Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cancer Res ; 77(9): 2279-2291, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28235765

ABSTRACT

IL6 produced by tumor cells promotes their survival, conferring a poor prognosis in patients with cancer. IL6 also contributes to immunosuppression of CD4+ T cell-mediated antitumor effects. In this study, we focused on the impact of IL6 trans-signaling mediated by soluble IL6 receptors (sIL6R) expressed in tumor-bearing hosts. Higher levels of sIL6R circulating in blood were observed in tumor-bearing mice, whereas the systemic increase of sIL6R was not prominent in tumor-bearing mice with myeloid cell-specific conditional deletion of IL6R even when tumor cells produced sIL6R. Abundant sIL6R was released by CD11b+ cells from tumor-bearing mice but not tumor-free mice. Notably, IL6-mediated defects in Th1 differentiation, T-cell helper activity for tumor-specific CD8+ T cells, and downstream antitumor effects were rescued by myeloid-specific deletion of sIL6R. Expression of the T-cell transcription factor c-Maf was upregulated in CD4+ T cells primed in tumor-bearing mice in an IL6-dependent manner. Investigations with c-Maf loss-of-function T cells revealed that c-Maf activity was responsible for IL6/sIL6R-induced Th1 suppression and defective T-cell-mediated antitumor responses. In patients with cancer, myeloid cell-derived sIL6R was also possibly associated with Th1 suppression and c-Maf expression. Our results argued that increased expression of sIL6R from myeloid cells and subsequent c-Maf induction were adverse events for counteracting tumor-specific Th1 generation. Overall, this work provides a mechanistic rationale for sIL6R targeting to improve the efficacy of T-cell-mediated cancer immunotherapy. Cancer Res; 77(9); 2279-91. ©2017 AACR.


Subject(s)
Carcinogenesis/genetics , Interleukin-6/genetics , Neoplasms/genetics , Proto-Oncogene Proteins c-maf/genetics , Animals , CD4-Positive T-Lymphocytes/pathology , Gene Expression Regulation, Neoplastic , Humans , Mice , Molecular Targeted Therapy , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplasms/pathology , Receptors, Interleukin-6/genetics , Th1 Cells
2.
J Hepatobiliary Pancreat Sci ; 24(2): 109-119, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28008721

ABSTRACT

BACKGROUND: iPS-ML are myeloid lineage cells with a proliferative capacity derived from induced pluripotent stem (iPS) cells. This study aimed to examine therapeutic effect of iPS-ML producing interferon-ß (iPS-ML/IFN-ß) towards primary and metastatic liver cancer and investigate the mechanism of that effect. METHODS: We established a xenograft model of liver metastasis by injecting the spleen of SCID mice with MKN-45 human gastric cancer cells and also a primary liver cancer model by injecting SK-HEP-1 human hepatocellular carcinoma cells into the liver. After cancer lesions were established, iPS-ML/IFN-ß was administered by intraperitoneal injection, and therapeutic effect was evaluated. RESULTS: The i.p. injection of iPS-ML/IFN-ß resulted in a significant retardation of cancer progression and prolonged mouse survival. The infiltration of i.p. administered iPS-ML into tumor lesions located below the liver capsule was observed, suggesting tumor-directed migration and penetration of the liver capsule by iPS-ML. The IFN-ß concentration in the liver was maintained at levels sufficient to exert an anti-cancer effect for at least 3 days post-injection, accounting for the potent therapeutic effect obtained by injection two to three times per week. CONCLUSIONS: This study demonstrates the therapeutic potential of the iPS-ML/IFN-ß in patients with liver cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/therapy , Induced Pluripotent Stem Cells/transplantation , Interferon-beta/therapeutic use , Liver Neoplasms/therapy , Myeloid Cells/transplantation , Stomach Neoplasms/therapy , Animals , Carcinoma, Hepatocellular/secondary , Cell- and Tissue-Based Therapy , Disease Models, Animal , Liver Neoplasms/secondary , Mice, SCID , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays
3.
PLoS One ; 11(4): e0152384, 2016.
Article in English | MEDLINE | ID: mdl-27050553

