Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Int J Urol ; 2024 May 09.
Article in English | MEDLINE | ID: mdl-38722221

ABSTRACT

OBJECTIVES: The JAVELIN Bladder 100 phase 3 trial showed that avelumab first-line maintenance + best supportive care significantly prolonged overall survival and progression-free survival versus best supportive care alone in patients with advanced urothelial carcinoma who were progression-free following first-line platinum-based chemotherapy. We report findings from J-AVENUE (NCT05431777), a real-world study of avelumab first-line maintenance therapy in Japan. METHODS: Medical charts of patients with advanced urothelial carcinoma without disease progression following first-line platinum-based chemotherapy, who received avelumab maintenance between February and November 2021, were reviewed. Patients were followed until June 2022. The primary endpoint was patient characteristics; secondary endpoints included time to treatment failure and progression-free survival. RESULTS: In 79 patients analyzed, median age was 72 years (range, 44-86). Primary tumor site was upper tract in 45.6% and bladder in 54.4%. The most common first-line chemotherapy regimen was cisplatin + gemcitabine (63.3%). Median number of chemotherapy cycles received was four. Best response to chemotherapy was complete response in 10.1%, partial response in 58.2%, and stable disease in 31.6%. Median treatment-free interval before avelumab was 4.9 weeks. With avelumab first-line maintenance therapy, the disease control rate was 58.2%, median time to treatment failure was 4.6 months (95% CI, 3.3-6.4), and median progression-free survival was 6.1 months (95% CI, 3.6-9.7). CONCLUSIONS: Findings from J-AVENUE show the effectiveness of avelumab first-line maintenance in patients with advanced urothelial carcinoma in Japan in clinical practice, with similar progression-free survival to JAVELIN Bladder 100 and previous real-world studies, supporting its use as a standard of care.

2.
Biochem Biophys Res Commun ; 476(4): 183-187, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27286705

ABSTRACT

P-glycoprotein (P-gp) is an ATP-binding cassette protein involved in cancer multi-drug resistance (MDR). It has been reported that infection with some bacteria and viruses induces changes in the activities of various drug-metabolizing enzymes and transporters, including P-gp. Although human adenoviruses (Ad) cause the common cold, the effect of Ad infection on MDR in cancer has not been established. In this study, we investigated whether Ad infection is a cause of MDR in A549, H441 and HCC827 non-small-cell lung cancer (NSCLC) cell lines, using an Ad vector system. We found that Ad vector infection of NSCLC cell lines induced P-gp mRNA expression, and the extent of induction was dependent on the number of Ad vector virus particles and the infection time. Heat-treated Ad vector, which is not infectious, did not alter P-gp mRNA expression. Uptake experiments with doxorubicin (DOX), a P-gp substrate, revealed that DOX accumulation was significantly decreased in Ad vector-infected A549 cells. The decrease of DOX uptake was blocked by verapamil, a P-gp inhibitor. Our results indicated that Ad vector infection of NSCLC cells caused MDR mediated by P-gp overexpression. The Ad vector genome sequence is similar to that of human Ad, and therefore human Ad infection of lung cancer patients may lead to chemoresistance in the clinical environment.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Adenoviridae Infections/complications , Antibiotics, Antineoplastic/pharmacology , Carcinoma, Non-Small-Cell Lung/complications , Doxorubicin/pharmacology , Drug Resistance, Multiple , Lung Neoplasms/complications , Adenoviridae/genetics , Adenoviridae/isolation & purification , Adenoviridae Infections/genetics , Antibiotics, Antineoplastic/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Doxorubicin/pharmacokinetics , Drug Resistance, Neoplasm , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Up-Regulation
3.
Int J Gynecol Cancer ; 23(2): 235-43, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23314283

