection were evaluated in vivo and in vitro to determine changes in the malignant phenotype. Transfection of HNSCC cell lines with PDX resulted in significant decrease or absence of tumorigenicity after s.c. inoculation into severe combined immunodeficient mice. Likewise, in vitro invasiveness was reduced approximately 50%. The in vivo invasion assay using tracheal xenotransplants showed even more drastic reductions of the invasive ability of PDX-transfected cells (up to an 80% decrease). PDX-transfected cells did not invade or penetrated less into the tracheal wall tissues than their vector alone-transfected counterparts. In addition, the former cells showed a remarkable decrease in MMP-2 processing and activity. After PDX transfection the cells were less efficient in processing the tumor progression-associated furin substrates transforming growth factor beta1 and pro-membrane type 1-MMP. These findings indicate that furin inhibition is a feasible approach to attenuate and even abolish certain critical attributes of the advanced malignant phenotype. Thus, furin should be considered as a promising target for cancer therapy.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Subtilisins/antagonists & inhibitors , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Furin , Humans , In Vitro Techniques , Matrix Metalloproteinases/metabolism , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation , Rats , Subtilisins/metabolism , Transfection , Tumor Cells, Cultured , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/pharmacology
14.
Proc Natl Acad Sci U S A ; 98(14): 7970-5, 2001 Jul 03.
Article in English | MEDLINE | ID: mdl-11427729

ABSTRACT

Rearrangements of the high mobility group protein I-C (HMGI-C) gene, consisting in the loss of the carboxyl-terminal tail, have been frequently detected in benign human tumors of mesenchymal origin. We have previously demonstrated that transgenic (TG) mice carrying a truncated HMGI-C construct (HMGI-C/T) exhibit a giant phenotype together with a predominantly abdominal/pelvic lipomatosis. Here, we report that HMGI-C/T TG mice develop natural killer (NK)-T/NK cell lymphomas starting from 12 months of age. We found an increased expression of IL-2 and IL-15 proteins and their receptors in these lymphomas, and we demonstrate that HMGI-C/T protein positively regulates their expression in vitro. Therefore, the HMGI-C/T-mediated chronic stimulation of the IL-2/IL-15 pathway could be responsible for the onset of NK-T/NK cell lymphomas in HMGI-C/T TG mice.


Subject(s)
High Mobility Group Proteins/genetics , Interleukin-15/immunology , Interleukin-2/immunology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/immunology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Genetic Predisposition to Disease , High Mobility Group Proteins/immunology , Humans , Killer Cells, Natural/pathology , Lymphoma, T-Cell/etiology , Lymphoma, T-Cell/pathology , Mice , Mice, Transgenic
15.
Cancer Epidemiol Biomarkers Prev ; 10(5): 455-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11352854

ABSTRACT

CCND1 gene amplification and cyclin D1 protein overexpression are indicators for poor prognosis in invasive head and neck carcinomas. Increased CCND1 gene dosage is a more sensitive prognostic factor than protein overexpression as evaluated by conventional immunohistochemical techniques. Qualitative immunohistochemistry cannot distinguish cyclin D1 overexpression accompanied by amplification of the CCND1 gene from overexpression associated with normal CCND1 gene copy number. To improve the sensitivity of cyclin D1 protein determination, we applied quantitative techniques of image analysis to evaluate cyclin D1 in 54 head and neck carcinomas. There was a significantly higher rate of occurrence of adverse events (P = 0.043) among patients with CCND1 gene amplification than among those without gene amplification. There was a strong association between CCND1 gene amplification (as detected by Southern blot analysis) and the highest nuclear score (by image cytometry of the immunostained tumor sections). The predominance of cells in the lowest nuclear score category was significantly associated with normal copy number (P = 0.005). Conversely, the highest nuclear score was a significant predictor of gene dosage (P = 0.02). Similarly, high nuclear score was a good predictor of death as the final outcome of the disease (P = 0.01). Although somewhat less accurate than Southern blotting, image cytometry of immunohistochemical cyclin D1 stain appears to be a promising tool that could be useful for other tumor marker expression studies.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cyclin D1/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Image Cytometry/methods , Adult , Aged , Blotting, Southern , Culture Techniques , Cyclin D1/analysis , Female , Gene Amplification , Humans , Immunohistochemistry , Male , Middle Aged , Prognosis , Sensitivity and Specificity
16.
Cancer Res ; 61(10): 3961-8, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11358813

