Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Semin Cancer Biol ; 86(Pt 3): 431-444, 2022 11.
Article in English | MEDLINE | ID: mdl-35124196

ABSTRACT

mRNA export, translation, splicing, cleavage or capping determine mRNA stability, which represents one of the primary aspects regulating gene expression and function. RNA-binding proteins (RBPs) bind to their target mRNAs to regulate multiple cell functions by increasing or reducing their stability. In recent decades, studies of the role of RBPs in tumorigenesis have revealed an increasing number of proteins impacting the prognosis, diagnosis and cancer treatment. Several RBPs have been identified based on their interactions with oncogenes or tumor suppressor genes in human cancers, which are involved in apoptosis, the epithelial-mesenchymal transition (EMT), DNA repair, autophagy, cell proliferation, immune response, metabolism, and the regulation of noncoding RNAs. In this review, we propose a model showing how RBP mutations influence tumorigenesis, and we update the current knowledge regarding the molecular mechanism by which RBPs regulate cancer. Special attention is being devoted to RBPs that represent prognostic and diagnostic factors in cancer patients.


Subject(s)
Neoplasms , RNA-Binding Proteins , Humans , RNA-Binding Proteins/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Neoplasms/genetics , Neoplasms/metabolism , Epithelial-Mesenchymal Transition/genetics
2.
Semin Cancer Biol ; 60: 166-180, 2020 02.
Article in English | MEDLINE | ID: mdl-31369817

ABSTRACT

The acquisition of genetic alterations, clonal evolution, and the tumor microenvironment promote cancer progression, metastasis and therapy resistance. These events correspond to the establishment of the great phenotypic heterogeneity and plasticity of cancer cells that contribute to tumor progression and resistant disease. Targeting resistant cancers is a major challenge in oncology; however, the underlying processes are not yet fully understood. Even though current treatments can reduce tumor size and increase life expectancy, relapse and multidrug resistance (MDR) ultimately remain the second cause of death in developed countries. Recent evidence points toward stem-like phenotypes in cancer cells, promoted by cancer stem cells (CSCs), as the main culprit of cancer relapse, resistance (radiotherapy, hormone therapy, and/or chemotherapy) and metastasis. Many mechanisms have been proposed for CSC resistance, such as drug efflux through ABC transporters, overactivation of the DNA damage response (DDR), apoptosis evasion, prosurvival pathways activation, cell cycle promotion and/or cell metabolic alterations. Nonetheless, targeted therapy toward these specific CSC mechanisms is only partially effective to prevent or abolish resistance, suggesting underlying additional causes for CSC resilience. This article aims to provide an integrated picture of the MDR mechanisms that operate in CSCs' behavior and to propose a novel model of tumor evolution during chemotherapy. Targeting the pathways mentioned here might hold promise and reveal new strategies for future clinical therapeutic approaches.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Autophagy , Biomarkers , DNA Damage , Disease Susceptibility , Endoplasmic Reticulum Stress , Epigenesis, Genetic , Exosomes/metabolism , Hippo Signaling Pathway , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms/drug therapy , Neoplasms/etiology , Neoplasms/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Unfolded Protein Response
3.
Oncol Rep ; 41(2): 1275-1283, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30483800

ABSTRACT

Prostate cancer (PCa) is the second most common type of male malignancy worldwide. The transcription factor zinc finger E­box binding homeobox 1 (ZEB1) is associated with epithelial­mesenchymal transition and is also involved in regulation of androgen receptor (AR) expression, the main ligands of which are testosterone and dihydrotestosterone (DHT). These androgens are synthesized through the steroidogenic pathway within the prostate, and their synthesis is altered in PCa. The present study aimed to determine the ZEB1­induced alterations in androgen synthesis and AR expression in the DU145 PCa cell line. Reverse transcription­quantitative polymerase chain reaction, western blotting and immunocytochemistry were used to determine the mRNA and protein expression levels, and cellular localization of steroidogenic pathway enzymes in the DU145 cell line in response to ZEB1 silencing. Furthermore, the concentrations of testosterone and DHT were detected in cell culture medium using ELISA. ZEB1­silenced cells exhibited an increase in testosterone and DHT production, an increase in AR expression and an alteration in the steroidogenic pathway. In particular, steroidogenic acute regulatory protein and 5α­reductase 2 expression levels were decreased, whereas cytochrome P450 family 17 subfamily A member 1, 5α­reductase 1, aldo­keto reductase family 1 member D1 and aldo­keto reductase family 1 member C2 expression levels were increased. In conclusion, the present study provided novel information regarding the regulation of intratumoral androgen production in PCa, which is relevant for the progression of the disease to a castration­resistant form.


