Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
1.
Birth Defects Res B Dev Reprod Toxicol ; 95(6): 386-94, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23169613

ABSTRACT

BACKGROUND: Psoralea corylifolia L. (PC) was commonly used to treat miscarriages clinically. The aim of this study was to examine its embryotoxicity in mice and embryonic stem cells (ESCs). METHODS: Quality control of PC extract including reference marker compounds, pesticide residues, and heavy metals was authenticated with HPLC, Gas chromatography-mass spectrometry (GC-MS), and inductively coupled plasma-mass spectrometry. Pregnant mice were randomly assigned into five groups and dosed with distilled water (G1), PC extract of 2 (G2), 4 (G3), or 8 g/kg/day (G4), and vitamin A (G5). Meanwhile, half maximal inhibitory concentration values for ESCs and 3T3 cells were identified in a cytotoxicity assay, and apoptosis in neuroepithelium was assessed by transmission electron microscopy. RESULTS: In the G4 group, a statistically significant decrease in the total fetus, live fetus, and gravid uterine weight, and increase in the resorbed fetus, postimplantation loss, and neuroepithelial apoptosis as well as maternal liver-weight were found (p < 0.05). CONCLUSIONS: PC extracts at 8 g/kg/day might cause fetal toxicity and maternal liver damage in mice, although it did not cause typical malformation and ESC's cytotoxicity in this experiment. Our data suggested that high dosage and long-term administration of PC preparations may not be safe for pregnant women.


Subject(s)
Embryonic Development/drug effects , Fetal Development/drug effects , Maternal Exposure/adverse effects , Plant Extracts/toxicity , Psoralea/chemistry , Teratogens/toxicity , 3T3 Cells/drug effects , 3T3 Cells/pathology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Embryo, Nonmammalian/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/pathology , Female , Fetal Resorption/chemically induced , Fetal Weight/drug effects , Gas Chromatography-Mass Spectrometry , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred ICR , Neuroepithelial Cells/drug effects , Neuroepithelial Cells/pathology , Neuroepithelial Cells/ultrastructure , Organ Size/drug effects , Plant Extracts/analysis , Plant Extracts/classification , Pregnancy , Teratogens/classification , Uterus/drug effects , Uterus/pathology , Vitamin A/toxicity
2.
BMC Musculoskelet Disord ; 13: 94, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22682502

ABSTRACT

BACKGROUND: It has been indicated that moderate or high dose of X-irradiation could delay fracture union and cause osteoradionecrosis, in part, mediated by its effect on proliferation and differentiation of osteoblasts. However, whether low dose irradiation (LDI) has similar roles on osteoblasts is still unknown. In this study, we investigated whether and to what extent LDI could affect the proliferation, differentiation and mineralization of osteoblasts in vitro. METHODS: The MC3T3-E1 cells were exposed to single dose of X-irradiation with 0, 0.1, 0.5, 1.0 Gy respectively. Cell proliferation, apoptosis, alkaline phosphatase (ALP) activity, and mineralization was evaluated by methylthiazoletetrazolium (MTT) and bromodeoxyuridine (BrdU) assay, flow cytometry, ALP viability kit and von Kossa staining, respectively. Osteocalcin (OCN) and core-binding factor α1 (Cbfα1) expressions were measured by real time-PCR and western blot, respectively. RESULTS: The proliferation of the cells exposed to 2.0 Gy was significantly lower than those exposed to ≤1.0 Gy (p < 0.05) from Day 4 to Day 8, measured by MTT assay and BrdU incorporation. For cells exposed to ≤1.0 Gy, increasing dosages of X-irradiation had no significant effect on cell proliferation and apoptosis. Importantly, LDI of 0.5 and 1 Gy increased ALP activities and mineralized nodules of MC3T3-E1 cells. In addition, mRNA and protein expressions of OCN and Cbfα1 were also markedly increased after treatment with LDI at 0.5 and 1 Gy. CONCLUSIONS: LDI have different effects on proliferation and differentiation of osteoblasts from those of high dose of X-irradiation, which might suggest that LDI could lead to promotion of fracture healing through enhancing the differentiation and mineralization of osteoblasts.


Subject(s)
Calcification, Physiologic/radiation effects , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Osteoblasts/radiation effects , 3T3 Cells/metabolism , 3T3 Cells/pathology , 3T3 Cells/radiation effects , Alkaline Phosphatase/metabolism , Animals , Apoptosis/radiation effects , Bromodeoxyuridine/metabolism , Calcium/analysis , Calcium/metabolism , Cell Line , Cell Survival/radiation effects , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Dose-Response Relationship, Radiation , Gene Expression/radiation effects , Mice , Osteoblasts/metabolism , Osteoblasts/pathology , Osteocalcin/genetics , Osteocalcin/metabolism , RNA, Messenger/metabolism , Tetrazolium Salts/metabolism , Thiazoles/metabolism
3.
Food Chem Toxicol ; 47(10): 2401-6, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19520133