ABSTRACT

Exisulind (Aptosyn) is a novel antineoplastic drug being developed for the prevention and treatment of precancerous and malignant diseases. In colon tumor cells, the drug induces apoptosis by a mechanism involving cyclic GMP (cGMP) phosphodiesterase inhibition, sustained elevation of cGMP, and protein kinase G activation. We studied the effect of exisulind on bladder tumorigenesis induced in rats by the carcinogen, N-butyl-N-(4-hydroxybutyl) nitrosamine. Exisulind at doses of 800, 1000, and 1200 mg/kg (diet) inhibited tumor multiplicity by 36, 47, and 64% and tumor incidence by 31, 38, and 61%, respectively. Experiments on the human bladder tumor cell line, HT1376, showed that exisulind inhibited growth with a GI(50) of 118 microM, suggesting that the antineoplastic activity of the drug in vivo involved a direct effect on neoplastic urothelium. Exisulind also induced apoptosis as determined by DNA fragmentation, caspase activation, and morphology. Analysis of phosphodiesterase (PDE) isozymes in HT1376 cells showed PDE5 and PDE4 isozymes that were inhibited by exisulind with IC(50)s of 112 and 116 microM, respectively. Inhibition of PDE5 appears to be pharmacologically relevant, because treatment of HT1376 cells increased cGMP and activated protein kinase G at doses that induce apoptosis, whereas cyclic AMP levels were not changed. Immunocytochemistry showed that PDE5 was localized in discrete perinuclear foci in HT1376 cells. Immunohistochemistry showed that PDE5 was overexpressed in human squamous and transitional cell carcinomas compared with normal urothelium. The data lead us to conclude that future clinical trials of exisulind for human bladder cancer treatment and/or prevention should be considered and suggest a mechanism of action involving cGMP-mediated apoptosis induction.


Subject(s)
Anticarcinogenic Agents/pharmacology , Sulindac/pharmacology , Urinary Bladder Neoplasms/prevention & control , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , 3',5'-Cyclic-GMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Animals , Apoptosis/drug effects , Cell Division/drug effects , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4 , Cyclic Nucleotide Phosphodiesterases, Type 5 , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Female , Humans , Inhibitory Concentration 50 , Microscopy, Fluorescence , Rats , Rats, Inbred F344 , Sulindac/analogs & derivatives , Tumor Cells, Cultured , Urinary Bladder Neoplasms/chemically induced , Urinary Bladder Neoplasms/metabolism
17.
Cancer Lett ; 162(2): 161-5, 2001 Jan 26.
Article in English | MEDLINE | ID: mdl-11146221

ABSTRACT

Murine and human skin express an abundance of lipoxygenase isoforms whose functions are not understood. Substantial data have implicated a role for the 'platelet-type' 12-lipoxygenase (P-12LO) metabolite, 12(S)-hydroxy-eicosatetraenoic acid (12-HETE), in a variety of tumor functions. Using P-12LO deficient mice, we sought to examine the role of the P-12LO pathway in tumor initiation and progression. Two distinct genetic strains of P-12LO deficient and wild-type mice, B6/129 Sv and SENCAR, were evaluated in two-stage carcinogenesis experiments. Carcinoma incidence was significantly reduced in the P-12LO deficient mice of the B6/129 Sv background but not the SENCAR-backcrossed mice. In contrast, papilloma incidence was reduced on the SENCAR background but not in the B6/129 Sv strain mice. A separate experiment employing a complete carcinogenesis protocol failed to find any difference in papilloma or carcinoma incidence. Overall, these data suggest that the P-12LO pathway may contribute to tumor incidence and progression in two-stage, but not complete, carcinogenesis, depending on the genetic background.