ABSTRACT

We previously reported a method to expand human monocytes through lentivirus-mediated introduction of cMYC and BMI1, and we named the monocyte-derived proliferating cells, CD14-ML. CD14-ML differentiated into functional DC (CD14-ML-DC) upon addition of IL-4, resulting in the generation of a large number of DC. One drawback of this method was the extensive donor-dependent variation in proliferation efficiency. In the current study, we found that introduction of BCL2 or LYL1 along with cMYC and BMI1 was beneficial. Using the improved method, we obtained CD14-ML from all samples, regardless of whether the donors were healthy individuals or cancer patients. In vitro stimulation of peripheral blood T cells with CD14-ML-DC that were loaded with cancer antigen-derived peptides led to the establishment of CD4+ and CD8+ T cell lines that recognized the peptides. Since CD14-ML was propagated for more than 1 month, we could readily conduct genetic modification experiments. To generate CD14-ML-DC that expressed antigenic proteins, we introduced lentiviral antigen-expression vectors and subjected the cells to 2 weeks of culture for drug-selection and expansion. The resulting antigen-expressing CD14-ML-DC successfully induced CD8+ T cell lines that were reactive to CMVpp65 or MART1/MelanA, suggesting an application in vaccination therapy. Thus, this improved method enables the generation of a sufficient number of DC for vaccination therapy from a small amount of peripheral blood from cancer patients. Information on T cell epitopes is not necessary in vaccination with cancer antigen-expressing CD14-ML-DC; therefore, all patients, irrespective of HLA type, will benefit from anti-cancer therapy based on this technology.


Subject(s)
Antigens/biosynthesis , Dendritic Cells/cytology , Lipopolysaccharide Receptors/immunology , Adult , Cell Differentiation , Cell Proliferation , Dendritic Cells/immunology , Female , Humans , Lymphocyte Activation , Male , Middle Aged , Young Adult
4.
Nat Commun ; 6: 6702, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25850032

ABSTRACT

Decline in immune function and inflammation concomitantly develop with ageing. Here we focus on the impact of this inflammatory environment on T cells, and demonstrate that in contrast to successful tumour elimination in young mice, replenishment of tumour-specific CD4(+) T cells fails to induce tumour regression in aged hosts. The impaired antitumour effect of CD4(+) T cells with their defective Th1 differentiation in an aged environment is restored by interleukin (IL)-6 blockade or IL-6 deficiency. IL-6 blockade also restores the impaired ability of CD4(+) T cells to promote CD8(+) T-cell-dependent tumour elimination in aged mice, which requires IFN-γ. Furthermore, IL-6-stimulated production of IL-4/IL-21 through c-Maf induction is responsible for impaired Th1 differentiation. IL-6 also contributes to IL-10 production from CD4(+) T cells in aged mice, causing attenuated responses of CD8(+) T cells. These findings suggest that IL-6 serves as an extrinsic factor counteracting CD4(+) T-cell-mediated immunity against tumour in old age.


Subject(s)
Aging/immunology , Cell Differentiation/immunology , Fibrosarcoma/immunology , Interleukin-6/immunology , Th1 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular/immunology , Interferon-gamma/immunology , Interleukin-10/immunology , Interleukin-4/immunology , Interleukins/immunology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Proto-Oncogene Proteins c-maf/immunology , Tumor Escape
5.
Stem Cell Res ; 13(3 Pt A): 442-53, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25460605

ABSTRACT

The purpose of this study was to evaluate the therapeutic potential of human induced pluripotent stem (iPS) cell-derived macrophage-like cells for Alzheimer's disease (AD). In previous studies, we established the technology to generate macrophage-like myeloid lineage cells with proliferating capacity from human iPS cells, and we designated the cells iPS-ML. iPS-ML reduced the level of Aß added into the culture medium, and the culture supernatant of iPS-ML alleviated the neurotoxicity of Aß. We generated iPS-ML expressing the Fc-receptor-fused form of a single chain antibody specific to Aß. In addition, we made iPS-ML expressing Neprilysin-2 (NEP2), which is a protease with Aß-degrading activity. In vitro, expression of NEP2 but not anti-Aß scFv enhanced the effect to reduce the level of soluble Aß oligomer in the culture medium and to alleviate the neurotoxicity of Aß. To analyze the effect of iPS-ML expressing NEP2 (iPS-ML/NEP2) in vivo, we intracerebrally administered the iPS-ML/NEP2 to 5XFAD mice, which is a mouse model of AD. We observed significant reduction in the level of Aß in the brain interstitial fluid following administration of iPS-ML/NEP2. These results suggested that iPS-ML/NEP2 may be a potential therapeutic agent in the treatment of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Induced Pluripotent Stem Cells/cytology , Macrophages/metabolism , Neprilysin/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Antigens, CD/metabolism , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Flow Cytometry , Hippocampus/metabolism , Hippocampus/pathology , Humans , Induced Pluripotent Stem Cells/transplantation , Macrophages/cytology , Macrophages/immunology , Mice , Microscopy, Fluorescence , Neprilysin/genetics , Transplantation, Heterologous
6.
J Immunol ; 193(4): 2024-33, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25031460