ABSTRACT

OBJECTIVE: The purpose of this study was to investigate the clinical significance of DNA methylation of the human papillomavirus (HPV) genome as a prognostic biomarker for cervical intraepithelial neoplasia (CIN). METHODS AND MATERIALS: Clinical samples (paraffin-embedded tissues obtained by conization/hysterectomy or initial punch biopsy) were collected from patients at the Gynecologic Oncology of the Hyogo Cancer Center with informed consent. We evaluated the methylation status of the L1 gene of the HPV genome by bisulfite sequencing, calculating the methylation ratio (L1MR) as (number of methylated CpGs in the analyzed region of the L1 gene) / (number of all CpGs in the analyzed region of the L1 gene) × 100. The methylation analysis and in situ hybridization were performed with serial tissue-section slices. RESULTS: DNA methylation was observed in the L1 gene, but not in the long control region of HPV-16, -18, or the other high-risk HPV types including HPV-31, -52, and -58. L1MR was associated with the CIN grade; the median L1MR was 2.3%, 11.2%, 35.2%, and 50.0% for CIN1, CIN2, CIN3, and squamous cell carcinoma, respectively. L1MRs also seemed to indicate physical status (integrated or episomal form) of the HPV genome in the host cell. L1MR of the progression group was significantly higher than that of the regression group. CONCLUSIONS: L1MR was associated with the CIN grade and indicated the HPV genome status in the host cell: high L1MR indicated HPV genome integration linked to progression from early-stage CINs, whereas low L1MR indicated an episomal HPV genome location in host cells. L1MR may be a prognostic indicator of CIN.


Subject(s)
Capsid Proteins/genetics , DNA Methylation , Oncogene Proteins, Viral/genetics , Uterine Cervical Dysplasia/diagnosis , Uterine Cervical Neoplasms/diagnosis , Cell Line, Tumor , CpG Islands , DNA Methylation/physiology , DNA, Viral/analysis , DNA, Viral/genetics , Female , Gene Expression Regulation, Viral/physiology , Humans , In Situ Hybridization , Papillomavirus Infections/complications , Papillomavirus Infections/diagnosis , Papillomavirus Infections/genetics , Papillomavirus Infections/virology , Polymerase Chain Reaction , Prognosis , Risk Factors , Uterine Cervical Neoplasms/etiology , Uterine Cervical Neoplasms/virology , Uterine Cervical Dysplasia/etiology , Uterine Cervical Dysplasia/virology
4.
Breast Cancer Res Treat ; 132(1): 165-73, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21594664

ABSTRACT

Aberrant promoter methylation of genes is a common molecular event in breast cancer. Thus, DNA methylation analysis is expected to be a new tool for cancer diagnosis. In this article, we have established a new, high-performance DNA methylation assay, the one-step methylation-specific polymerase chain reaction (OS-MSP) assay, which is optimized for analyzing gene methylation in serum DNA. The OS-MSP assay is designed to detect aberrant promoter methylation of GSTP1, RASSF1A, and RARß2 genes in serum DNA. Moreover, two quality control markers were designed for monitoring the bisulfite conversion efficiency and measuring the DNA content in the serum. Serum samples were collected from patients with primary (n = 101, stages I-III) and metastatic breast cancers (n = 58) as well as from healthy controls (n = 87). If methylation of at least one of the three genes was observed, the OS-MSP assay was considered positive. The sensitivity of this assay was significantly higher than that of the assay involving conventional tumor markers (CEA and/or CA15-3) for stages I (24 vs. 8%) and II (26 vs. 8%) breast cancer and similar to that of the assay involving the conventional tumor markers for stage III (18 vs. 19%) and metastatic breast cancers (55 vs. 59%). The results of the OS-MSP assay and those of the assay involving CEA and/or CA15-3 seemed to compensate for each other because sensitivity of these assays increased to 78% when used in combination for metastatic breast cancer. In conclusion, we have developed a new OS-MSP assay with improved sensitivity and convenience; thus, this assay is more suitable for detecting aberrant promoter methylation in serum DNA. Moreover, the combination of the OS-MSP assay and the assay involving CEA and/or CA15-3 is promising for enhancing the sensitivity of diagnosis of metastatic breast cancer.


Subject(s)
Biomarkers, Tumor/blood , Breast Neoplasms/diagnosis , Carcinoma, Ductal, Breast/diagnosis , DNA/blood , Glutathione S-Transferase pi/blood , Receptors, Retinoic Acid/blood , Tumor Suppressor Proteins/blood , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Breast Neoplasms/blood , Breast Neoplasms/pathology , Carcinoembryonic Antigen/blood , Carcinoma, Ductal, Breast/blood , Carcinoma, Ductal, Breast/secondary , Case-Control Studies , DNA/genetics , DNA Methylation , Female , Genetic Association Studies , Glutathione S-Transferase pi/genetics , Humans , Lymphatic Metastasis , Middle Aged , Molecular Diagnostic Techniques , Mucin-1/blood , Neoplasm Staging , Polymerase Chain Reaction/methods , Promoter Regions, Genetic , Receptors, Retinoic Acid/genetics , Sensitivity and Specificity , Sequence Analysis, DNA , Tumor Suppressor Proteins/genetics
5.
Am J Physiol Heart Circ Physiol ; 300(3): H1021-31, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21193589

