Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Sci Rep ; 12(1): 10850, 2022 06 27.
Article in English | MEDLINE | ID: mdl-35760982

ABSTRACT

Heparan sulfates have long been known to intracellularly accumulate in Alzheimer's disease neurons, where they colocalize with neurofibrillary tangles made of abnormally phosphorylated and aggregated tau protein. However, the reasons and consequences of the heparan sulfates accumulation in the Alzheimer's cells are not yet well understood. Previously, we showed that the neural heparan sulfate 3-O-sulfotransferase HS3ST2 is critical for the abnormal phosphorylation of tau in Alzheimer's disease-related tauopathy. Using cell models of tauopathy we showed that intracellular 3-O-sulfatated heparan sulfates interact with tau inducing its abnormal phosphorylation. However, it is unknown whether HS3ST2 expression induces the intracellular aggregation of tau in cells. Here, by using replicative pEBV plasmids, we engineered HEK293 cells to stably express HS3ST2 together with human tau carrying or not the P301S mutation. We show that HS3ST2 gain of function induces the cell autonomous aggregation of tau not only in cells expressing tauP301S, but also in cells expressing the wild type tau. Our engineered cells mimicked both the HS intracellular accumulation observed in neurons of Alzheimer's disease and the tau aggregation characteristic of tauopathy development and evolution. These results give evidence that the neural HS3ST2 plays a critical role in the cell autonomous self-aggregation of tau.


Subject(s)
Alzheimer Disease , Sulfotransferases/metabolism , Tauopathies , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , HEK293 Cells , Heparitin Sulfate/metabolism , Humans , Neurofibrillary Tangles/metabolism , Phosphorylation , Sulfates/metabolism , Tauopathies/metabolism , tau Proteins/genetics , tau Proteins/metabolism
2.
Oncogene ; 36(16): 2328-2336, 2017 04 20.
Article in English | MEDLINE | ID: mdl-27819670

ABSTRACT

Heat shock protein 110 (HSP110) is induced by different stresses and, through its anti-apoptotic and chaperoning properties, helps cells survive these adverse situations. In colon cancers, HSP110 is abnormally abundant. We have recently shown that colorectal cancer patients with microsatellite instability (MSI) had an improved response to chemotherapy because they harbor an HSP110-inactivating mutation (HSP110DE9). In this work, we used patient biopsies, human colorectal cancer cells grown in vitro and in vivo (xenografts), and intestinal crypts to demonstrate that HSP110 is also involved in colon cancer growth. We showed that HSP110 induces colon cancer cell proliferation and that this effect is associated with STAT3 activation, specifically an increase in STAT3 phosphorylation, nuclear translocation and transcription factor activity. STAT3 inhibition blocks the proliferative effect of HSP110. From a molecular standpoint, we demonstrated that HSP110 directly binds to STAT3, thereby facilitating its phosphorylation by JAK2. Finally, we showed a correlation between HSP110 expression and STAT3 phosphorylation in colon cancer patient samples. Thus, the expression of HSP110 in colon cancer contributes to STAT3-dependent tumor growth and the frequent inactivating mutation of this chaperone is probably an important event underlying the improved prognosis in colon cancer displaying MSI.


Subject(s)
Colorectal Neoplasms/pathology , HSP110 Heat-Shock Proteins/metabolism , STAT3 Transcription Factor/metabolism , Animals , Biopsy , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Humans , Intestinal Mucosa/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Phosphorylation , Protein Binding
3.
Nucleic Acids Res ; 35(11): 3535-50, 2007.
Article in English | MEDLINE | ID: mdl-17483520