ABSTRACT

We established a method to generate a large quantity of myeloid lineage cells from mouse embryonic stem (ES) cells, termed ES cell-derived proliferating myeloid cell lines (ES-ML). ES-ML continuously proliferated in the presence of M-CSF and GM-CSF. ES-ML genetically modified to express an anti-HER2 (neu) mAb single-chain V region fragment reduced the number of cocultured mouse Colon-26 cancer cells expressing HER2. Stimulation of ES-ML with IFN-γ plus LPS or TNF resulted in almost complete killing of the Colon-26 cells by the ES-ML, and the cytotoxicity was mediated, in part, by NO produced by ES-ML. When ES-ML were injected into mice with i.p. established Colon-26 tumors, they efficiently infiltrated the tumor tissues. Injection of ES-ML with rIFN-γ and LPS inhibited cancer progression in the mouse peritoneal cavity. Coinjection of TNF-transfected or untransfected ES-ML with rIFN-γ inhibited cancer growth and resulted in prolonged survival of the treated mice. In this experiment, transporter associated with Ag processing (TAP)1-deficient ES-ML exhibited therapeutic activity in MHC-mismatched allogeneic recipient mice. Despite the proliferative capacity of ES-ML, malignancy never developed from the transferred ES-ML in the recipient mice. In summary, TAP-deficient ES-ML with anticancer properties exhibited a therapeutic effect in allogeneic recipients, suggesting the possible use of TAP-deficient human-induced pluripotent stem cell-derived proliferating myeloid cell lines in cancer therapy.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Colonic Neoplasms/therapy , Embryonic Stem Cells/immunology , Macrophages/immunology , Animals , Antibodies, Monoclonal/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Cytotoxicity, Immunologic , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interferon-gamma/pharmacology , Lipopolysaccharides , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pluripotent Stem Cells/immunology , Receptor, ErbB-2/immunology , Single-Chain Antibodies/immunology , Transplantation, Homologous
7.
Oncoimmunology ; 3(1): e27927, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24800175

ABSTRACT

We established a method to produce a large quantity of myeloid cells from human inducible pluripotent stem cells (iPSCs). When injected intraperitoneally into mice carrying established peritoneal tumors, iPSC-derived myeloid cells (iPS-MCs) efficiently accumulated within neoplastic lesions. The intraperitoneal injection of iPS-MCs expressing interferon ß significantly inhibited the growth of human gastric and pancreatic cancers implanted in the peritoneal cavity of immunocompromised mice.

8.
Vet Microbiol ; 167(3-4): 364-71, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24139177

ABSTRACT

The bovine leukemia virus (BLV) Tax protein is believed to play a crucial role in leukemogenesis by the virus. BLV usually causes asymptomatic infections in cattle, but only one-third develop persistent lymphocytosis that rarely progress after a long incubation period to lymphoid tumors, namely enzootic bovine leucosis (EBL). In the present study, we demonstrated that the BLV tax genes could be divided into two alleles and developed multiplex PCR detecting an L233P mutation of the Tax protein. Then, in order to define the relationship between the Tax protein and leukemogenicity, we examined 360 tumor samples randomly collected from dairy or breeding cattle in Japan, of which Tax proteins were categorized, for age at the time of diagnosis of EBL. The ages of 288 animals (80.0%) associated with L233-Tax and those of 70 animals (19.4%) with P233-Tax individually followed log-normal distributions. Only the two earliest cases (0.6%) with L233-Tax disobeyed the log-normal distribution. These findings suggest that the animals affected by EBL were infected with the virus at a particular point in life, probably less than a few months after birth. Median age of those with P233-Tax was 22 months older than that with L233-Tax and geometric means exhibited a significant difference (P<0.01). It is also quite unlikely that viruses carrying the particular Tax protein infect older cattle. Here, we conclude that BLV could be divided into two categories on the basis of amino acid at position 233 of the Tax protein, which strongly correlated with leukemogenicity.


Subject(s)
Enzootic Bovine Leukosis/virology , Gene Products, tax/genetics , Leukemia Virus, Bovine/genetics , Leukemia Virus, Bovine/pathogenicity , Mutation/genetics , Age Distribution , Alleles , Animals , Cattle , Female , Gene Expression Regulation, Viral , Genetic Variation , Japan , Phylogeny , Polymerase Chain Reaction
9.
Hum Immunol ; 74(10): 1400-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23811433

ABSTRACT

Anticancer vaccination therapies with monocyte-derived dendritic cells (DC) are widely conducted. A large number of primary monocytes (approximately 10(8) cells) are needed to generate the number of DC required to achieve an effect upon vaccination, and monocytes are usually purified from peripheral blood mononuclear cells obtained by apheresis procedure, which is somehow invasive for cancer patients. As a means to facilitate the generation of DC for therapeutic use, we herein report a method to amplify human monocytes. We found that lentivirus-mediated transduction of cMYC along with BMI1 induced proliferation of CD14(+) monocytes derived from 9 out of 12 blood donors, and we named the monocyte-derived proliferating cells CD14-ML. Their proliferation continued for 3-5 weeks in the presence of M-CSF and GM-CSF, resulting in 20-1000-fold amplification. Importantly, the expanded CD14-ML differentiated into fully functional DC (CD14-ML-DC) upon the addition of IL-4 to the culture. We successfully stimulated autologous CD8(+) T cells with CD14-ML-DC pulsed with cytomegalovirus peptide or MART-1 peptide to generate antigen-specific CTL lines. This is the first report describing the method for in vitro expansion of human peripheral blood monocytes.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/metabolism , Gene Expression , Monocytes/cytology , Monocytes/metabolism , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins c-myc/genetics , Antigen Presentation/immunology , Cancer Vaccines/immunology , Cell Differentiation , Cell Line , Cytomegalovirus/immunology , Dendritic Cells/immunology , Humans , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , MART-1 Antigen/immunology , Monocytes/immunology , Myeloid Cells/cytology , Myeloid Cells/immunology , Myeloid Cells/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
10.
PLoS One ; 8(6): e67567, 2013.
Article in English | MEDLINE | ID: mdl-23826321

ABSTRACT

We recently developed a method to generate myeloid cells with proliferation capacity from human iPS cells. iPS-ML (iPS-cell-derived myeloid/macrophage line), generated by introducing proliferation and anti-senescence factors into iPS-cell-derived myeloid cells, grew continuously in an M-CSF-dependent manner. A large number of cells exhibiting macrophage-like properties can be readily obtained by using this technology. In the current study, we evaluated the possible application of iPS-ML in anti-cancer therapy. We established a model of peritoneally disseminated gastric cancer by intraperitoneally injecting NUGC-4 human gastric cancer cells into SCID mice. When iPS-ML were injected intraperitoneally into the mice with pre-established peritoneal NUGC-4 tumors, iPS-ML massively accumulated and infiltrated into the tumor tissues. iPS-ML expressing IFN-ß (iPS-ML/IFN-ß) significantly inhibited the intra-peritoneal growth of NUGC-4 cancer. Furthermore, iPS-ML/IFN-ß also inhibited the growth of human pancreatic cancer MIAPaCa-2 in a similar model. iPS-ML are therefore a promising treatment agent for peritoneally disseminated cancers, for which no standard treatment is currently available.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Interferon-beta/metabolism , Myeloid Cells/metabolism , Myeloid Cells/transplantation , Peritoneal Neoplasms/therapy , Stem Cell Transplantation , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Cell Proliferation , Cytokines/pharmacology , Humans , Injections, Intraperitoneal , Macrophages/pathology , Mice , Mice, SCID , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Peritoneal Neoplasms/secondary , Peritoneum/metabolism , Peritoneum/pathology , Receptor, ErbB-2/metabolism , Single-Chain Antibodies/metabolism
11.
Arch Virol ; 156(7): 1137-41, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21387204

ABSTRACT

Bovine leukemia virus (BLV) infection in cattle causes persistent lymphocytosis, and a few percent of infected animals develop lymphoid tumors, namely enzootic bovine leukosis (EBL). In this study, a 440-bp fragment of the env gene was amplified from 204 tumor samples collected from different regions of Japan and analyzed by restriction fragment length polymorphism (RFLP) to determine the association of BLV with EBL. Of the seven RFLP types defined, types I, II, and III were dominant and found in 12.7, 75.0, and 8.3% of tumor samples, respectively. Cattle harboring type III virus were significantly older than other animals at the time of diagnosis of EBL. Type III viruses were found in approximately 33% and 5.5% of Japanese Black and Holstein cattle, respectively, with EBL. These findings indicate that genetically distinct BLV was associated with EBL in Japan and that the genetic profile may influence the leukemogenicity of the virus.