ABSTRACT

In this study, we investigated the effect of sulfated glucosamine (SGlc) on adipogenesis of 3T3-L1 adipocytes during differentiation of preadipocytes into adipocytes by measuring lipid accumulation and adipogenesis related factors. Treatment with SGlc reduced the triglyceride content in Oil-Red O staining and enhanced glycerol secretion in adipocytes in a dose-dependent manner. In addition, SGlc induced the down-regulation of adipogenesis related factors and adipocyte specific gene promoters. Moreover, treatment of 3T3-L1 adipocytes with SGlc activated the phosphorylated adenosine monophosphate-activated protein kinase (AMPK) alpha and beta along with their substrate, acetyl-CoA carboxylase (ACC). These results suggest that inhibitory effect of SGlc on adipocyte differentiation might be mediated through the up-regulation of AMPK pathway.


Subject(s)
3T3 Cells/drug effects , AMP-Activated Protein Kinases/drug effects , Adipocytes/drug effects , Adipogenesis/drug effects , Anti-Obesity Agents/pharmacology , Glucosamine/analogs & derivatives , 3T3 Cells/metabolism , 3T3 Cells/pathology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Adipogenesis/genetics , Animals , Carrier Proteins , Cell Differentiation/drug effects , Gene Expression Regulation/drug effects , Glucosamine/pharmacology , Glycerol/metabolism , Lipid Metabolism/drug effects , Mice , Perilipin-1 , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Messenger/genetics , Signal Transduction/drug effects , Sterol Esterase/genetics , Sterol Esterase/metabolism , Triglycerides/metabolism
4.
Polim Med ; 38(2): 21-31, 2008.
Article in English, Polish | MEDLINE | ID: mdl-18810984

ABSTRACT

INTRODUCTION: The aim of this study was to evaluate polyasparagine (PAA), a new, promising scaffold. PAA was compared with alginate, which is used in cell transplantations and may be regarded as a standard. MATERIALS AND METHODS: In vitro cell viability on both scaffolds was assessed. C57B1 mice were injected s. c. alginate or PAA with or without 3T3 cells. After two months specimens from sites of injection were examined and blood samples were taken for enzymatic activity estimation. Cathepsin D activity and alpha1-antitrypsin levels were measured. RESULTS: In vitro cell viability was lowest on PAA and highest in control group. Increase in levels of enzymes measured was observed in response to PAA and alginate and was lower in case of polymer seeded with cells. Increase in alpha1-antitrypsin levels was lower in case of PAA in comparison to alginate. Scaffold degradation in histopathological specimens was visible. CONCLUSION: The results indicate that PAA implants undergo biodegradation and nonspecific inflammatory response is comparable to alginate.


Subject(s)
Cell Culture Techniques/methods , Materials Testing , Peptides , Tissue Scaffolds , 3T3 Cells/metabolism , 3T3 Cells/pathology , Alginates , Animals , Cathepsin D/metabolism , Cell Proliferation , Cell Survival , Cells, Cultured , Glucuronic Acid , Hexuronic Acids , Mice , Mice, Inbred C57BL , Trypsin/metabolism
5.
Chem Res Toxicol ; 20(10): 1543-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17824666

ABSTRACT

There is a great interest in characterizing the biological properties of natural compounds obtained from plants, especially polyphenols. We studied the structure-activity-cytotoxicity relationships of polyphenolic fractions obtained from grape pomace and pine bark. These fractions contained similar polymerised flavonoids but different percentages of pyrogallol groups that confer on them different biological properties. The human keratinocyte cell line HaCaT and the mouse fibroblast cell line 3T3 were used to study the cytotoxicity of the different fractions after 24, 48, and 72 h of exposure. Antioxidant activity of the fractions was evaluated by measuring the inhibition of hemolysis mediated by AAPH. Our results demonstrate that the polyphenolic fractions studied show high antioxidant capacity in a concentration range that is not harmful to normal human cells. Pine fractions presented slightly lower antioxidant activity than grape fractions but are less cytotoxic. This data provides useful information to help design safe antioxidant products that act without altering critical cell functions.


Subject(s)
Antioxidants/pharmacology , Biflavonoids/pharmacology , Catechin/pharmacology , Pinus/chemistry , Proanthocyanidins/pharmacology , Vitis/chemistry , 3T3 Cells/drug effects , 3T3 Cells/pathology , Amidines/pharmacology , Animals , Antioxidants/chemistry , Biflavonoids/chemistry , Catechin/chemistry , Cell Line, Transformed , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemical Fractionation , Erythrocytes/drug effects , Fibroblasts/drug effects , Fibroblasts/pathology , Flavonoids/chemistry , Flavonoids/pharmacology , Hemolysis/drug effects , Hemolysis/physiology , Humans , Keratinocytes/drug effects , Keratinocytes/pathology , Mice , Phenols/chemistry , Phenols/pharmacology , Plant Bark/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , Polyphenols , Proanthocyanidins/chemistry , Structure-Activity Relationship
6.
Altern Lab Anim ; 34(2): 151-75, 2006 May.
Article in English | MEDLINE | ID: mdl-16704290