ABSTRACT

Much effort has long been devoted to unraveling the coordinated cellular response to genotoxic insults. In view of the difficulty of obtaining human biological samples of homogeneous origin, I have established a set of stable human clones where one DNA repair gene has been stably silenced by means of RNA interference. I used pEBVsiRNA plasmids that greatly enhance long-term gene silencing in human cells. My older clones reached >500 days in culture. Knock-down HeLa clones maintained a gene silencing phenotype for an extended period in culture, demonstrating that I was able to mimic cells from cancer-prone syndromes. I have silenced >20 genes acting as sensors/transducers (ATM, ATR, Rad50, NBS1, MRE11, PARG and KIN17), or of different DNA repair pathways. In HeLa cells, I have switched off the expression of genes involved in nucleotide excision repair (XPA, XPC, hHR23A, hHR23B, CSA and CSB), nonhomologous end-joining (DNA-PKcs, XRCC4 and Ligase IV), homologous recombination repair (Rad51 and Rad54), or base excision repair (Ogg1 and Ligase III). These cells displayed the expected DNA repair phenotype. We could envisage untangling the complex network between the different DNA repair pathways. In this study, no viral vehicles, with their attendant ethical and safety concerns, were used.


Subject(s)
DNA Repair , RNA Interference , Acid Anhydride Hydrolases , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Clone Cells , DNA Damage , DNA Repair Enzymes/antagonists & inhibitors , DNA Repair Enzymes/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Etoposide/toxicity , Gamma Rays , Genetic Vectors , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , MRE11 Homologue Protein , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Plasmids/genetics , Ultraviolet Rays
4.
Radiat Res ; 159(6): 748-58, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12751957

ABSTRACT

The human KIN17 protein is a chromatin-associated protein involved in DNA replication. Certain tumor cell lines overproduce KIN17 protein. Among 16 cell lines, the highest KIN17 protein level was observed in H1299 non-small cell lung cancer cells, whereas the lowest was detected in MeWo melanoma cells. Cells displaying higher KIN17 protein levels exhibited elevated RPA70 protein contents. High KIN17 protein levels may be a consequence of the tumorigenic phenotype or a prerequisite for tumor progression. Twenty-four hours after exposure to ionizing radiation, after the completion of DNA repair, a co-induction of chromatin-bound KIN17 and RPA70 proteins was detected. Etoposide, an inhibitor of topoisomerase II generating double-strand breaks, triggered the concentration of KIN17 into punctuate intranuclear foci. KIN17 may be associated with unrepaired DNA sites. Flow cytometry analysis revealed that 48 h after transfection the uppermost KIN17-positive RKO cells shifted in the cell cycle toward higher DNA content, suggesting that KIN17 protein induced defects in chromatin conformation. Cells displaying reduced levels of KIN17 transcript exhibited a sixfold increased radiosensitivity at 2 Gy. The KIN17 protein may be a component of the DNA replication machinery that participates in the cellular response to unrepaired DSBs, and an impaired KIN17 pathway leads to an increased sensitivity to ionizing radiation.


Subject(s)
DNA-Binding Proteins/physiology , Nuclear Proteins , Radiation Tolerance , Animals , Cell Nucleus/metabolism , Chromatin/chemistry , DNA Damage , DNA Replication , DNA-Binding Proteins/analysis , Enzyme-Linked Immunosorbent Assay , Etoposide/pharmacology , Gamma Rays , Humans , Mice , RNA-Binding Proteins , Tumor Cells, Cultured
5.
Exp Cell Res ; 250(2): 499-509, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10413603

ABSTRACT

To characterize the biological role of Kin17 protein, a mammalian nuclear protein which participates in the response to UV and ionizing radiation and binds to curved DNA, EBV-derived vectors carrying (Mm)Kin17 cDNA were constructed and transfected in tumorigenic cells harboring different p53 profiles (HeLa, H1299, and HCT116) and in immortalized HEK 293 cells. (Mm)Kin17 protein expression induced a tremendous decrease in cell proliferation of the three tumorigenic cell lines 2 weeks after transfection. Transfection of HEK 293 cells with an pEBVCMV(Mm)Kin17 plasmid gave rise to numerous (Mm)Kin17-expressing cells which constantly disappeared with time, preventing the establishment of (Mm)Kin17-expressing cells. Several independent clones were isolated from HEK 293 cells carrying a pEBVMT(Mm)Kin17 vector. The two clones described here (B223.1 and B223.2) exhibited different (Mm)Kin17 protein levels and displayed a gradual decrease in their proliferative capacities. In B223.1 cells, the basal expression of (Mm)Kin17 greatly reduced plating efficiency and cell growth. B223.1 cell morphology was altered, with numerous round-shaped cells whose spreading on the culture support was hampered. We observed giant multinucleated cells or cells containing micronuclei-like structures and/or multilobed nuclei. To conclude, (Mm)Kin17 overexpression reduced the proliferation of tumorigenic cells independently of their p53 status and modified cell growth and cell morphology of established HEK 293 cells producing (Mm)Kin17 protein. It is likely that (Mm)Kin17 may interfere with DNA replication.