Subject(s)
Enzootic Bovine Leukosis/virology , Genetic Heterogeneity , Leukemia Virus, Bovine/genetics , Animals , Cattle , Japan , Leukemia Virus, Bovine/isolation & purification , Polymorphism, Restriction Fragment Length , Viral Envelope Proteins/genetics
12.
Virus Res ; 155(1): 343-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21095208

ABSTRACT

Bovine leukemia virus (BLV) infection of cattle has been increasing yearly in Japan although several European countries have successfully eradicated the infection. In the present study, phylogenetic analysis on the env gene obtained from 64 tumor samples found in different regions in Japan was carried out in order to define the genetic background of BLV strains prevailing in the country. Most of the Japanese isolates were found to reside in the consensus cluster or genotype 1 of BLV strains (Rodriguez et al., 2009). Out of them, 21 isolates and 10 isolates exhibited the identical sequences, respectively. Only one isolate was classified into the different genotype related to the US isolates. Analysis on the deduced amino acids of gp51 demonstrated the sequence diversity in the neutralizing domain. These data may indicate that two major populations of BLV prevailed throughout Japan, whereas antigenic variants also exist. It was further proved that multiple invasion of the genetically different BLV strains have occurred in Japan.


Subject(s)
Enzootic Bovine Leukosis/epidemiology , Enzootic Bovine Leukosis/virology , Gene Products, env/genetics , Leukemia Virus, Bovine/classification , Leukemia Virus, Bovine/genetics , Animals , Antibodies, Neutralizing/immunology , Cattle , Cluster Analysis , Epitopes/genetics , Japan/epidemiology , Leukemia Virus, Bovine/immunology , Leukemia Virus, Bovine/isolation & purification , Molecular Epidemiology , Molecular Sequence Data , Phylogeny , RNA, Viral/genetics , Sequence Analysis, DNA , Sequence Homology
13.
Drug Metab Pharmacokinet ; 19(2): 120-9, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15499178

ABSTRACT

The purpose of this study was to evaluate a contribution of polymorphic cytochrome P450 (CYP) 3A5 to the oxidation of diltiazem, midazolam and testosterone by liver microsomes from Japanese subjects. Twenty-seven liver samples were classified into three groups according to the CYP3A5 genotypes; CYP3A5(*)1/(*)1 (n=3), (*)1/(*)3 (n=12) and (*)3/(*)3 (n=12). The results of genotyping and immunochemical quantitation of CYP3A5 protein showed a good accordance between the CYP3A5 genotype and CYP3A5 content but not CYP3A4 content in liver microsomes. The expression levels of hepatic CYP3A5 protein ranged from 20 to 60% of the sum of CYP3A4 and CYP3A5 contents in subjects with at least one wild type allele ((*)1). The CYP3A5 contents correlated well with liver microsomal activities of diltiazem N-demethylation, midazolam 1'- and 4-hydroxylations and testosterone 6beta-hydroxylation among subjects carrying at least one (*)1 allele. In addition, the correlation coefficients of CYP3A5 contents with the rates of diltiazem N-demethylation, midazolam 1'-hydroxylation and testosterone 6beta- hydroxylation were higher than those of CYP3A4, although the value of CYP3A5 with the midazolam 4-hydroxylation rate was similar to that of CYP3A4. Kinetic analyses revealed a biphasic diltiazem N-demethylation in liver microsomes from subjects carrying the (*)1 allele. The apparent V(max)/K(m) values for recombinant CYP3A5 indicated the greater contributions to diltiazem N-demethylation and midazolam 1'-hydroxylation as compared with CYP3A4. These results suggest that polymorphic CYP3A5 contributes markedly to the drug oxidations, particularly diltiazem N-demethylation, midazolam 1'- hydroxylation and testosterone 6beta-hydroxylation by liver microsomes from Japanese subjects.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/enzymology , Pharmaceutical Preparations/metabolism , Calcium Channel Blockers/metabolism , Cytochrome P-450 CYP3A , DNA/genetics , DNA Primers , Dealkylation , Diltiazem/metabolism , Escherichia coli/metabolism , GABA Modulators/metabolism , Genotype , Humans , Hydroxylation , In Vitro Techniques , Japan , Kinetics , Midazolam/metabolism , NADPH-Ferrihemoprotein Reductase/genetics , NADPH-Ferrihemoprotein Reductase/metabolism , Oxidation-Reduction , Reverse Transcriptase Polymerase Chain Reaction , Testosterone/metabolism
14.
Mol Pharmacol ; 65(2): 326-34, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14742674

ABSTRACT

CYP3A4, the most abundant form of cytochrome P450 in the human adult liver, shows wide interindividual variation in its activity. This variability is thought to be caused largely by transcriptional and genetic factors, yet the underlying mechanisms are poorly understood. The purpose of this study was to clarify the mechanisms controlling the CYP3A4 gene transcription and to search for genetic polymorphisms in the 5'-flanking region of the CYP3A4 gene. Transient transfection of human hepatoma HepG2 cells and of normal human hepatocytes with a series of CYP3A4 promoter-luciferase reporter plasmids revealed that a region from -11.4 to -10.5 kilobases, designated the constitutive liver enhancer module of CYP3A4 (CLEM4), was important for the constitutive activation of the CYP3A4 gene. Gel shift assay using nuclear extracts prepared from HepG2 cells showed that HNF-1alpha, HNF-4alpha, USF1, and AP-1 interacted with CLEM4. Furthermore, the introduction of mutations into their binding sites demonstrated that essentially all sites were required for the maximal enhancer activity. Screening for genetic polymorphisms within CLEM4 in genomic DNA from French persons, we identified the novel variant, TGT insertion between -11,129 and -11,128 (-11,129_-11,128insTGT), whose allele frequency was 3.1%. The -11,129_-11,128insTGT resulted in the disruption of USF1 binding and a 36% reduction of the enhancer activity. These results suggest that CLEM4 is a constitutive enhancer of the CYP3A4 gene in the liver and that -11,129_-11,128insTGT may at least partly contribute to the interindividual variability of CYP3A4 expression.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Enhancer Elements, Genetic/genetics , Polymorphism, Genetic/genetics , 5' Flanking Region/genetics , Base Sequence , Binding Sites/genetics , Cell Line, Tumor , Cytochrome P-450 CYP3A , Genetic Variation/genetics , Humans , Molecular Sequence Data
15.
Pharmacogenetics ; 13(11): 689-95, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14583682

ABSTRACT

One of seven poor metabolizers of coumarin found in Thai subjects was previously genotyped as heterozygote for the CYP2A6*4 (whole deletion) and CYP2A6*9. Thus, we aimed to investigate the relationship between the genetic polymorphism in the TATA box of the CYP2A6 gene (CYP2A6*9), expression levels of CYP2A6 mRNA and coumarin 7-hydroxylase activities in human livers. Levels of CYP2A6 mRNA were quantified by real-time quantitative reverse transcriptase-polymerase chain reaction. The mean expression levels of CYP2A6 mRNA in individuals with CYP2A6*1/*4, CYP2A6*1/*9 and CYP2A6*4/*9 were 58%, 71% and 21% of the individuals genotyped as CYP2A6*1/*1, respectively. The mean in-vitro coumarin 7-hydroxylase activities in subjects carrying CYP2A6*1/*4, CYP2A6*1/*9 and CYP2A6*4/*9 were 41%, 71% and 12%, respectively, compared to those of the subjects judged as wild-type. Vmax values for coumarin 7-hydroxylation in the liver microsomes from human subjects with genotypes of CYP2A6*1/*1, CYP2A6*1/*4, CYP2A6*1/*9 and CYP2A6*4/*9 were 0.58, 0.26, 0.44 and 0.13 nmol/min/nmol total P450, respectively. CYP2A6 protein levels in human liver microsomes with the CYP2A6*4 and the CYP2A6*9 alleles were markedly decreased. These results suggest that the genetic polymorphism in the promoter region of the CYP2A6 gene (CYP2A6*9) reduced the expression levels of CYP2A6 mRNA and protein in human livers, resulting in the decrease of coumarin 7-hydroxylase activities. Individuals judged as CYP2A6*4/*9 were expected to be poor metabolizers, having extremely low activity of CYP2A6.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Microsomes, Liver/metabolism , Mixed Function Oxygenases/metabolism , Polymorphism, Genetic , Promoter Regions, Genetic , Adult , Alleles , Asian People , Cytochrome P-450 CYP2A6 , Female , Gene Frequency , Heterozygote , Humans , Kinetics , Male , Microsomes, Liver/enzymology , Middle Aged , Polymorphism, Single Nucleotide , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...