ABSTRACT

A database of over 280 chemicals has been compiled by using a mouse 3T3-L1 fibroblast-like cell line in exponential growth, exposed to chemicals for 72 hours in a 96-well tissue culture plate format, and determining cell number via the Kenacid blue (KB) assay for total protein. Ranking the chemicals according to their basal cytotoxicity, expressed as the concentration (mM) that inhibits increase in total cellular protein over 72 hours by 50% (the ID50 value) shows a wide range of ID50 values, from 0.00003 mM to 10,096 mM. This information includes the results for MEIC chemicals 1-50, and we have now added basal cytotoxicity data for 23 of the next 25 MEIC chemicals. When the neutral red uptake (NRU) assay was performed with the same cell cultures, before the KB assay, very similar indications of basal cytotoxicity were obtained. Comparisons between the results with 3T3-L1 cells and with a human fibroblast-like cell line, BCL-D1 showed a significant difference in order of magnitude of the ID50 value for only 5 of 52 chemicals. However, there was a difference in ID50 value of more than one order of magnitude for 8 of 24 chemicals tested with an undifferentiated teratocarcinoma cell line, F9.


Subject(s)
Animal Testing Alternatives , Databases, Factual , Toxicity Tests/methods , 3T3 Cells/drug effects , 3T3 Cells/metabolism , 3T3 Cells/pathology , Animals , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Mice , Neutral Red , Organic Chemicals , Proteins/metabolism , Xenobiotics/classification , Xenobiotics/toxicity
7.
Oncogene ; 25(6): 899-906, 2006 Feb 09.
Article in English | MEDLINE | ID: mdl-16247467

ABSTRACT

Galpha(12), the alpha-subunit of G12, which has been referred to as the gep oncogene, stimulates mitogenic pathways in different cell types and readily induces neoplastic transformation of fibroblast cell lines. Recently, we have shown that the oncogenic pathway activated by Galpha(12) involves the receptor tyrosine kinase platelet derived growth factor receptor-alpha (PDGFRalpha) and JAK3. In the present study, we demonstrate that the GTPase-deficient activated mutant of Galpha(12) activates signal transducer and activator of transcription 3 (STAT3) via PDGFRalpha as well as JAK3. Here we show that Galpha(12) stimulates the phosphorylation of STAT3 at both Tyrosine-705 and Serine-727 residues. Studies to delineate the mechanism by which Galpha(12) stimulates STAT3 have indicated that the Tyrosine-705-phosphorylation of STAT3 involves the tyrosine kinases, Janus Kinase-3 as well as Src kinase, whereas the Serine-727 phosphorylation of STAT3 occurs via the receptor tyrosine kinase, PDGFRalpha and phosphatidylinositol 3-OH kinase pathway. Our results also indicate that the coexpression of the dominant negative, DNA binding mutant of STAT3 (STAT3DB) inhibits the foci formation as well as anchorage-independent growth of Galpha(12)QL-transfectants, thereby establishing the critical role of STAT3 in Galpha(12)QL-mediated neoplastic cell growth. The results presented here demonstrate, for the first time, the ability of Galpha(12) to recruit multiple receptor-, nonreceptor-, and Ser/Thr kinases to stimulate STAT3-signaling to promote neoplastic transformation.


Subject(s)
Cell Transformation, Neoplastic/genetics , GTP-Binding Protein alpha Subunits, G12-G13/genetics , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , STAT3 Transcription Factor/metabolism , 3T3 Cells/pathology , Animals , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Janus Kinase 3 , Mice , Mutation , Oncogenes , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Serine/metabolism , Signal Transduction , Tyrosine/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
8.
Oncogene ; 23(34): 5748-58, 2004 Jul 29.
Article in English | MEDLINE | ID: mdl-15195135

ABSTRACT

Epithelial cell adhesion molecule (EpCAM) is a membrane glycoprotein expressed on adenomatous and simple epithelia, where it is involved in homophilic adhesion at the basolateral membrane. Carcinomas strongly overexpress EpCAM through an, as yet, unknown mechanism. Interestingly, otherwise EpCAM-negative squamous epithelia are seen to express EpCAM concomitant with their transformation and de-differentiation. The amount of EpCAM and the number of expressing cells both increase with the grade of dysplasia. Despite an important amount of data correlating the expression of EpCAM with cellular proliferation and de-differentiation, such as the coexpression with Ki-67, a marker for proliferation, it is unknown whether EpCAM may directly contribute to carcinogenesis. Here, we show that EpCAM has a direct impact on cell cycle and proliferation, and the ability to rapidly upregulate the proto-oncogene c-myc and cyclin A/E. Human epithelial 293 cells as well as murine NIH3T3 fibroblasts expressing EpCAM had a decreased requirement for growth factors, enhanced metabolic activity and colony formation capacity. Importantly, the inhibition of EpCAM expression with antisense mRNA led to a strong decrease in proliferation and metabolism in human carcinoma cells. Moreover, domain swapping experiments demonstrated that the intracellular part of EpCAM is necessary and sufficient to transduce the effects described. Thus, the data presented here highlight the role of EpCAM, demonstrating for the first time a direct link to cell cycle and proliferation.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Proto-Oncogene Proteins c-myc/genetics , 3T3 Cells/metabolism , 3T3 Cells/pathology , Animals , Antigens, Neoplasm/genetics , Carcinogenicity Tests , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Cell Adhesion Molecules/genetics , Cell Cycle/physiology , Cell Division/physiology , Cells, Cultured , Cyclin A/genetics , Cyclin A/metabolism , Cyclin E/genetics , Cyclin E/metabolism , Epithelial Cell Adhesion Molecule , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation , Humans , Mice , Protein Structure, Tertiary , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/metabolism , Up-Regulation
9.
Oncogene ; 23(34): 5815-22, 2004 Jul 29.
Article in English | MEDLINE | ID: mdl-15184870