ABSTRACT

Portal hypertension, a major complication of cirrhosis, is caused by both increased portal blood flow due to arterial vasodilation and augmented intrahepatic vascular resistance due to sinusoidal constriction. In this study, we examined the possible involvement of resident macrophages in the tone regulation of splanchnic blood vessels using bile duct ligated (BDL) portal hypertensive rats and an in vitro organ culture method. In BDL cirrhosis, the number of ED2-positive resident macrophages increased by two- to fourfold in the vascular walls of the mesenteric artery and extrahepatic portal vein compared with those in sham-operated rats. Many ED1-positive monocytes were also recruited into this area. The expression of inducible nitric oxide (NO) synthase (iNOS) mRNA was increased in the vascular tissues isolated from BDL rats, and accordingly, nitrate/nitrite production was increased. Immunohistochemistry revealed that iNOS was largely expressed in ED1-positive and ED2-positive cells. We further analyzed the effect of iNOS expression on vascular smooth muscle contraction using an in vitro organ culture system. iNOS mRNA expression and nitrate production significantly increased in vascular tissues (without endothelium) incubated with 1 µg/ml lipopolysaccharide (LPS) for 6 h. Immunohistochemistry indicated that iNOS was largely expressed in ED2-positive resident macrophages. α-Adrenergic-stimulated contractility of the mesenteric artery was greatly suppressed by LPS treatment and was restored by N(G)-nitro-L-arginine methyl ester (NO synthase inhibitor); in contrast, portal vein contractility was largely unaffected by LPS. Sodium nitroprusside (NO donor) and 8-bromo-cGMP showed greater contractile inhibition in the mesenteric artery than in the portal vein with decreasing myosin light chain phosphorylation. In the presence of an α-adrenergic agonist, the mesenteric artery cytosolic Ca(2+) level was greatly reduced by sodium nitroprusside; however, the portal vein Ca(2+) level was largely unaffected. These results suggest that the induction of iNOS in monocytes/macrophages contributes to a hypercirculatory state in the cirrhosis model rat in which the imbalance of the responsiveness of visceral vascular walls to NO (mesenteric artery >> portal vein) may account for the increased portal venous flow in portal hypertension.


Subject(s)
Hypertension, Portal/physiopathology , Macrophages/enzymology , Muscle Contraction/physiology , Muscle, Smooth, Vascular/physiopathology , Nitric Oxide Synthase Type II/physiology , Splanchnic Circulation/physiology , Animals , Calcium/physiology , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Hypertension, Portal/enzymology , Lipopolysaccharides/pharmacology , Liver Cirrhosis/enzymology , Liver Cirrhosis/physiopathology , Macrophages/physiology , Male , Mesenteric Arteries/enzymology , Mesenteric Arteries/physiopathology , Monocytes/enzymology , Monocytes/physiology , Muscle, Smooth, Vascular/enzymology , Myosin Light Chains/metabolism , Myosin Light Chains/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Nitrates/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitroprusside/pharmacology , Portal Vein/enzymology , Portal Vein/physiopathology , Rats , Rats, Sprague-Dawley , Vasodilator Agents/pharmacology
6.
Mol Endocrinol ; 21(12): 2907-18, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17761946

ABSTRACT

The estrogen receptor (ER)-alpha (ESR1) is a key regulatory molecule in mammary epithelial cell development. Loss of ER-alpha in breast cancer is correlated with poor prognosis, increased recurrence after treatment, and an elevated incidence of metastasis. A proposed molecular pathway by which ER-alpha acts to constrain invasive growth in breast cancer cells involves direct, ER-alpha-dependent expression of metastasis-associated protein 3, a cell-type-specific component of the Mi-2/NuRD chromatin remodeling complex. MTA3 in turn represses expression of Snail, a transcription factor linked to epithelial to mesenchymal transition and cancer metastasis. To elucidate its role(s) in epithelial to mesenchymal transition (EMT), we expressed Snail in the noninvasive, ER-alpha-positive MCF-7 cell line. Snail expression led to decreased cell-cell adhesion and increased cell invasiveness. Furthermore, we observed loss of ER-alpha expression at both the RNA and protein level that was accompanied by direct interaction of Snail with regulatory DNA sequences at the ESR1 locus. A consequence of loss of ER-alpha function in this system was the increased abundance of key components of the TGF-beta signaling pathway. Thus, cross-talk among ER-alpha, Snail, and the TGF-beta pathway appears to control critical phenotypic properties of breast cancer cells.