Subject(s)
Cell Division , DNA-Binding Proteins/physiology , Nuclear Proteins , Animals , Blotting, Western , Cadmium/pharmacology , Cell Count , Cell Line, Transformed , Cell Size , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/metabolism , Cyclin B/metabolism , DNA-Binding Proteins/genetics , Fluorescent Antibody Technique, Indirect , Gene Expression/drug effects , Giant Cells/metabolism , Humans , In Situ Hybridization, Fluorescence , Mice , Proliferating Cell Nuclear Antigen/metabolism , RNA-Binding Proteins , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Zinc/pharmacology
6.
Cytotechnology ; 26(1): 29-38, 1998 Jan.
Article in English | MEDLINE | ID: mdl-22359004

ABSTRACT

To characterize the differences between fibrotic myofibroblasts and normal fibroblasts, we studied two differentiation markers: α-smooth muscle (SM) actin, a specific marker of myofibroblast differentiation, and ß-actin, which is overexpressed in the fibrotic tissue. Experiments were performed on fibroblasts isolated from normal pig skin and on subcutaneous myofibroblasts isolated from pig radiation-induced fibrosis. Three culture models were used: cells in monolayers, equivalent dermis, consisting of fibroblasts embedded into a matrix composed of type I collagen, and in vitro reconstituted skin, in which the matrix and containing life fibroblasts were overlaid with keratinocytes. Samples were studied using immunofluorescence and western-blotting. In monolayers cultures, both fibrosis and normal cells expressed α-SM actin. Furthermore, similar amounts of ß-actin protein were found. In these conditions, the resulting alterations in the phenotypes of cells made comparison of cultured fibrotic and normal cells irrelevant. Under the two 3-D culture models, normal fibroblasts no longer expressed α-SM actin. They expressed ß-actin at the basal level. Moreover, the fibrotic myofibroblasts in both 3-D models retained their differentiation features, expressing α-SM actin and overexpressing ß-actin. We found that this normalization was mainly related to the genomic programmation acquired by the cells in the tissue. Cellular motility and microenvironment were also involved, whereas cellular proliferation was not a major factor. Consequently, both three-dimensional models allowed the study of radiation-induced fibrosis in vitro, provided good extrapolations to in vivo conditions and avoided certain of culture artefacts.

7.
Arch Dermatol Res ; 289(8): 448-56, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9266022

ABSTRACT

In eukaryotic cells, various proteins homologous to the E. coli RecA protein are involved in the elimination of DNA damage. These proteins contribute to the repair of double-strand breaks and to genetic recombination. The mouse Kin17 protein is recognised by antibodies directed against the RecA protein. Kin17 has a zinc-finger domain allowing binding to curved DNA stretching over illegitimate recombination junctions. In the present study, we identified the human counterpart of the mouse Kin17 protein (named HsKin17) in skin cells. We employed an in vitro reconstructed skin model composed of an epidermal sheath lying on a dermal matrix with human fibroblasts embedded in rat collagen type I. The maturation programme (proliferation versus differentiation) of keratinocytes was highly dependent on stromal cells. Immunohistochemical staining of frozen sections obtained from skin specimens was monitored by an interactive laser cytometer. In this way we analysed protein levels in both dermal and epidermal compartments. After having characterised the epithelium, we focused our attention on HsKin17 expression. We detected HsKin17 in human keratinocytes. HsKin17 protein levels increased in proliferating epithelial keratinocytes after 7 days of culture. After 2 weeks of culture, epidermal sheaths acquired most of the differentiated features of mature epithelium. At this time, HsKin17 protein dropped below measurable levels in the stratum corneum, and diminished in nucleated cells. This study showed that HsKin17 is expressed in human reconstructed epithelium under conditions of hyperproliferation.