ABSTRACT

The RNA-binding motif (RRM) gene on Y chromosome (RBMY), encoding a male germ cell-specific RNA-binding protein associated with spermatogenesis, was found inserted by hepatitis B virus (HBV) DNA in one childhood hepatocellular carcinoma (HCC). This study is aimed to explore the oncogenic potential of the RBMY protein. The RBMY transcripts, expressed exclusively in the testis of normal people, were detected by reverse transcription-polymerase chain reaction in 36% of HCCs from 90 males and in 67% of hepatoblastoma from six boys. The nontumor liver counter parts, cirrhotic liver tissues from children with biliary atresia, and other types of cancers, such as bile duct, colon, stomach, lung, prostate, and kidney, were all negative for RBMY expression. One to four types of RBMY transcripts, including wild type and variants with N-terminal RRM deletion, C-terminal SRGY (serine-arginine-glycine-tyrosine) boxes deletion, or deletion of both domains, were found in the testis and liver cancer tissues. The wild-type RBMY protein was expressed in the nucleus and demonstrated its tumorigenicity by transformation of mouse fibroblast NIH3T3 cells and in vivo tumor formation. The RBMY variant protein with deletion of C-terminal exons 9-12 was trapped in the cytoplasm and showed decreased tumorigenicity. Our results suggest that RBMY is a new candidate oncogene specific for male liver cancer.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , RNA-Binding Proteins/genetics , Testis/physiology , 3T3 Cells/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Carcinogenicity Tests , Cell Transformation, Neoplastic/genetics , Child , Child, Preschool , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Cirrhosis, Biliary/genetics , Male , Mice , Mice, Inbred BALB C , Middle Aged , Neoplasms/genetics , Nuclear Proteins , Organ Specificity , Reference Values , Sequence Deletion
10.
J Cancer Res Clin Oncol ; 130(5): 301-5, 2004 May.
Article in English | MEDLINE | ID: mdl-15014984

ABSTRACT

PURPOSE: The effects of cigarette smoking on the association between inflammation and cancer were studied, since some bacteria induce the production of tumor necrosis factor-alpha (TNF-alpha), a proinflammatory cytokine and endogenous tumor promoter, in cells. METHODS: Bacteria from a gargled solution from the buccal cavity of 20 individuals were cultured in the presence of 4 mg/ml cigarette-smoke condensates. Although cigarette-smoke condensates inhibited growth of Staphylococcus aureus strongly and that of Staphylococcus warneri weakly, tobacco tar-resistant S. aureus and S. warneri were obtained. RESULTS: One tobacco tar-resistant S. aureus strain (Sa-TA10) induced expression of the TNF-alpha gene in both Bhas 42 cells (v-Ha-ras transfected BALB/3T3 cells) and human lung cancer cell line H226B, while one tobacco tar-resistant S. warneri (Sw-TA75) did not induce it significantly. Moreover, Sa-TA10 induced formation of transformed foci and soft-agar colony in Bhas 42 cells in cooperation with the v-Ha-ras gene. The results suggested that Sa-TA10 has carcinogenic potential, whereas Sw-TA75 does not. CONCLUSION: These data suggest that tobacco tar-resistant S. aureus, with carcinogenic potential, is present in the buccal cavity of some individuals, and that cigarette smoking simultaneously inhibits growth of most of the bacteria and selects carcinogenic bacteria.