Subject(s)
Dihydrotestosterone/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Testosterone/biosynthesis , Zinc Finger E-box-Binding Homeobox 1/physiology , Cell Line, Tumor , Dihydrotestosterone/analysis , Gene Silencing , Humans , Male , Prostate/metabolism , Prostatic Neoplasms, Castration-Resistant/chemistry , Receptors, Androgen/metabolism , Testosterone/analysis , Zinc Finger E-box-Binding Homeobox 1/genetics
4.
Oncogene ; 33(9): 1124-34, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-23455324

ABSTRACT

Prostate tumor overexpressed-1 (PTOV1), a modulator of the Mediator transcriptional regulatory complex, is expressed at high levels in prostate cancer and other neoplasias in association with a more aggressive disease. Here we show that PTOV1 interacts directly with receptor of activated protein C kinase 1 (RACK1), a regulator of protein kinase C and Jun signaling and also a component of the 40S ribosome. Consistent with this interaction, PTOV1 was associated with ribosomes and its overexpression promoted global protein synthesis in prostate cancer cells and COS-7 fibroblasts in a mTORC1-dependent manner. Transfection of ectopic PTOV1 enhanced the expression of c-Jun protein without affecting the levels of c-Jun or RACK1 mRNA. Conversely, knockdown of PTOV1 caused significant declines in global protein synthesis and c-Jun protein levels. High levels of PTOV1 stimulated the motility and invasiveness of prostate cancer cells, which required c-Jun, whereas knockdown of PTOV1 strongly inhibited the tumorigenic and metastatic potentials of PC-3 prostate cancer cells. In human prostate cancer samples, the expression of high levels of PTOV1 in primary and metastatic tumors was significantly associated with increased nuclear localization of active c-Jun. These results unveil new functions of PTOV1 in the regulation of protein translation and in the progression of prostate cancer to an invasive and metastatic disease.


Subject(s)
Neoplasm Proteins/genetics , Protein Biosynthesis/genetics , Proto-Oncogene Proteins c-jun/genetics , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , COS Cells , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line , Cell Line, Tumor , Cell Movement/genetics , Chlorocebus aethiops , Disease Progression , Dogs , Humans , Madin Darby Canine Kidney Cells , Male , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neoplasm Proteins/metabolism , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-jun/metabolism , Receptors for Activated C Kinase , Receptors, Cell Surface , Ribosomes/genetics , Ribosomes/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
5.
Cancer Metastasis Rev ; 33(1): 115-41, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24375388

ABSTRACT

Ribosome biogenesis is the most demanding energetic and metabolic expenditure of the cell. The nucleolus, a nuclear compartment, coordinates rRNA transcription, maturation, and assembly into ribosome subunits. The transcription process is highly coordinated with ribosome biogenesis. In this context, ribosomal proteins (RPs) play a crucial role. In the last decade, an increasing number of studies have associated RPs with extraribosomal functions related to proliferation. Importantly, the expression of RPs appears to be deregulated in several human disorders due, at least in part, to genetic mutations. Although the deregulation of RPs in human malignancies is commonly observed, a more complex mechanism is believed to be involved, favoring the tumorigenic process, its progression and metastasis. This review explores the roles of the most frequently mutated oncogenes and tumor suppressor genes in human cancer that modulate ribosome biogenesis, including their interaction with RPs. In this regard, we propose a new focus for novel therapies.