Subject(s)
DNA-Binding Proteins/analysis , Nuclear Proteins , Skin/chemistry , Adult , Animals , Basement Membrane/cytology , Cell Differentiation , Cells, Cultured , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Female , Humans , Keratinocytes/chemistry , Mice , RNA-Binding Proteins , Rats , Skin/cytology
8.
Radiat Res ; 147(4): 442-50, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9092924

ABSTRACT

Kin17 is a mammalian nuclear protein sharing a slight sequence homology with the bacterial RecA protein. Kin17 has a zinc-finger motif and binds efficiently to curved DNA, a genomic topology associated with illegitimate recombination junctions. We investigated the relationship between the level of Kin17 protein and genomic alteration due to either impaired wild-type p53 functions or exposure to gamma rays. We used BP cells, a rodent epithelial cell system. The cell lines used were syngeneic and harbored wild-type or mutant p53 alleles and exhibited different sensitivities to gamma irradiation. In radioresistant cells (wild-type p53 genotype), the level of Kin17 protein peaked 30 min after a low dose of radiation (2 Gy), whereas maximum accumulation of p53 protein was observed 3 h postirradiation. Radiosensitive cells carrying the same mutation in both alleles of the p53 gene showed elevated basal levels of both Kin17 and p53 proteins and failed to accumulate Kin17 and p53 proteins after exposure to ionizing radiation. These cells exhibited enhanced cell death by apoptosis after gamma irradiation. Our results indicate that Kin17 protein accumulated immediately after DNA damage in cells carrying a wild-type p53 genotype, and that levels of constitutive Kin17 protein increased in highly proliferating tumorigenic cells when wild-type p53 functions were abrogated.


Subject(s)
Apoptosis/radiation effects , Cell Survival/radiation effects , DNA-Binding Proteins/biosynthesis , Gene Expression/radiation effects , Nuclear Proteins , Tumor Suppressor Protein p53/biosynthesis , Animals , Benzo(a)pyrene/toxicity , Cell Cycle/radiation effects , Cell Line , Cobalt Radioisotopes , Dose-Response Relationship, Radiation , Epithelium , Gamma Rays , Genes, p53/radiation effects , Kinetics , Lung , Point Mutation , Rats , Zinc Fingers
9.
Biochimie ; 79(9-10): 599-606, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9466698

ABSTRACT

We used antibodies against E coli RecA protein to identify in mouse cells a 45-kDa DNA-binding protein called kin17, which has an active zinc finger and a nuclear localisation signal. Kin17 protein produced in E coli binds preferentially to the curved DNA of a bacterial promoter in vivo and in vitro, suggesting a transcriptional regulation activity. The fact that in rodent cells kin17 protein levels increase after gamma-irradiation suggests its participation in a cellular response to ionising radiation. We raised polyclonal antibodies against the whole kin17 protein and against its derived synthetic peptides. We report the detection of kin17 protein and of truncated forms of the protein by Western blot or by immunocytochemistry after transient overexpression in cultured human cells. Our results indicate that the cross-reactivity with the anti-RecA antibodies is due to an antigenic determinant located in the core of kin17 protein, between residues 129 and 228. The kin17 protein is located in the nucleus and is concentrated in small nuclear dot-like structures throughout the nucleoplasm. The RecA homologous region seems to play an essential role in the localisation of kin17 protein since the deletion of this particular region dramatically changes the form and the distribution of the intranuclear foci. We hypothesise that these dot-like structures reflect nuclear metabolism compartmentalization.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/biosynthesis , Nuclear Proteins , Animals , Blotting, Western , Cell Nucleus/genetics , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Mice , Mutagenesis, Insertional , Mutagenesis, Site-Directed , Nuclear Localization Signals/genetics , Protein Structure, Tertiary , RNA-Binding Proteins , Rec A Recombinases/genetics , Sequence Deletion , Subcellular Fractions/metabolism , Transfection , Zinc Fingers/genetics
10.
Biochimie ; 77(11): 854-60, 1995.
Article in English | MEDLINE | ID: mdl-8824764