Subject(s)
Carcinogens/adverse effects , Drug Resistance, Bacterial , Mouth Mucosa/microbiology , Staphylococcus aureus/growth & development , Tobacco Smoke Pollution/adverse effects , 3T3 Cells/pathology , Adult , Animals , Cell Transformation, Neoplastic/drug effects , Cells, Cultured , Gene Expression , Humans , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , RNA, Messenger/metabolism , Staphylococcus aureus/drug effects , Staphylococcus aureus/isolation & purification , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
11.
Oncogene ; 22(38): 5946-57, 2003 Sep 04.
Article in English | MEDLINE | ID: mdl-12955073

ABSTRACT

CrkII belongs to the adaptor protein family that plays a crucial role in signal transduction. In order to better understand the biological functions of CrkII, we focused on the regulation of gene expression by CrkII. Various transcriptional control elements were examined for their activation by CrkII-expression, and we found that CrkII selectively activates the serum response element (SRE), a transcriptional control element of immediate-early genes. This SRE activation induced by CrkII-overexpression was mediated by the serum response factor (SRF) via Rho. Indeed, we confirmed that the amount of activated Rho was increased in the CrkII-expressing cells. Moreover, we showed that when overexpressed, CrkII induces the cellular transformation of NIH 3T3 cells and that a dominant negative mutant of Rho suppresses this transformation, strongly suggesting that activation of Rho is essential for the transforming activity by CrkII. Furthermore, we also found that CrkII and Galpha12, a member of the heterotrimeric G proteins, synergistically activates Rho as well as the SRF, and that an SH3 mutant of CrkII can inhibit the Galpha12-induced activation of SRF. These results strongly suggest that CrkII is involved in the activation of Rho and SRF by Galpha12. Our study provides strong evidence that Rho activation plays a crucial role in CrkII-mediated signals to induce gene expression and cellular transformation.


Subject(s)
Cell Transformation, Neoplastic , Proto-Oncogene Proteins/metabolism , Serum Response Element/physiology , Serum Response Factor/metabolism , rho GTP-Binding Proteins/metabolism , 3T3 Cells/metabolism , 3T3 Cells/pathology , Animals , GTP-Binding Protein alpha Subunits, G12-G13 , Gene Expression Regulation , Genes, Reporter , Heterotrimeric GTP-Binding Proteins/genetics , Heterotrimeric GTP-Binding Proteins/metabolism , Mice , Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-crk , Serum Response Factor/genetics , Signal Transduction , Transcriptional Activation , rho GTP-Binding Proteins/genetics
12.
Oncogene ; 22(38): 6081-9, 2003 Sep 04.
Article in English | MEDLINE | ID: mdl-12955087

ABSTRACT

The cholecystokinin 2 receptor (CCK2R) increases proliferation of normal and neoplastic gastrointestinal cells and activates various mitogenic signaling pathways when stimulated by gastrin. To study the incidence of permanent activation of this receptor in tumorigenicity, a constitutively active mutant was generated by replacing residue Glu151 in the conserved E/DRY motif by Ala. Expression of the E151A-CCK2R mutant in NIH-3T3 cells causes ligand-independent activation of phospholipase C and ornithine decarboxylase, two enzymes critical for mitogenesis. Strikingly, the constitutive activity of this mutant was associated with dramatic alteration of NIH-3T3 cell morphology, enhanced cell proliferation and invasion. Moreover, injection of cells expressing E151A-CCK2R in nude mice resulted in the development of large and rapidly growing tumors. By contrast, none of these effects was observed with cells expressing the wild-type CCK2R, indicating that the tumorigenic properties of the E151A-CCK2R mutant is the result of its constitutive activation. To date, this is the first report that provides evidence for the high tumorigenic effect of a constitutively active CCK2R mutant, thus raising a potential role of the CCK2R in human cancer.


Subject(s)
Cell Transformation, Neoplastic/genetics , Point Mutation , Receptors, Cholecystokinin/genetics , Receptors, Cholecystokinin/metabolism , 3T3 Cells/pathology , Alanine/genetics , Amino Acid Motifs , Animals , Carcinogenicity Tests , Cell Division/genetics , Enzyme Activation , Female , Glutamic Acid/genetics , Humans , Mice , Mice, Nude , Ornithine Decarboxylase/metabolism , Receptor, Cholecystokinin B , Type C Phospholipases/metabolism
13.
Blood ; 102(13): 4535-40, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-12920032

ABSTRACT

The ability of viral or mutated cellular oncogenes to initiate neoplastic events and their poor immunogenicity have considerably undermined their potential use as immunotherapeutic tools for the treatment of human cancers. Using an Epstein-Barr virus-encoded oncogene, latent membrane protein 1 (LMP1), as a model, we report a novel strategy that both deactivates cellular signaling pathways associated with the oncogenic phenotype and reverses poor immunogenicity. We show that cotranslational ubiquitination combined with N-end rule targeting of LMP1 enhanced the intracellular degradation of LMP1 and total blockade of LMP1-mediated nuclear factor-kappaB (NF-kappaB) and signal transducer and activator of transcription (STAT) activation in human cells. In addition, although murine cells expressing LMP1 were uniformly tumorigenic, this oncogenicity was completely abrogated by covalent linkage of LMP1 with ubiquitin, while an enhanced CD8+ T cell response to a model epitope fused to the C-terminus of LMP1 was observed following immunization with ubiquitinated LMP1. These observations suggest that proteasomal targeting of tumor-associated oncogenes could be exploited therapeutically by either gene therapy or vaccination.