Subject(s)
Carcinogenesis/genetics , Neoplasms/genetics , Ribosomal Proteins/genetics , Tumor Suppressor Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Models, Genetic , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Ribosomal Proteins/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Signal Transduction/genetics , Tumor Suppressor Proteins/metabolism
6.
Gut ; 53(7): 993-1000, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15194650

ABSTRACT

BACKGROUND: Overexpression of tissue plasminogen activator (t-PA) in pancreatic cancer cells promotes invasion and proliferation in vitro and tumour growth and angiogenesis in vivo. AIMS: To understand the mechanisms by which t-PA favours cancer progression, we analysed the surface membrane proteins responsible for binding specifically t-PA and studied the contribution of this interaction to the t-PA promoted invasion of pancreatic cancer cells. METHODS: The ability of t-PA to activate plasmin and a fluorogenic plasmin substrate was used to analyse the nature of the binding of active t-PA to cell surfaces. Specific binding was determined in two pancreatic cancer cell lines (SK-PC-1 and PANC-1), and complex formation analysed by co-immunoprecipitation experiments and co-immunolocalisation in tumours. The functional role of the interaction was studied in Matrigel invasion assays. RESULTS: t-PA bound to PANC-1 and SK-PC-1 cells in a specific and saturable manner while maintaining its activity. This binding was competitively inhibited by specific peptides interfering with the interaction of t-PA with annexin II. The t-PA/annexin II interaction on pancreatic cancer cells was also supported by co-immunoprecipitation assays using anti-t-PA antibodies and, reciprocally, with antiannexin II antibodies. In addition, confocal microscopy showed t-PA and annexin II colocalisation in tumour tissues. Finally, disruption of the t-PA/annexin II interaction by a specific hexapeptide significantly decreased the invasive capacity of SK-PC-1 cells in vitro. CONCLUSION: t-PA specifically binds to annexin II on the extracellular membrane of pancreatic cancer cells where it activates local plasmin production and tumour cell invasion. These findings may be clinically relevant for future therapeutic strategies based on specific drugs that counteract the activity of t-PA or its receptor annexin II, or their interaction at the surface level.


Subject(s)
Annexin A2/metabolism , Pancreatic Neoplasms/metabolism , Plasminogen/metabolism , Tissue Plasminogen Activator/metabolism , Binding, Competitive , Cell Membrane/metabolism , Humans , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , Neoplasm Proteins/physiology , Pancreatic Neoplasms/pathology , Tissue Plasminogen Activator/physiology , Tumor Cells, Cultured
7.
Oncogene ; 20(12): 1455-64, 2001 Mar 22.
Article in English | MEDLINE | ID: mdl-11313889

ABSTRACT

In a search for molecular markers of progression in prostate cancer by means of differential display, we have identified a new gene, which we have designated PTOV1. Semiquantitative RT-PCR has established that nine out of 11 tumors overexpress PTOV1 at levels significantly higher than benign prostatic hyperplasia or normal prostate tissue. The human PTOV1 protein consists almost entirely of two repeated blocks of homology of 151 and 147 amino acids, joined by a short linker peptide, and is encoded by a 12-exon gene localized in chromosome 19q13.3. A Drosophila melanogaster PTOV1 homolog also contains two tandemly arranged PTOV blocks. A second gene, PTOV2, was identified in humans and Drosophila, coding for proteins with a single PTOV homology block and unrelated amino- and carboxyl-terminal extensions. A 1.8-Kb PTOV1 transcript was detected abundantly in normal human brain, heart, skeletal muscle, kidney and liver, and at low levels in normal prostate. Immunocytochemical analysis and expression of chimeric GFP-PTOV1 proteins in cultured cells showed a predominantly perinuclear localization of PTOV1. In normal prostate tissue and in prostate adenomas, PTOV1 was undetectable or expressed at low levels, whereas nine out of 11 prostate adenocarcinomas showed a strong immunoreactivity, with a focal distribution in areas of carcinoma and prostatic intraepithelial neoplasia. Therefore, PTOV1 is a previously unknown gene, overexpressed in early and late stages of prostate cancer. The PTOV homology block represents a new class of conserved sequence blocks present in human, rodent and fly proteins.