Subject(s)
Cell Differentiation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Estrogen Receptor alpha/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Transcription Factors/metabolism , Acetylation , Breast Neoplasms/metabolism , Cell Line, Tumor , Down-Regulation , Estrogen Receptor alpha/genetics , Histones/metabolism , Humans , Introns/genetics , Microarray Analysis , Phenotype , Promoter Regions, Genetic/genetics , Protein Binding , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription, Genetic/genetics , Transforming Growth Factor beta/metabolism
7.
Arch Anim Nutr ; 61(1): 31-41, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17361946

ABSTRACT

Responses of whole body protein synthesis (WBPS) and glucose irreversible loss rate (ILR) were compared between dietary starch and sucrose in four male goats. Diets were fed at 1.2 times maintenance requirements of ME and CP with 30% of the ME as starch, starch plus sucrose or sucrose, twice daily. The diets consisted of 33, 32, 11 and 24% of alfalfa hay, corn, soybean meal and the carbohydrates, respectively. The WBPS and glucose ILR during 5-7 h after feeding were determined by an isotope dilution method of [2H5]phenylalanine, [2H2]tyrosine, [2H4]tyrosine and [13C6]glucose. Sucrose elevated ammonia nitrogen and lowered acetate concentrations in the rumen, but did not differ from starch in nitrogen retention. Glucose ILR and WBPS were similar between the carbohydrates. It was concluded that dietary sucrose would have effects similar to starch on WBPS and glucose kinetics in the absorptive state in goats fed a high-concentrate diet.


Subject(s)
Dietary Sucrose/pharmacology , Glucose/pharmacokinetics , Goats/metabolism , Proteins/metabolism , Starch/pharmacology , Animal Nutritional Physiological Phenomena , Animals , Carbon Isotopes , Deuterium , Dietary Carbohydrates/pharmacology , Dietary Proteins/metabolism , Dietary Supplements , Energy Intake/physiology , Energy Metabolism , Male , Nutritional Requirements , Protein Biosynthesis/drug effects
8.
Article in English | MEDLINE | ID: mdl-16580858

ABSTRACT

An isotope dilution experiment was conducted to determine the effect of metabolizable energy intake (MEI) as starch on whole body protein synthesis (WBPS), nitrogen (N) retention and glucose irreversible loss rate (ILR) in four adult goats (Capra hircus). The goats were fed isonitrogenous diets containing three different metabolizable energy (1.0, 1.5 and 2.0 times maintenance) twice daily. Energy above maintenance was supplemented with cornstarch. The WBPS and glucose ILR during 5 to 7 h after feeding were measured by a primed-continuous infusion of [2H5]phenylalanine, [2H2]tyrosine, [2H4]tyrosine and [13C6]glucose for 4 h, with measurements of plasma concentrations of metabolites and insulin. Ruminal characteristics were also determined. Increasing MEI improved N retention, despite decreased digestible N. Increasing MEI decreased ruminal pH and ammonia nitrogen. In plasma, decreased urea N, increased total amino N and tyrosine, and trends for increases in phenylalanine and insulin resulted from increasing MEI. Increasing MEI increased ILR of glucose, phenylalanine and tyrosine, and hydroxylation rate of phenylalanine and WBPS. We conclude that in goats increasing MEI as starch enhances WBPS in the absorptive state and N retention, despite a decrease in digestible N. These changes are probably associated with both decreased ammonia absorption and increased amino acid absorption.


Subject(s)
Glucose/metabolism , Goats/metabolism , Nitrogen/metabolism , Protein Biosynthesis/physiology , Starch/pharmacology , Animals , Blood Urea Nitrogen , Butyrates/blood , Dietary Supplements , Energy Intake/physiology , Energy Metabolism , Fatty Acids/blood , Insulin/blood , Isotopes/pharmacology , Lactic Acid/blood , Male , Phenylalanine/blood , Propionates/blood , Tyrosine/blood
9.
Mol Endocrinol ; 18(12): 2937-49, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15358836

ABSTRACT

Metastasis-associated protein 3 (MTA3) is a cell type-specific subunit of the Mi-2/NuRD transcriptional corepressor complex. In breast cancer cells, MTA3 and the Mi-2/NuRD complex mediate repression of Snail, a transcription factor that promotes epithelial to mesenchymal transitions. Thus, MTA3 functions to maintain a differentiated, epithelial status in breast cancer. Interestingly, in mammary epithelial cells, MTA3 biosynthesis requires both functional estrogen receptor (ER) and estradiol. Here we have investigated the molecular basis for estrogen and ER-dependent expression of MTA3 in breast cancer cells. Molecular dissection of the MTA3 promoter using transient transfection assays identified a composite element required for high-level transcription consisting of an SP1 site in close proximity to a consensus estrogen response element half-site. Depletion of either SP1 or ER-alpha by RNA interference led to loss of MTA3 transcript in multiple breast cancer cell lines, indicating a requirement for both transcription factors in expression of endogenous MTA3. The MTA3 gene thus joins a growing list of loci regulated by both SP1 and ER.


Subject(s)
Breast Neoplasms/genetics , Estrogens/physiology , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Response Elements/genetics , Sp1 Transcription Factor/physiology , Base Sequence , Breast Neoplasms/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor alpha/physiology , Estrogens/pharmacology , Female , Genes, Reporter/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Luciferases/analysis , Luciferases/genetics , Molecular Sequence Data , Neoplasm Proteins/metabolism , RNA Interference , RNA, Messenger/analysis , RNA, Messenger/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Trans-Activators , Transcription, Genetic
10.
Mol Cell Biol ; 24(17): 7559-66, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15314165

ABSTRACT

Snail and Slug are closely related transcriptional repressors involved in embryonic patterning during metazoan development. In human cancer, aberrant expression of Snail and/or Slug has been correlated with invasive growth potential, a property primarily attributed to their ability to directly repress transcription of genes whose products are involved in cell-cell adhesion, such as E-cadherin, occludin, and claudins. To investigate the molecular mechanisms of alterations in epithelial cell fate mediated by aberrant expression of Snail or Slug, we analyzed the consequences of exogenous expression of these factors in human cancer cells. Aberrant expression of either Snail or Slug led to changes in cell morphology, the loss of normal cell-cell contacts, and the acquisition of invasive growth properties. Snail or Slug expression also promoted resistance to programmed cell death elicited by DNA damage. Detailed molecular analysis revealed direct transcriptional repression of multiple factors with well-documented roles in programmed cell death. Depletion of endogenous Snail by RNA interference led to increased sensitivity to DNA damage accompanied by increased expression of the proapoptotic factors identified as targets of Snail. Thus, aberrant expression of Snail or Slug may promote tumorigenesis through increased resistance to programmed cell death.


Subject(s)
DNA Damage , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Transcription Factors/metabolism , Animals , Apoptosis/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic , DNA-Binding Proteins/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , In Situ Nick-End Labeling , Phenotype , RNA Interference , Snail Family Transcription Factors , Transcription Factors/genetics
11.
J Vet Med Sci ; 66(4): 409-15, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15133271

ABSTRACT

Several studies have indicated that viruses require a specific cytoskeletal structure for replication in host cells. In this study, we examined the role of actin fiber in the replication of canine distemper virus (CDV), belonging to the Morbillivirus genus of the family Paramyxoviridae. For this purpose, we used two actin depolymerizing agents, cytochalasin-D (C-D) and mycalolide-B (ML-B). In Vero cells, C-D disrupted actin fibers distributed in the cytosol, but peripheral actin fibers remained intact. On the other hand, ML-B completely disrupted the actin fibers distributed in both areas. Treatment of Vero cells with C-D or ML-B inhibited the replication of CDV. Double staining of CDV-infected Vero cells with antibody to N-protein and rhodamine-phalloidin revealed the presence of N-protein in mid-cytoplasm. However, the N-protein was specifically localized at the submembrane region in the presence of C-D, whereas it was clustered in the presence of ML-B. Viral mRNA levels of N- and H-proteins were rather increased by treatment with C-D or ML-B. The treatment with ML-B strongly inhibited N-protein expression, whereas C-D only slightly inhibited N-protein expression. These results suggest that actin microfilaments distributed in the cytoplasm and on the membrane region in host cells may have a different role in the process of CDV replication.


Subject(s)
Actin Cytoskeleton/physiology , Cytochalasin D/pharmacology , Distemper Virus, Canine/physiology , Oxazoles/pharmacology , RNA, Messenger/metabolism , Virus Replication/drug effects , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Cell Membrane/metabolism , Chlorocebus aethiops , Cytoplasm/metabolism , DNA Primers , Distemper Virus, Canine/genetics , Electrophoresis, Agar Gel , Fluorescent Antibody Technique , Marine Toxins , Nucleocapsid Proteins , Vero Cells , Virus Replication/physiology
12.
Biochim Biophys Acta ; 1677(1-3): 52-7, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15020045

ABSTRACT

The vertebrate Mi-2/NuRD complex is a multi-subunit protein complex containing both histone deacetylase and nucleosome-dependent ATPase subunits. Current models predict that this complex functions primarily in transcriptional repression. Surprisingly, every subunit of this complex presents heterogeneity at the protein and gene level. This raises the intriguing possibility of functional specialization resulting from incorporation of unique gene products into the complex. The MTA (metastasis-associated) proteins represent one class of alternative subunits of the human Mi-2/NuRD complex. The members of this family in human cells are differentially expressed depending on cell type and on physiologic parameters. We summarize evidence supporting the view that the alternative subunits of the complex that have arisen during vertebrate evolution endow unique functional properties.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Helicases/metabolism , Histone Deacetylases/metabolism , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Animals , Autoantigens/genetics , Autoantigens/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA Helicases/genetics , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Histone Deacetylases/genetics , Humans , Macromolecular Substances , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Molecular Sequence Data , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phylogeny , Protein Isoforms , Repressor Proteins/genetics , Repressor Proteins/metabolism , Trans-Activators , Transcription Factors
13.
Oncogene ; 22(54): 8716-22, 2003 Nov 27.
Article in English | MEDLINE | ID: mdl-14647466

ABSTRACT

Membrane-type 1 matrix metalloproteinase (MT1-MMP/MMP-14) has been believed a key enzyme in tumor invasion, because it is expressed in a variety of malignant human tumors, and overexpression of the enzyme enhances the ability of cellular invasiveness. However, it has not necessarily been clarified whether the endogenously expressed MT1-MMP in human tumors plays a critical role in their invasiveness. We used RNA silencing technology to downregulate the endogenous MT1-MMP expression in human tumor cells (fibrosarcoma HT1080 and gastric carcinoma MKN-28 cell lines), and evaluated the effect on the invasion of a reconstituted basement membrane (Matrigel). Transfection of a double-stranded RNA targeted to the MT1-MMP gene decreased the level of the enzyme to less than 10-20% without affecting production of other MMPs. According to the degree of silencing, activation of proMMP-2 was inhibited. CD44 shedding was also inhibited, but only in part. Decreased MT1-MMP levels were also reflected in reduced cell motility on hyaluronan (HA) and invasion in Matrigel. Thus, specific downregulation of MT1-MMP expression was sufficient to cause significant inhibition of the migration and invasion of tumor cells, even though other MMPs continued to be expressed.


Subject(s)
Gene Silencing , Metalloendopeptidases/antagonists & inhibitors , Neoplasms/therapy , Cell Line, Tumor , Cell Movement , Enzyme Activation , Enzyme Precursors/metabolism , Gelatinases/metabolism , Humans , Hyaluronan Receptors/physiology , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Metalloendopeptidases/physiology , Neoplasm Invasiveness , RNA, Double-Stranded/physiology , RNA, Messenger/analysis , Stomach Neoplasms/pathology
14.
Biochem Soc Symp ; (70): 253-62, 2003.
Article in English | MEDLINE | ID: mdl-14587298

ABSTRACT

Membrane-type 1 matrix metalloproteinase (MT1-MMP) is an integral membrane proteinase that performs processing of cell surface proteins and degradation of extracellular matrix (ECM) components. Through these proteolytic events, MT1-MMP regulates various cellular functions, including ECM turnover, promotion of cell migration and invasion, and morphogenic responses to extracellular stimuli. MT1-MMP has to be regulated strictly to accomplish its function appropriately at various steps, including at the transcriptional and post-translational levels. MT1-MMP was originally identified as an invasion-promoting enzyme expressed in malignant tumour cells, and also as a specific activator of proMMP-2, which is believed to play a role in invasion of the basement membrane. Since then, it has attracted attention as a membrane-associated MMP that promotes cancer cell invasion and angiogenesis by endothelial cells. Although MT1-MMP has now become one of the best characterized enzymes in the MMP family, there remain numerous unanswered questions. In this chapter, we summarize our recent findings on how MT1-MMP is regulated during cell migration, and how cell migration is regulated by MT1-MMP.


Subject(s)
Cell Movement/physiology , Matrix Metalloproteinase 1/physiology , Endocytosis , Hyaluronan Receptors/physiology
15.
Cell ; 113(2): 207-19, 2003 Apr 18.
Article in English | MEDLINE | ID: mdl-12705869

ABSTRACT

Estrogen receptor is a key regulator of proliferation and differentiation in mammary epithelia and represents a crucial prognostic indicator and therapeutic target in breast cancer. Mechanistically, estrogen receptor induces changes in gene expression through direct gene activation and also through the biological functions of target loci. Here, we identify the product of human MTA3 as an estrogen-dependent component of the Mi-2/NuRD transcriptional corepressor in breast epithelial cells and demonstrate that MTA3 constitutes a key component of an estrogen-dependent pathway regulating growth and differentiation. The absence of estrogen receptor or of MTA3 leads to aberrant expression of the transcriptional repressor Snail, a master regulator of epithelial to mesenchymal transitions. Aberrant Snail expression results in loss of expression of the cell adhesion molecule E-cadherin, an event associated with changes in epithelial architecture and invasive growth. These results establish a mechanistic link between estrogen receptor status and invasive growth of breast cancers.


Subject(s)
Breast Neoplasms/genetics , Carcinoma/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/genetics , Histone Deacetylases/metabolism , Neoplasm Proteins/metabolism , Receptors, Estrogen/metabolism , Amino Acid Sequence/genetics , Base Sequence/genetics , Breast Neoplasms/metabolism , Cadherins/genetics , Cadherins/metabolism , Carcinoma/metabolism , Cell Differentiation/genetics , Cell Division/genetics , Cell Transformation, Neoplastic/metabolism , DNA Mutational Analysis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , HeLa Cells , Histone Deacetylases/genetics , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Molecular Sequence Data , Neoplasm Proteins/genetics , Receptors, Estrogen/genetics , Repressor Proteins/genetics , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
16.
EMBO J ; 21(15): 3949-59, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12145196

ABSTRACT

Membrane-type 1 matrix metalloproteinase (MT1- MMP) localizes at the front of migrating cells and degrades the extracellular matrix barrier during cancer invasion. However, it is poorly understood how the polarized distribution of MT1-MMP at the migration front is regulated. Here, we demonstrate that MT1-MMP forms a complex with CD44H via the hemopexin-like (PEX) domain. A mutant MT1-MMP lacking the PEX domain failed to bind CD44H and did not localize at the lamellipodia. The cytoplasmic tail of CD44H, which comprises interfaces that associate with the actin cytoskeleton, was important for its localization at lamellipodia. Overexpression of a CD44H mutant lacking the cytoplasmic tail also prevented MT1-MMP from localizing at the lamellipodia. Modulation of F-actin with cytochalasin D revealed that both CD44H and MT1-MMP co-localize closely with the actin cytoskeleton, dependent on the cytoplasmic tail of CD44H. Thus, CD44H appears to act as a linker that connects MT1-MMP to the actin cytoskeleton and to play a role in directing MT1-MMP to the migration front. The PEX domain of MT1-MMP was indispensable in promoting cell migration and CD44H shedding.


Subject(s)
Hyaluronan Receptors/physiology , Metalloendopeptidases/metabolism , Pseudopodia/metabolism , Actins/analysis , Animals , CHO Cells/cytology , Cell Membrane/metabolism , Cell Movement/physiology , Chlorocebus aethiops , Cricetinae , Cricetulus , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Fibroblasts/cytology , Hemopexin/chemistry , Humans , Hyaluronan Receptors/chemistry , Hyaluronan Receptors/genetics , Macromolecular Substances , Matrix Metalloproteinases, Membrane-Associated , Protein Interaction Mapping , Protein Structure, Tertiary , Protein Transport , Recombinant Fusion Proteins/physiology , Sequence Deletion , Structure-Activity Relationship , Transfection , Tumor Cells, Cultured/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...