Subject(s)
Cysteine Endopeptidases/metabolism , Genes, Viral , Herpesvirus 4, Human/immunology , Multienzyme Complexes/metabolism , Oncogenes , Protein Processing, Post-Translational , Viral Matrix Proteins/metabolism , 3T3 Cells/pathology , 3T3 Cells/transplantation , 3T3 Cells/virology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Transformation, Viral , Epitopes/immunology , H-2 Antigens/immunology , Humans , Male , Mice , Mice, Nude , NF-kappa B/metabolism , Neoplasms, Experimental/etiology , Phenotype , Proteasome Endopeptidase Complex , Recombinant Fusion Proteins/immunology , Transcription, Genetic , Transfection , Ubiquitin/metabolism , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/immunology
14.
Mol Cell Biol ; 23(9): 3043-51, 2003 May.
Article in English | MEDLINE | ID: mdl-12697807

ABSTRACT

Despite the central role of TATA-binding protein (TBP) in transcription, changes in cellular TBP concentration produce selective effects on gene expression. Moreover, TBP is up-regulated by oncogenic signaling pathways. These findings suggest that TBP could be a nexus in pathways that regulate cell proliferation and that genetic lesions that result in cellular transformation may produce their effects at least in part through TBP. We provide evidence consistent with this hypothesis, demonstrating that increases in TBP expression contribute to cellular transformation. A Ras-mediated increase in TBP expression is required for full Ras transforming activity. TBP overexpression induces cells to grow in an anchorage-independent manner and to form tumors in athymic mice. These effects on cellular transformation require changes in RNA polymerase II-dependent transcription and on the selective recruitment of TBP to promoters via its DNA binding activity. TBP expression is elevated in human colon carcinomas relative to normal colon epithelium. Both Ras-dependent and Ras-independent mechanisms mediate increases in TBP expression in colon carcinoma cell lines. We conclude that TBP may be a critical component in dysregulated signaling that occurs downstream of genetic lesions that cause tumors.


Subject(s)
Carcinoma/genetics , Carcinoma/metabolism , Cell Transformation, Neoplastic , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , TATA-Box Binding Protein/metabolism , 3T3 Cells/metabolism , 3T3 Cells/pathology , Animals , Carcinogenicity Tests , Cells, Cultured , Epithelium/metabolism , Humans , Mice , Mice, Nude , RNA Polymerase II/metabolism , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , TATA-Box Binding Protein/genetics , Up-Regulation , ras Proteins/genetics , ras Proteins/metabolism
15.
Oncogene ; 22(10): 1501-10, 2003 Mar 13.
Article in English | MEDLINE | ID: mdl-12629513

ABSTRACT

Platelet-derived growth factors (PDGFs) are important for normal tissue growth and maintenance. Overexpression of the classical PDGFs, PDGF-A and PDGF-B, has been linked to several diseases, including cancer, fibrotic disease and atherosclerosis. Recently, two novel PDGFs, PDGF-C and PDGF-D, were discovered. It has not yet been established whether PDGF-C and PDGF-D are linked to disease phenotypes like the classical PDGFs. PDGF-B, the cellular homologue of the viral simian sarcoma oncogene v-sis, is known to potently induce cellular transformation through activation of PDGF receptor (PDGFR)-beta. In this work, we have determined the transformation efficacy of PDGF-D in comparison with that of PDGF-C and PDGF-B. PDGF-D is a potent transforming growth factor for NIH/3T3 cells, and the transformed cells displayed stress fibre reorganization, increased proliferation rate, anchorage-independent growth in soft agar, ability to induce tumours in nude mice, and upregulation of vascular endothelial growth factor. Morphological analyses of the vasculatures from the PDGF-isoform-expressing tumours revealed marked differences suggesting differential signalling through the two PDGF receptors in tumour vessel development and remodelling. In summary, these results suggest that PDGF-D induce cellular transformation and promote tumour growth by accelerating the proliferation rate of the tumour cells, and by stimulation of tumour neovascularization.


Subject(s)
Angiogenesis Inducing Agents/physiology , Cell Transformation, Neoplastic , Lymphokines , Platelet-Derived Growth Factor/physiology , 3T3 Cells/pathology , Actins/metabolism , Actins/ultrastructure , Animals , Carcinogenicity Tests , Cell Division/genetics , Down-Regulation , Female , Gene Expression Regulation , Humans , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic , Proto-Oncogene Proteins c-sis/physiology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Tumor Stem Cell Assay
16.
Cancer Gene Ther ; 10(2): 152-60, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12536204