Subject(s)
Arachidonate 12-Lipoxygenase/physiology , Blood Platelets/enzymology , Skin Neoplasms/enzymology , 9,10-Dimethyl-1,2-benzanthracene , Animals , Arachidonate 12-Lipoxygenase/deficiency , Arachidonate 12-Lipoxygenase/genetics , Arachidonic Acid/metabolism , Carcinogens , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/enzymology , Female , Mice , Mice, Inbred SENCAR , Mice, Knockout , Papilloma/chemically induced , Papilloma/enzymology , Skin Neoplasms/chemically induced
18.
J Natl Cancer Inst ; 92(23): 1934-40, 2000 Dec 06.
Article in English | MEDLINE | ID: mdl-11106685

ABSTRACT

BACKGROUND: Multidrug resistance-associated protein (MRP) 1 and canalicular multispecific organic anion transporter (cMOAT or MRP2) are adenosine triphosphate-binding cassette transporters that confer resistance to anticancer agents. In addition to these two transporters, there are at least four other human MRP subfamily members (MRP3 through MRP6). We and others reported previously that MRP3 is capable of conferring resistance to certain anticancer agents. In this study, we investigated whether MRP4 (MOAT-B), whose transcript accumulates to the highest levels in prostate tissue, has the capacity to confer drug resistance. METHODS: MRP4-transfected NIH3T3 cells were generated, and their drug sensitivity was analyzed. The subcellular localization of MRP4 was assessed by immunohistochemical analysis in transfected cells and in prostate tissue. Statistical tests were two-sided. RESULTS: MRP4 was detected as a 170-kd protein that was localized in the plasma membrane and cytoplasm of transfected cells. The MRP4 transfectants displayed 5.5-fold increased resistance to methotrexate in short-term drug-exposure assays (P=.022) and exhibited decreased cellular accumulation of this agent at 4 hours (P=.006) and 24 hours (P<.001). In continuous-exposure assays, however, the MRP4 transfectants did not display increased resistance for either methotrexate or natural product cytotoxic agents (anthracyclines, etoposide, vinca alkaloids, and paclitaxel [Taxol]). However, the transfectants did show increased resistance (2.3-fold) for the anti-acquired immunodeficiency syndrome nucleoside analogue 9-(2-phosphonylmethoxyethyl)adenine (PMEA) (P=.022) in continuous-exposure assays. Consistent with MRP4's plasma membrane localization in transfected cells, analysis of prostate tissue showed that MRP4 protein was localized primarily in the basolateral plasma membranes of tubuloacinar cells. CONCLUSIONS: These results indicate that MRP4 confers resistance to short-term methotrexate and continuous PMEA treatment. Given its structure, drug resistance profile and subcellular localization, MRP4 probably functions as an amphipathic anion efflux pump whose substrate range includes glutamate and phosphate conjugates.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , Antineoplastic Agents/pharmacology , Carrier Proteins/genetics , Drug Resistance, Multiple , Drug Resistance, Neoplasm , 3T3 Cells , ATP Binding Cassette Transporter, Subfamily B/physiology , Acrylates/pharmacology , Animals , Anion Transport Proteins , Antibiotics, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Carrier Proteins/physiology , Cloning, Molecular , DNA, Complementary/genetics , Drug Screening Assays, Antitumor , Humans , Immunoblotting , Immunohistochemistry , Male , Methotrexate/pharmacology , Mice , Polymers/pharmacology , Prostate/metabolism , Transfection
19.
Cancer Res ; 60(21): 5972-6, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11085515

ABSTRACT

The transcription factor E2F-1, a downstream regulator of the p16-cyclinD-Rb pathway, is required for cell cycle progression. Evidence shows that overexpression of E2F-1 can either promote or inhibit development of tumors, depending on tissue or experimental conditions. To study whether the E2F-1 gene plays a role in tumor progression, the expression of E2F-1 protein was evaluated in 10 human head and neck squamous cell carcinoma cell lines using Western blot analysis. In addition, the invasive ability of these cell lines was determined by evaluating the penetration of cell lines into the tracheal wall in an in vivo invasion assay using deepithelialized tracheas transplanted into the s.c. tissue of Scid mice. This study showed that the aggressive cell lines had higher expression of E2F-1 than the less invasive cell lines. To evaluate the hypothesis that E2F-1 enhances invasiveness, we selected two cell lines, SCC9 and SCC12, for a gene transfer experiment. These cell lines exhibited low invasive ability with low expression of E2F-1. Two stable clones with overexpression of transfected E2F-1 gene and two clones with their respective vector-alone control were selected from each cell line for in vivo invasion evaluation. The clones containing the transfected E2F-1 gene had significantly higher invasive ability than their respective vector-alone clones. Flow cytometry showed that parental, transfected E2F-1, and vector-alone cells had a similar proliferation pattern under normal culture conditions. Nevertheless, transfected E2F-1 cells exhibited a higher portion of cells in S phase than the control cells after serum-starvation and refeeding. The results indicated that overexpression of E2F-1 plays a positive role in cell cycle reentry from quiescence and is associated with increased in vivo invasiveness.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Transcription Factors/genetics , Animals , Carcinoma, Squamous Cell/metabolism , Cell Division/genetics , E2F Transcription Factors , E2F1 Transcription Factor , Gene Expression , Gene Transfer Techniques , Head and Neck Neoplasms/metabolism , Humans , Immunohistochemistry , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation , Rats , Rats, Inbred F344 , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/biosynthesis , Transplantation, Heterologous , Tumor Cells, Cultured
20.
Mol Carcinog ; 28(2): 63-9, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10900462

ABSTRACT

Processing of latent precursor proteins by proprotein convertases (PCs) into their biologically active products is a common mechanism required for many important biologic functions. This process is tightly regulated, leading to the generation of active peptides and proteins including neuropeptides and polypeptide hormones, protein tyrosine phosphatases, growth factors and their receptors, and enzymes including matrix metalloproteases (MMPs). These processing reactions occurs at pairs of basic amino acids. Within the past several years, a novel family of Ca(2+)-dependent serine proteases has been identified, all of which possess homology to the endoproteases subtilisin (bacteria) and kexin (yeast). This family of PCs is currently comprised of fewer than a dozen members, known as furin/paired basic amino-acid-cleaving enzyme (PACE), PC1/PC3, PC2, PC4, PACE4, PC5/PC6, and PC7/PC8/lymphoma proprotein convertase. They share a high degree of amino-acid identity of 50-75% within their catalytic domains. Despite the relatively high degree of homology in the PC family, only PACE4 and furin localize to the same chromosome: mouse chromosome 7 and human chromosome 15. Recent reports have supported a possible functional role for PCs in tumorigenesis. For instance, convertases have been shown to be expressed in various tumor lines and human primary tumors. Furin and PACE4 process stromelysin 3 (MMP-11 or Str-3), an MMP involved in tumor invasion, into its mature, active form. Similarly, a growing family of MMPs, known as membrane-type metalloproteinases (MT-MMPs), and growth factors and adhesion molecules such as E-cadherin show similar amino-acid motifs and thus could be activated by furin and PACE4. These data, taken together with the high expression levels of PACE4 in 50% of murine chemically induced spindle cell tumors, confer to PACE4 and possibly other PCs a possible functional role in the activation of MMPs and consequently in tumor cell invasion and tumor progression. This was further supported by the remarkable enhancement in the invasive ability of the PACE4-transfected murine tumor cell lines. Mol. Carcinog. 28:63-69, 2000.


Subject(s)
Membrane Proteins , Neoplasms/pathology , Serine Endopeptidases/metabolism , Subtilisins/metabolism , Animals , Furin , Humans , Mice , Neoplasms/enzymology , Proprotein Convertases , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...