ABSTRACT

We have sought to characterize the molecular basis of the sensitivity to ionising radiation and to identify the genes involved in the cellular response of mammalian cells to such radiation. Using the Escherichia coli model, we tested the hypothesis that functional domains of RecA protein are represented in proteins of mammalian cells. We review here the results obtained in the detection of nuclear proteins of mammalian cells that are recognized by anti-RecA antibodies. We have called them kin proteins. Kin proteins likely play a role in DNA metabolism. We summarize the cloning of the mouse Kin-17 cDNA and our work on the identification and preliminary characterisation of the biochemical properties of mouse kin17 protein, a new nuclear protein able to recognize bent DNA and suspected to be involved in illegitimate recombination. We briefly describe our latest experiments on the molecular characterisation of the mouse Kin-17 gene. Finally, we discuss the properties of kin17 protein and the possible participation of kin17 protein in DNA transactions like transcription or recombination.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Nuclear Proteins/metabolism , Rec A Recombinases/immunology , Animals , Antibodies/immunology , Chromosomes , Cloning, Molecular , Conserved Sequence , DNA, Complementary , DNA-Binding Proteins/genetics , Genes , Mammals , Mice , Nuclear Proteins/genetics , Plant Proteins/metabolism , Proteins/immunology
11.
Cancer Res ; 54(13): 3361-4, 1994 Jul 01.
Article in English | MEDLINE | ID: mdl-8012950

ABSTRACT

A rat lung cell population had been treated with benzo(a)pyrene, and a set of different epithelial cell lines was derived from it. These cell lines carried either a wild-type or mutant p53 gene and represented grading states of neoplastic development. We demonstrate here that the cells lacking both wild-type p53 alleles display a significant decrease in survival after gamma-irradiation with doses of 2 to 12 Gy, compared with their counterparts carrying wild-type p53 alleles. This is the first reported model in which cells bearing a mutation of the p53 gene display enhanced sensitivity to ionizing radiation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Genes, p53/genetics , Lung/pathology , Point Mutation , Animals , Benzo(a)pyrene , Cell Line , Cell Survival , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/pathology , Gene Deletion , Genes, p53/drug effects , Radiation Tolerance , Rats
12.
Cancer Res ; 52(19): 5213-8, 1992 Oct 01.
Article in English | MEDLINE | ID: mdl-1382840

ABSTRACT

We have developed a model system for assessing the demethylating potential of external agents. Disruption in the DNA methylation pattern was evaluated at the translational level of the Escherichia coli beta-galactosidase coding gene (lacZ). We have constructed a clonal cell line (A4/4 cells) derived from the adenovirus-transformed human embryonic kidney 293 strain. The A4/4 cells contain the E. coli lacZ gene under the control of the mouse metallothionein 1 promoter which is down-regulated by a natural DNA methylation pattern. Furthermore, the lacZ transcription is also regulated by the E. coli lac operator/repressor system and by mouse metallothionein 1 metal responsiveness offering a wide range in lacZ expression. In this system, the beta-galactosidase activity was only recovered in the presence of a demethylating agent such as 5-azacytidine. The demethylating potential of 5-azacytidine, 5-aza 2'-deoxycytidine and sodium butyrate was rapidly assessed by a flow cytometric method using fluorescein di-beta-D galactopyranoside as a fluorescent probe. A tremendous induction of lacZ expression was triggered by these drugs. Analysis of cell cycles showed little disruptions with 5-azacytidine and sodium butyrate, but an important blockage in the S-phase following 5-aza 2'-deoxycytidine treatment was observed. This approach allows a rapid identification and study of environmental demethylating agents.