ABSTRACT

Cancer gene therapy based on the use of suicide genes, such as the thymidine kinase gene, is not producing satisfactory results. Several approaches have been delineated to enhance the therapeutic responses, including augmentation of the bystander effect, the combination of the herpes simplex virus thymidine kinase-ganciclovir (HSVTK-GCV) system into replication competent adenoviruses and others. Moreover, because usually less than 20% of human malignant cells are in S-phase, the HSVTK-GCV system is not as efficient as expected. To increase the cytotoxic effects of the HSVTK-GCV system, we hypothesized that concomitant expression of E1a protein, which drives cells to proliferation and S-phase, could increase the effects of the HSVTK-GCV system. Several retroviruses were constructed carrying bicistronic sequences of TK and E1a 12S genes under the control of the CMV promoter. The constructions were tested in murine (NIH-3T3, MSC11A5) and human cells (IMR90, HeLa, MDA-MB435). A clear increase of the HSVTK-GCV system killing effect in nonconfluent cells was observed in the cells studied, especially in NIH-3T3, MSC11A5, IMR90, and MDA-MB435 expressing cells. In confluence, the NIH3T3 and IMR90 E1a-TK-expressing cells were also very sensitive and most malignant E1a-TK-expressing cells showed an irreversible G2-M cell cycle arrest. Moreover, the concomitant expression of adenovirus E1a and the HSVTK-GCV system increased the sensitivity to anticancer agents such as cisplatin. These results show that adenovirus E1a protein expression clearly enhances the cytotoxic effects of the HSVTK-GCV system and the response to treatment with cisplatin.


Subject(s)
Adenovirus E1A Proteins/pharmacology , Antibiotics, Antineoplastic/pharmacology , Ganciclovir/pharmacology , Genetic Therapy/methods , Simplexvirus/enzymology , Thymidine Kinase/pharmacology , 3T3 Cells/drug effects , 3T3 Cells/pathology , Adenovirus E1A Proteins/genetics , Animals , Cell Cycle/genetics , Cisplatin/pharmacology , Cyclin B/metabolism , Cyclin B1 , Drug Synergism , Genetic Engineering/methods , HeLa Cells/drug effects , HeLa Cells/pathology , Humans , Mice , Retroviridae/genetics , Simplexvirus/genetics , Thymidine Kinase/biosynthesis , Thymidine Kinase/genetics , Tumor Cells, Cultured
17.
Oncogene ; 22(2): 186-97, 2003 Jan 16.
Article in English | MEDLINE | ID: mdl-12527888

ABSTRACT

The RON receptor tyrosine kinase is a member of the MET proto-oncogene family that has been implicated in regulating motile-invasive phenotypes in certain types of epithelial cancers. The purpose of this study was to determine if RON expression is altered in primary human colorectal adenocarcinomas. Results from immunohistochemical staining showed that RON is highly expressed in the majority of colorectal adenocarcinomas (29/49 cases). Accumulated RON is also constitutively active with autophosphorylation in tyrosine residues. Moreover, three splicing variants of RON, namely RONdelta165, RONdelta160, and RONdelta155 were detected and cloned from two primary colon cancer samples. These RON variants were generated by deletions in different regions in extracellular domains of the RON beta chain. Functional studies showed that expression of RONdelta160 or RONdelta155 in Martin-Darby canine kidney cells resulted in increased cell dissociation (scatter-like activity). RON variants, RONdelta160 and RONdelta155, also exerted the ability to induce multiple focus formation and sustain anchorage-independent growth of transfected NIH3T3 cells. Moreover, NIH3T3 cells expressing RONdelta160 or RONdelta155 formed tumors in athymic nude mice and colonized in the lungs. These data suggest that RON expression is altered in certain primary colon cancers. Abnormal accumulation of RON variants may play a role in the progression of certain colorectal cancers in vivo.


Subject(s)
Adenocarcinoma/metabolism , Colorectal Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism , 3T3 Cells/pathology , Adenocarcinoma/genetics , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , Base Sequence , Carcinogenicity Tests , Cell Transformation, Neoplastic , Cells, Cultured , Cloning, Molecular , Colorectal Neoplasms/genetics , Female , Genetic Variation , Humans , Intestinal Mucosa/metabolism , Male , Mice , Mice, Nude , Middle Aged , Molecular Sequence Data , Proto-Oncogene Mas , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Cell Surface/genetics , Reference Values
18.
Nat Biotechnol ; 21(1): 41-6, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12459735

ABSTRACT

Semiconductor quantum dots (QDs) are among the most promising emerging fluorescent labels for cellular imaging. However, it is unclear whether QDs, which are nanoparticles rather than small molecules, can specifically and effectively label molecular targets at a subcellular level. Here we have used QDs linked to immunoglobulin G (IgG) and streptavidin to label the breast cancer marker Her2 on the surface of fixed and live cancer cells, to stain actin and microtubule fibers in the cytoplasm, and to detect nuclear antigens inside the nucleus. All labeling signals are specific for the intended targets and are brighter and considerably more photostable than comparable organic dyes. Using QDs with different emission spectra conjugated to IgG and streptavidin, we simultaneously detected two cellular targets with one excitation wavelength. The results indicate that QD-based probes can be very effective in cellular imaging and offer substantial advantages over organic dyes in multiplex target detection.


Subject(s)
Biomarkers, Tumor/metabolism , Nanotechnology/methods , Receptor, ErbB-2/metabolism , Spectrometry, Fluorescence/methods , Staining and Labeling/methods , 3T3 Cells/metabolism , 3T3 Cells/pathology , Animals , Antigens, Neoplasm/analysis , Biomarkers, Tumor/analysis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Crystallization/methods , Diagnostic Imaging/instrumentation , Diagnostic Imaging/methods , Electrochemistry , Fibroblasts/metabolism , Fibroblasts/pathology , Fluorescent Antibody Technique/methods , Fluorescent Dyes , Humans , Mice , Microchemistry/methods , Microspheres , Receptor, ErbB-2/analysis , Semiconductors , Spectrometry, Fluorescence/instrumentation , Staining and Labeling/instrumentation , Tumor Cells, Cultured
19.
Biochemistry ; 41(41): 12350-8, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12369824

ABSTRACT

Cdc42 is a small GTP-binding protein which has been implicated in a number of cellular activities, including cell morphology, motility, cell-cycle progression, and malignant transformation. While GTPase-defective forms of Cdc42 inhibit cell growth, a mutation [Cdc42(F28L)] that allows the constitutive exchange of GDP for GTP and is GTPase-competent induces cellular transformation. These results suggest that Cdc42 must cycle between its GTP- and GDP-bound states to stimulate cell growth. In attempting to design Cdc42 molecules with more potent transforming activity, we set out to generate other types of Cdc42 mutants capable of constitutive GDP-GTP exchange. Here, we describe one such mutant, generated by changing a conserved aspartic acid residue at position 118 to an asparagine. The Cdc42(D118N) protein exchanges GDP for GTP more rapidly than wild-type Cdc42, but significantly more slowly than the Cdc42(F28L) mutant. Despite its slower rate of activation, the Cdc42(D118N) mutant is more potent at inducing cellular transformation than the Cdc42(F28L) protein, and causes a significant loss in actin stress fibers, reminiscent of what is observed with fibroblasts transformed by oncogenic Ras mutants. Effector-loop mutations made within the D118N background inhibit Cdc42-induced transformation and Cdc42-mediated antiapoptotic (survival) activity to similar extents. In addition, mutating aspartic acid 121 (to asparagine), which forms part of a caspase cleavage site (DLRD, residues 118-121 of Cdc42), in combination with the F28L mutation generates a Cdc42 molecule [Cdc42(F28L/D121N)] with transforming activity significantly stronger than that of Cdc42(F28L). Thus, mutations that combine some capacity for cycling between the GTP- and GDP-bound states with increased survival against apoptotic signals yield Cdc42 molecules with the maximum capability for inducing cellular transformation.


Subject(s)
Amino Acid Substitution/genetics , Apoptosis/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Trans-Activators/genetics , cdc42 GTP-Binding Protein/genetics , 3T3 Cells/cytology , 3T3 Cells/metabolism , 3T3 Cells/pathology , Animals , Asparagine/genetics , Aspartic Acid/genetics , COS Cells , Cell Division/genetics , Cell Transformation, Neoplastic/metabolism , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , Humans , Mice , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Protein Binding/genetics , Trans-Activators/biosynthesis , Trans-Activators/metabolism , Trans-Activators/physiology , Transfection , cdc42 GTP-Binding Protein/biosynthesis , cdc42 GTP-Binding Protein/metabolism , cdc42 GTP-Binding Protein/physiology
20.
J Toxicol Sci ; 27(3): 229-37, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12238146

ABSTRACT

We investigated and compared the cytotoxicity of 16 reference compounds in four in vitro systems: primary cultured rat hepatocytes, hepatoma HepG2 cell line, non-hepatic HeLa and Balb/c 3T3 cell lines. After 24 hr of exposure to the test compounds, the water-soluble tetrazolium salts WST-1 assay was used as an endpoint to evaluate cytotoxicity. Acetaminophen, diclofenac sodium cyclophosphamide and disulfiram displayed from 2 to more than 10 times higher IC50 values in three cell lines than in rat primary cultured hepatocytes. The cytotoxic effects of aspirin, amiodarone, clorfibiric acid, chlorpromazine, erythomycin, lithocholic acid, cisplatin and quinidine in rat hepatocytes were similar or 2 times stronger than those observed in cell lines. Ketoconazole resulted in the lowest IC50 value in the HeLa cell line. The data suggested that the compounds which are known to be metabolism-mediated liver toxicants have a differential hepatotoxicity in vitro and that primary cultured rat hepatocytes could represent a valuable tool for both screening and study of the effects of bio-transformation on the cytotoxicity of new chemical entities and xenobiotics in vitro.


Subject(s)
3T3 Cells/drug effects , HeLa Cells/drug effects , Hepatocytes/drug effects , Toxicity Tests/methods , Xenobiotics/toxicity , 3T3 Cells/pathology , Animals , Dose-Response Relationship, Drug , HeLa Cells/pathology , Hepatocytes/pathology , Humans , In Vitro Techniques , Male , Mice , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...