Subject(s)
Biomarkers, Tumor , Drosophila Proteins , Neoplasm Proteins , Prostatic Neoplasms/genetics , Proteins/genetics , Recombinant Fusion Proteins , Sequence Homology, Amino Acid , Adenocarcinoma/genetics , Amino Acid Sequence , Base Sequence , Cell Compartmentation , Chromosome Mapping , Chromosomes, Human, Pair 19 , Databases, Factual , Humans , In Situ Hybridization, Fluorescence , Male , Mediator Complex , Molecular Sequence Data , Prostatic Hyperplasia/genetics , Proteins/isolation & purification , RNA, Messenger/isolation & purification , RNA, Neoplasm/isolation & purification , Tissue Distribution
8.
Am J Pathol ; 153(1): 201-12, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9665481

ABSTRACT

Because hepatocyte growth factor (HGF) is a potent mitogen for normal human exocrine pancreas cells (NPCs) in vitro, we have analyzed the expression of HGF and its receptor, Met, in NPC and pancreas cancer cells and studied its effects in vitro. Using immunohistochemistry, Northern blotting, and reverse transcription-polymerase chain reaction, we examined the expression of HGF and Met in normal pancreas and pancreas cancer. Scatter assays, wound-healing assays, and migration through transwell filters were used to study HGF-stimulated motility of IMIM-PC-2 cancer cells. In tumors, HGF is mainly detected in stromal cells, whereas Met is overexpressed in cancer cells with an unpolarized distribution. In vitro, HGF stimulates motogenesis but not proliferation in cancer cells. Cell motility is accompanied by a rapid decrease in the cytoskeleton-bound E-cadherin, an acceleration of cellular adhesion to the substrate, an up-regulation of urokinase plasminogen activator (u-PA) RNA and protein, and a change in the solubility and proteolysis of the u-PA receptor. Cell motility is significantly reduced by inhibitors of u-PA proteolytic activity such as antibodies neutralizing u-PA activity, plasminogen activator inhibitor 1, and amiloride. These results show that a paracrine loop of HGF activation may participate in the development or progression of pancreas cancer. In vitro, the HGF-stimulated motogenesis of pancreas cancer cells involves the activation of the u-PA/u-PA receptor proteolytic system, suggesting its role in the invasive stages of tumor progression.


Subject(s)
Cadherins/metabolism , Hepatocyte Growth Factor/metabolism , Pancreatic Neoplasms/metabolism , Urokinase-Type Plasminogen Activator/physiology , Antibodies/pharmacology , Blotting, Northern , Blotting, Western , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Hepatocyte Growth Factor/pharmacology , Humans , Immunoenzyme Techniques , Microscopy, Confocal , Pancreas/metabolism , Plasminogen/pharmacology , Plasminogen Activator Inhibitor 1/pharmacology , Polymerase Chain Reaction , Proto-Oncogene Proteins c-met/metabolism , Receptors, Cell Surface/metabolism , Receptors, Urokinase Plasminogen Activator , Tissue Plasminogen Activator/antagonists & inhibitors , Tissue Plasminogen Activator/metabolism , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Urokinase-Type Plasminogen Activator/metabolism
9.
J Biol Chem ; 273(24): 15091-8, 1998 Jun 12.
Article in English | MEDLINE | ID: mdl-9614119

ABSTRACT

The phorbol ester phorbol 12-myristate 13-acetate induces remarkable phenotypic changes in intestinal HT-29 M6 cells; these changes consist of loss of homotypic adhesion and inactivation of E-cadherin. In parallel, cell growth is retarded. We have transfected HT-29 M6 cells with an activated form of the conventional protein kinase Calpha (cPK-Calpha). Expression of this isoform induced the acquisition of a scattered phenotype, similar to that adopted by cells after addition of phorbol 12-myristate 13-acetate, with very low cell-to-cell aggregation and undetectable levels of functional E-cadherin. These cell clones were highly motile and rapidly invaded embryonic chick heart fragments. Furthermore, cells expressing activated-cPK-Calpha showed decreased proliferation in comparison to control clones. We have also studied how these two apparently antagonistic changes affect the tumorigenic ability of HT-29 M6 cells. When the different cell clones were xenografted into athymic mice, the effect on cell growth seemed to predominate. Expression of activated-cPK-Calpha significantly reduced the size of the tumors; the cells with the highest level of expression did not even form subcutaneous tumors. Besides their smaller size, the morphology of these tumors was clearly different from those originated by HT-29 M6 cells, and they could be defined as infiltrative on anatomo-pathological basis. These results indicate that cPK-Calpha controls both cell-to-cell adhesion and proliferation of intestinal cells.


Subject(s)
Cell Adhesion/genetics , Cell Movement/genetics , Neoplasm Invasiveness/physiopathology , Protein Kinase C/physiology , Animals , Cadherins/analysis , Carbazoles/pharmacology , Cell Size/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Indoles/pharmacology , Maleimides/pharmacology , Mice , Mice, Nude , Mutagenesis, Site-Directed/genetics , Neoplasms, Experimental/pathology , Protein Kinase C/genetics , Tetradecanoylphorbol Acetate/antagonists & inhibitors , Tetradecanoylphorbol Acetate/pharmacology , Transfection/genetics , Transplantation, Heterologous/pathology , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/analysis
10.
Oncogene ; 16(5): 625-33, 1998 Feb 05.
Article in English | MEDLINE | ID: mdl-9482108

ABSTRACT

Plasminogen activators (PAs) play an important role in tumor cell invasion. We have analysed the expression of tissue-type PA (t-PA), urokinase-type PA (u-PA), and their respective receptors, annexin II and u-PAR, in normal and neoplastic cultures of pancreatic cells, as well as in pancreatic tissues, and have examined their role in tumor invasiveness in vitro. Using Northern blotting, Western blotting, and ELISA, t-PA is detected in cultured pancreas cancer cells displaying a well differentiated phenotype but it is undetectable in less differentiated cells and in normal pancreatic cultures. In contrast, u-PA transcripts, protein, and enzymatic activity are detected both in cancer cells and in normal cultures. Higher levels of u-PAR and annexin II are present in cancer cells than in normal cultures and, in SK-PC-1 cells, both receptors are localized in the basolateral membrane. In vitro invasion assays indicate that both t-PA and u-PA contribute to the invasiveness of SK-PC-1 cells through reconstituted extracellular matrix. To determine the relevance of these studies to pancreas cancer, immunohistochemical assays have been used to examine the expression of t-PA, u-PA, and their receptors in normal and neoplastic tissues. t-PA is absent from normal pancreas and from tumor associated pancreatitis, whereas it is detected in the majority of pancreas cancer tissues (16/17). Annexin II is also overexpressed in some tumors (5/13). u-PAR is overexpressed in most tumor samples examined (14/15), while u-PA is weakly detected in a low number of cases (3/14); both u-PAR and u-PA are overexpressed in areas of tumor associated pancreatitis. Indirect evidences indicate that K-ras and p53 mutated proteins can regulate the expression of PAs. In pancreatic cancer we have found an association between codon 12 K-ras mutations and t-PA expression (P=0.04). These results support the contention that, in the exocrine pancreas, activation of t-PA is more specifically associated to neoplastic transformation and to the invasive phenotype, whereas the induction of u-PA/u-PAR system might be more relevant to inflammatory or non-neoplastic events.


Subject(s)
Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Tissue Plasminogen Activator/physiology , Annexin A2/biosynthesis , Blotting, Northern , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Neoplasm Invasiveness , Pancreas/enzymology , Pancreatic Neoplasms/genetics , Phenotype , Receptors, Cell Surface/biosynthesis , Receptors, Urokinase Plasminogen Activator , Tissue Plasminogen Activator/biosynthesis , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/biosynthesis , Urokinase-Type Plasminogen Activator/physiology
11.
Mol Cell Biol ; 18(1): 576-89, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9418904

ABSTRACT

By means of differential RNA display, we have isolated a cDNA corresponding to transcripts that are down-regulated upon differentiation of the goblet cell-like HT-29-M6 human colon carcinoma cell line. These transcripts encode proteins originally identified as CROC-1 on the basis of their capacity to activate transcription of c-fos. We show that these proteins are similar in sequence, and in predicted secondary and tertiary structure, to the ubiquitin-conjugating enzymes, also known as E2. Despite the similarities, these proteins lack a critical cysteine residue essential for the catalytic activity of E2 enzymes and, in vitro, they do not conjugate or transfer ubiquitin to protein substrates. These proteins constitute a distinct subfamily within the E2 protein family and are highly conserved in phylogeny from yeasts to mammals. Therefore, we have designated them UEV (ubiquitin-conjugating E2 enzyme variant) proteins, defined as proteins similar in sequence and structure to the E2 ubiquitin-conjugating enzymes but lacking their enzymatic activity (HW/GDB-approved gene symbol, UBE2V). At least two human genes code for UEV proteins, and one of them, located on chromosome 20q13.2, is expressed as at least four isoforms, generated by alternative splicing. All human cell types analyzed expressed at least one of these isoforms. Constitutive expression of exogenous human UEV in HT-29-M6 cells inhibited their capacity to differentiate upon confluence and caused both the entry of a larger proportion of cells in the division cycle and an accumulation in G2-M. This was accompanied with a profound inhibition of the mitotic kinase, cdk1. These results suggest that UEV proteins are involved in the control of differentiation and could exert their effects by altering cell cycle distribution.


Subject(s)
Cell Cycle , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Ligases/genetics , Amino Acid Sequence , Base Sequence , Cell Cycle/genetics , Cell Differentiation/genetics , Chromosome Mapping , Chromosomes, Human, Pair 20 , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Gene Expression Regulation , Humans , Ligases/biosynthesis , Molecular Sequence Data , Mucus/metabolism , Sequence Alignment , Tumor Cells, Cultured , Ubiquitin-Conjugating Enzymes
12.
FEBS Lett ; 385(1-2): 72-6, 1996 Apr 29.
Article in English | MEDLINE | ID: mdl-8641471

ABSTRACT

We have used subtractive hybridization to isolate cDNAs overexpressed in SK-PC-1 pancreas cancer cells. Forty-five independent clones corresponding to 11 genes were identified. Their expression in cultured pancreas cancer cells, normal pancreas tissue, and normal exocrine pancreas cultures was examined by Northern blotting. cDNA clones can be grouped into two broad categories: (1) those corresponding to genes expressed at high levels both in tumor cell lines and in primary cultures of normal pancreas, but not in normal tissue (i.e. thymosin beta4(3), cytokeratin 18, beta-actin, pyruvate kinase and mitochondrial genes); and (2) those corresponding to genes expressed at high levels in pancreas cancer cultures but not in normal pancreas tissue or cultured cells (i.e. tissue-type plasminogen activator and cathepsin H). The overexpression of these proteases in pancreas cancers suggests that they play a role in the aggressive biological behavior of this tumor.


Subject(s)
Antigens, Neoplasm/genetics , Cathepsins/genetics , Cell Adhesion Molecules , Cysteine Endopeptidases , Membrane Glycoproteins/genetics , Pancreatic Neoplasms/genetics , Tissue Plasminogen Activator/genetics , Cathepsin H , Cloning, Molecular/methods , Cytoskeletal Proteins/genetics , DNA, Complementary/genetics , DNA, Neoplasm/genetics , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Nucleic Acid Hybridization , Pancreas/physiology , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/immunology , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Sequence Analysis, DNA , Tumor Cells, Cultured
13.
Biochem J ; 301 ( Pt 1): 105-10, 1994 Jul 01.
Article in English | MEDLINE | ID: mdl-8037656

ABSTRACT

beta-Tubulin synthesized in vitro in rabbit reticulocyte lysate is found associated with 900 kDa complexes (C900) containing T Complex Polypeptide 1 (TCP1), heat-shock protein (hsp) 70 and other unidentified proteins, with smaller 300 kDa complexes (C300) of unknown nature, in dimeric association with reticulocyte alpha-tubulin and in monomeric forms. Pulse-chase experiments indicated that production of fully functional beta-tubulin was preceded by its association with C900 and C300 multimolecular complexes and by the appearance of beta-monomers. The high-molecular-mass forms appeared as intermediate products in the process leading to fully functional dimerizable beta-tubulin. C300-associated tubulin can be released as beta-monomer by addition of a cofactor present in reticulocyte lysate. Here a 25 kDa protein which releases tubulin monomers from C300 has been identified and characterized. The protein specifically released monomers from C300, but not from C900, in a process favoured by GTP.


Subject(s)
Tubulin/metabolism , Animals , Cytosol/metabolism , DNA, Complementary/genetics , Guanosine Triphosphate/metabolism , In Vitro Techniques , Macromolecular Substances , Mice , Molecular Weight , Protein Conformation , Protein Folding , Protein Precursors/chemistry , Protein Precursors/metabolism , Proteins/chemistry , Proteins/metabolism , Rabbits , Reticulocytes/metabolism , Tubulin/chemistry , Tubulin/genetics
14.
Arch Biochem Biophys ; 310(2): 428-32, 1994 May 01.
Article in English | MEDLINE | ID: mdl-8179328

ABSTRACT

Binding of heat-shock protein (hsp70) to polymerized tubulin has been investigated by in vitro experiments. The tubulin region involved in binding to hsp70 corresponds to the carboxy-terminal residues 431-444, also involved in the association with other microtubule-associated proteins (MAPs). Additionally, the putative tubulin binding motif in the hsp70 protein contains a sequence related to the motif described for MAP1B protein.


Subject(s)
Heat-Shock Proteins/metabolism , Tubulin/metabolism , Amino Acid Sequence , Animals , Binding, Competitive , Brain/metabolism , Cattle , Cloning, Molecular , Electrophoresis, Polyacrylamide Gel , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/isolation & purification , Humans , Kinetics , Microtubule Proteins/chemistry , Microtubule Proteins/metabolism , Molecular Sequence Data , Molecular Weight , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Biosynthesis , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Reticulocytes/metabolism , Transcription, Genetic , Tubulin/chemistry , Tubulin/isolation & purification
15.
J Cell Sci ; 106 ( Pt 2): 627-32, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8282766

ABSTRACT

A toroid multisubunit complex of 800-900 kDa has been implicated in assisting protein folding of at least two cytoplasmic proteins, actin and tubulin. This process is dependent on the presence of magnesium ions and ATP hydrolysis. In vitro translation of cDNAs encoding different alpha- and beta-tubulin isotypes also gives rise to the formation of complexes of about 300 kDa. These complexes have been functionally implicated in the incorporation of tubulin monomers within the tubulin heterodimer. This work shows that, in addition to ATP hydrolysis, the incorporation of newly synthesized tubulin subunits into functional heterodimers requires GTP hydrolysis in the presence of magnesium ions. A two-step process is suggested, a first ATP-dependent step in which the 900 kDa complexes are implicated in a similar way to the step taking place in actin folding, and a second GTP-dependent step in which the 300 kDa complexes are involved in the assembly of the heterodimer.


Subject(s)
Guanosine Triphosphate/metabolism , Intracellular Signaling Peptides and Proteins , Microtubule-Associated Proteins , Tubulin/metabolism , Adenosine Triphosphate/metabolism , Animals , Hydrolysis , Magnesium/metabolism , Mice , Molecular Weight , Nuclear Proteins/metabolism , Protein Conformation , Protein Folding , Rabbits , Rats , Tubulin/chemistry , Tubulin/genetics , Ubiquitin-Protein Ligases , t-Complex Genome Region
16.
Mol Cell Biol ; 11(3): 1334-43, 1991 Mar.
Article in English | MEDLINE | ID: mdl-1996095

ABSTRACT

The 5' flanking region of the mouse N-ras gene was investigated to determine the elements governing transcriptional activity of the gene. The promoter did not contain typical TATA or CCAAT boxes, and according to primer extension and RNase protection analyses, transcription started at several sites. These assays also confirmed the short nucleotide distance interposed between the N-ras transcription unit and the previously described upstream unr gene. Chromatin studies performed by digestion of nuclei with DNase I revealed the presence of four hypersensitive sites: a, b, c, and d. Deletion mutagenesis of the 5' flanking region revealed sequences responsible for both promotion and inhibition of transcription. These sequences resided within 230 bp upstream of the transcription initiation site. Hypersensitive site b colocalized with the 76-bp segment with promoter activity. The negative regulatory element at position -180 colocalized with hypersensitive site a, was active on the N-ras promoter in stable as well as transient assays, and down-regulated the heterologous herpes simplex virus thymidine kinase promoter. Footprint analysis and in vivo transfection-competition experiments indicated that a trans-acting factor is responsible for the negative effect on transcription. The interaction between the cis-acting negative regulatory element and the promoter region may play a role in the tissue- and developmental-stage-specific patterns of expression of the N-ras gene.


Subject(s)
Promoter Regions, Genetic , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogenes , Regulatory Sequences, Nucleic Acid , Animals , Base Sequence , DNA Mutational Analysis , DNA-Binding Proteins/physiology , Mice , Molecular Sequence Data , Oligonucleotides/chemistry , Structure-Activity Relationship , Transcription Factors/physiology , Transcription, Genetic
17.
Nucleic Acids Res ; 18(16): 4891-9, 1990 Aug 25.
Article in English | MEDLINE | ID: mdl-2204029

ABSTRACT

The mammalian N-ras gene is believed to play a role in cellular proliferation, differentiation, and transformation. While investigating N-ras, we isolated cDNA's that originate from a closely linked upstream gene. RNase protection assays reveal that this gene, unr, is transcribed in the same direction as N-ras and that its 3' end is located just 130 base pairs away from the point at which N-ras transcription begins. The close spatial relationship between the two genes is conserved in all species from which the N-ras gene has been isolated. An open reading frame, potentially encoding a 798 amino acid protein, is contained within the unr cDNA. Neither the primary protein structure nor the nucleic acid sequence of unr is homologous to any other known gene, including N-ras. Unr transcripts are detected in mouse, rat and human cells, and Southern analysis indicates that the unr locus found immediately upstream of the N-ras gene is transcriptionaly active in the mouse since only a single copy of unr is detected in this species. Unr produces multiple transcripts that differ in their 3' ends and are apparently created through the differential use of multiple polyadenylation sites located in the 3' untranslated region of the gene. Both unr and N-ras are expressed in all tissues examined. In the testis, both genes are developmentally regulated, with an increase in expression occurring upon testicular maturation. Thus the two genes may be coordinately regulated, at least in certain circumstances. Our findings suggest that a thorough analysis of the relationship that exists between the two genes could potentially provide insights into the regulation and/or function of N-ras.


Subject(s)
Gene Expression Regulation , Genes, ras , Genes , Genetic Linkage , Testis/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Base Sequence , Blotting, Southern , DNA/genetics , DNA/isolation & purification , Gene Library , Humans , Male , Mice , Molecular Sequence Data , RNA-Binding Proteins/metabolism , Rats , Ribonucleases/metabolism , Sequence Homology, Nucleic Acid , Testis/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...