Subject(s)
Azacitidine/analogs & derivatives , Azacitidine/analysis , Butyrates/analysis , DNA, Bacterial/drug effects , Azacitidine/pharmacology , Base Sequence , Butyrates/pharmacology , Butyric Acid , Cell Cycle/drug effects , Cell Transformation, Viral/physiology , Cells, Cultured , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Decitabine , Escherichia coli/enzymology , Escherichia coli/genetics , Flow Cytometry , Fluorometry , Gene Expression Regulation, Enzymologic/drug effects , Humans , Kidney/cytology , Lac Operon/drug effects , Lac Operon/genetics , Methylation , Molecular Sequence Data , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , beta-Galactosidase/drug effects , beta-Galactosidase/genetics
13.
Exp Cell Res ; 200(2): 263-71, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1572395

ABSTRACT

To detect xenobiotics affecting the DNA methylation pattern we have established a human clonal cell line (A4/4) derived from the 293 epithelial cells. This cell line is stable after more than 1 year in culture and we report here its characteristics. The A4/4 cells carry the Escherichia coli lacI gene and the symmetric lacO sequence upstream of the beta-galactosidase coding gene (lacZ). Both sequences have been independently integrated into the genomic DNA. The lacZ gene is under the control of the metal-inducible mouse metallothionein-I (mMT-I) promoter. Vector integration followed by cell ageing in culture gave rise to the methylation of the 5'CpG3' sites in the mMT-I promoter and the 5' part of the lacZ gene. The reactivation of the lacZ gene by 5-azacytidine (5-Aza-CR) treatment allowed a tremendous lacZ expression which is correlated with demethylation in the mMT-I promoter and its neighboring regions. This enhanced transcription is easily quantified by measuring the beta-galactosidase activity in cell extracts. 5-Aza-CR, the specific isopropyl beta-D-thiogalactoside inducer, and heavy metal ions added together trigger an increase of the beta-galactosidase activity up to 600-fold over the basal level. These cells can be used for a rapid assessment of the demethylating potential of environmental chemicals.


Subject(s)
Cell Line , DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Expression Regulation , Clone Cells , DNA Replication , DNA, Recombinant , Humans , In Vitro Techniques , Lac Operon , Metallothionein/genetics , Methylation , Plasmids , Promoter Regions, Genetic , Restriction Mapping
14.
Biochim Biophys Acta ; 1130(1): 68-74, 1992 Feb 28.
Article in English | MEDLINE | ID: mdl-1311956

ABSTRACT

We have investigated the use of various Epstein-Barr virus (EBV)-based vectors bearing the two components of the Escherichia coli lac operator-repressor (lacO, lacI) complex. Our aim was to develop a model system of gene expression by looking at the transcription of the bacterial beta-galactosidase coding gene (lacZ) in 293 human embryonic kidney cells. Several vectors have been built carrying different promoters upstream of the lacI and lacZ genes and in which natural or synthetic operator sequences were inserted in the 5' part of the lacZ gene. In transient expression assays we achieved efficient lacZ gene repression which could be released by the specific inducer isopropyl beta-D-thiogalactoside (IPTG). A stable transformed cell line carrying two EBV-derived plasmids with the building blocks of the lac operator/repressor system was established. This cell line allowed us to achieve a wide range of lacZ gene regulation. In this cell line IPTG alone could remove the repression to trigger a 5-fold increase of lacZ expression. Heavy metal ions, which induced the mouse metallothionein I promoter located upstream of the lacZ gene, added together with IPTG gave rise to a 40-fold induction of lacZ expression.


Subject(s)
Gene Expression Regulation/drug effects , Genetic Vectors/genetics , Isopropyl Thiogalactoside/pharmacology , Lac Operon/genetics , Base Sequence , Cadmium/pharmacology , Cell Line , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , Escherichia coli/genetics , Herpesvirus 4, Human/genetics , Humans , Molecular Sequence Data , Plasmids/genetics , Promoter Regions, Genetic/genetics , Repressor Proteins/genetics , Transcription, Genetic/genetics , Zinc/pharmacology , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL