Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
Epigenetics Chromatin ; 16(1): 37, 2023 10 04.
Article in English | MEDLINE | ID: mdl-37794499

ABSTRACT

BACKGROUND: Genome-wide DNA methylation (DNAme) profiling of the placenta with Illumina Infinium Methylation bead arrays is often used to explore the connections between in utero exposures, placental pathology, and fetal development. However, many technical and biological factors can lead to signals of DNAme variation between samples and between cohorts, and understanding and accounting for these factors is essential to ensure meaningful and replicable data analysis. Recently, "epiphenotyping" approaches have been developed whereby DNAme data can be used to impute information about phenotypic variables such as gestational age, sex, cell composition, and ancestry. These epiphenotypes offer avenues to compare phenotypic data across cohorts, and to understand how phenotypic variables relate to DNAme variability. However, the relationships between placental epiphenotyping variables and other technical and biological variables, and their application to downstream epigenome analyses, have not been well studied. RESULTS: Using DNAme data from 204 placentas across three cohorts, we applied the PlaNET R package to estimate epiphenotypes gestational age, ancestry, and cell composition in these samples. PlaNET ancestry estimates were highly correlated with independent polymorphic ancestry-informative markers, and epigenetic gestational age, on average, was estimated within 4 days of reported gestational age, underscoring the accuracy of these tools. Cell composition estimates varied both within and between cohorts, as well as over very long placental processing times. Interestingly, the ratio of cytotrophoblast to syncytiotrophoblast proportion decreased with increasing gestational age, and differed slightly by both maternal ethnicity (lower in white vs. non-white) and genetic ancestry (lower in higher probability European ancestry). The cohort of origin and cytotrophoblast proportion were the largest drivers of DNAme variation in this dataset, based on their associations with the first principal component. CONCLUSIONS: This work confirms that cohort, array (technical) batch, cell type proportion, self-reported ethnicity, genetic ancestry, and biological sex are important variables to consider in any analyses of Illumina DNAme data. We further demonstrate the specific utility of epiphenotyping tools developed for use with placental DNAme data, and show that these variables (i) provide an independent check of clinically obtained data and (ii) provide a robust approach to compare variables across different datasets. Finally, we present a general framework for the processing and analysis of placental DNAme data, integrating the epiphenotype variables discussed here.


Subject(s)
DNA Methylation , Placenta , Humans , Pregnancy , Female , Infant, Newborn , Placenta/metabolism , Epigenesis, Genetic , Gestational Age , Genome
3.
Clin Transl Sci ; 9(1): 51-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26842729

ABSTRACT

Resistance to cytarabine remains a major challenge in the treatment of acute myeloid leukemia (AML). Based on previous studies implicating ABCC4/MRP4 in the transport of nucleosides, we hypothesized that cytarabine is sensitive to ABCC4-mediated efflux, thereby decreasing its cytotoxic response against AML blasts. The uptake of cytarabine and its monophosphate metabolite was found to be facilitated in ABCC4-expressing vesicles and intracellular retention was significantly impaired by overexpression of human ABCC4 or mouse Abcc4 (P < 0.05). ABCC4 was expressed highly in AML primary blasts and cell lines, and cytotoxicity of cytarabine in cells was increased in the presence of the ABCC4 inhibitors MK571 or sorafenib, as well as after ABCC4 siRNA. In Abcc4-null mice, cytarabine-induced hematological toxicity was enhanced and ex vivo colony-forming assays showed that Abcc4-deficiency sensitized myeloid progenitors to cytarabine. Collectively, these studies demonstrate that ABCC4 plays a protective role against cytarabine-mediated insults in leukemic and host myeloid cells.


Subject(s)
Cytarabine/pharmacology , Multidrug Resistance-Associated Proteins/metabolism , Myeloid Progenitor Cells/pathology , Animals , Biological Transport/drug effects , Cell Death/drug effects , Cell Line, Tumor , Child, Preschool , Gene Knockdown Techniques , Gene Silencing/drug effects , Humans , Leukemia, Myeloid, Acute/pathology , Mice, Inbred C57BL , Myeloid Progenitor Cells/drug effects , Myeloid Progenitor Cells/metabolism
4.
Leukemia ; 26(6): 1383-90, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22189900

ABSTRACT

BCL2 is deregulated in diffuse large B-cell lymphoma (DLBCL) by the t(14;18) translocation, gene amplification and/or nuclear factor-κB signaling. RNA-seq data have recently shown that BCL2 is the most highly mutated gene in germinal center B-cell (GCB) DLBCL. We have sequenced BCL2 in 298 primary DLBCL biopsies, 131 additional non-Hodgkin lymphoma biopsies, 24 DLBCL cell lines and 51 germline DNAs. We found frequent BCL2 mutations in follicular lymphoma (FL) and GCB DLBCL, but low levels of BCL2 mutations in activated B-cell DLBCL, mantle cell lymphoma, small lymphocytic leukemia and peripheral T-cell lymphoma. We found no BCL2 mutations in GC centroblasts. Many mutations were non-synonymous; they were preferentially located in the flexible loop domain, with few in BCL2-homology domains. An elevated transition/transversions ratio supports that the mutations result from somatic hypermutation. BCL2 translocations correlate with, and are likely important in acquisition of, additional BCL2 mutations in GCB DLBCL and FL. DLBCL mutations were not independently associated with survival. Although previous studies of BCL2 mutations in FL have reported mutations to result in pseudo-negative BCL2 protein expression, we find this rare in de-novo DLBCL.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Mantle-Cell/genetics , Lymphoma, T-Cell, Peripheral/genetics , Mediastinal Neoplasms/genetics , Mutation/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Cytogenetic Analysis , Humans , Immunoenzyme Techniques , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Mantle-Cell/metabolism , Lymphoma, T-Cell, Peripheral/metabolism , Mediastinal Neoplasms/metabolism , Protein Transport , Proto-Oncogene Proteins c-bcl-2/metabolism
5.
Leukemia ; 25(10): 1570-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21647154

ABSTRACT

To investigate the frequency of isocitrate dehydrogenase 1 (IDH1) and 2 (IDH2) mutations in pediatric acute myeloid leukemia (AML) and acute lymphoid leukemia (ALL), we sequenced these genes in diagnostic samples from 515 patients (227 AMLs and 288 ALLs). Somatic IDH1/IDH2 mutations were rare in ALL (N=1), but were more common in AML, occurring in 3.5% (IDH1 N=3 and IDH2 N=5), with the frequency higher in AMLs with a normal karyotype (9.8%). The identified IDH1 mutations occurred in codon 132 resulting in replacement of arginine with either cysteine (N=3) or histidine (N=1). By contrast, mutations in IDH2 did not affect the homologous residue but instead altered codon 140, resulting in replacement of arginine with either glutamine (N=4) or tryptophan (N=1). Structural modeling of IDH2 suggested that codon 140 mutations disrupt the enzyme's ability to bind its substrate isocitrate. Accordingly, recombinant IDH2 R140Q/W were unable to carry out the decarboxylation of isocitrate to α-ketoglutarate (α-KG), but instead gained the neomorphic activity to reduce α-KG to R(-)-2-hydroxyglutarete (2-HG). Analysis of primary leukemic blasts confirmed high levels of 2-HG in AMLs with IDH1/IDH2 mutations. Interestingly, 3/5 AMLs with IDH2 mutations had FLT3-activating mutations, raising the possibility that these mutations cooperate in leukemogenesis.


Subject(s)
Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/genetics , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Base Sequence , Child , Chromatography, Ion Exchange , DNA Primers , Humans , In Situ Hybridization, Fluorescence , Leukemia, Myeloid, Acute/enzymology , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Tandem Mass Spectrometry
6.
Annu Rev Pharmacol Toxicol ; 46: 381-410, 2006.
Article in English | MEDLINE | ID: mdl-16402910

ABSTRACT

The protein variously named ABCG2/BCRP/MXR/ABCP is a recently described ATP-binding cassette (ABC) transporter originally identified by its ability to confer drug resistance that is independent of Mrp1 (multidrug-resistance protein 1) and Pgp (P-glycoprotein). Unlike Mrp1 and Pgp, ABCG2 is a half-transporter that must homodimerize to acquire transport activity. ABCG2 is found in a variety of stem cells and may protect them from exogenous and endogenous toxins. ABCG2 expression is upregulated under low-oxygen conditions, consistent with its high expression in tissues exposed to low-oxygen environments. ABCG2 interacts with heme and other porphyrins and protects cells and/or tissues from protoporphyrin accumulation under hypoxic conditions. Individuals who carry ABCG2 alleles that have impaired function may be more susceptible to porphyrin-induced toxicity. Abcg2 knock-out models have allowed in vivo studies of Abcg2 function in host and cellular defense. In combination with immunohistochemical analyses, these studies have revealed how ABCG2 influences the absorption, distribution, and excretion of drugs and cytotoxins.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/physiology , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Chromosomes/genetics , Cloning, Molecular , Drug Therapy , Gene Expression Regulation , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Stem Cells/physiology
7.
Article in English | MEDLINE | ID: mdl-15364287

ABSTRACT

The estrildid finches (Aves: Passeriformes: Estrildidae) of Africa, Asia, and Australia have been the focus of several recent tests of sexual selection theory. Many estrildids display bright red, orange, or yellow colors in the beak or plumage, which typically are generated by the presence of carotenoid pigments. In this study, we used high-performance liquid chromatography to investigate the carotenoid content of feathers and other colorful tissues in seven species of estrildids. Star finches (Neochmia ruficauda) and diamond firetails (Stagonopleura guttata) circulated two main dietary carotenoids (lutein and zeaxanthin) through the blood and liver and used both to acquire a yellow plumage color. However, five other estrildids (common waxbill, Estrilda astrild; black-rumped waxbill, Estrilda troglodytes; zebra waxbill, Amandava subflava; red avadavat, Amandava amandava; and zebra finch, Taeniopygia guttata) circulated these same dietary carotenoids along with two metabolites (dehydrolutein and anhydrolutein) through the blood and/or liver and used all four as yellow plumage colorants. We subsequently tracked the distribution of these pigments using a published phylogeny of estrildid finches to determine the evolutionary pattern of carotenoid metabolism in these birds. We found that finches from the most ancient tribe of estrildids (Estrildini) possessed the ability to metabolize dietary carotenoids. Although carotenoids from the most ancestral extant estrildid species have yet to be analyzed, we hypothesize (based on their relationships with other songbirds known to have such metabolic capabilities) that these finches inherited from their ancestors the capability to metabolize carotenoids. Interestingly, later in estrildid evolution, certain taxa lost the ability to metabolize dietary carotenoids (e.g., in the Poephilini), suggesting that the occurrence of carotenoid metabolism can be labile and is likely shaped by the relative costs and benefits of color signaling across different species.


Subject(s)
Biological Evolution , Carotenoids/analysis , Finches/physiology , Pigmentation/physiology , Animals , Carotenoids/blood , Feathers/chemistry , Female , Finches/classification , Liver Extracts/chemistry , Male , Phylogeny , Sex Characteristics , Species Specificity
9.
AAPS PharmSci ; 4(3): E14, 2002.
Article in English | MEDLINE | ID: mdl-12423063

ABSTRACT

Nucleotide efflux (especially cyclic nucleotides) from a variety of mammalian tissues, bacteria, and lower eukaryotes has been studied for several decades. However, the molecular identity of these nucleotide efflux transporters remained elusive, despite extensive knowledge of their kinetic properties and inhibitor profiles. Identification of the subfamily of adenosine triphosphate (ATP) binding cassette transporters, multidrug resistance protein (MRP) subfamily, permitted rapid advances because some recently identified MRP family members transport modified nucleotide analogs (ie, chemotherapeutic agents). We first identified, MRP4, based on its ability to efflux antiretroviral compounds, such as azidothymidine monophosphate (AZT-MP) and 9-(2-phosphonyl methoxyethyl) adenine (PMEA), in drug-resistant and also in transfected cell lines. MRP5, a close structural homologue of MRP4 also transported PMEA. MRP4 and MRP5 confer resistance to cytotoxic thiopurine nucleotides, and we demonstrate MRP4 expression varies among acute lymphoblastic leukemias, suggesting this as a factor in response to chemotherapy with these agents. The ability of MRP4 and MRP5 to transport 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP) suggests they may play a biological role in cellular signaling by these nucleotides. Finally, we propose that MRP4 may also play a role in hepatic bile acid homeostasis because loss of the main bile acid efflux transporter, sister of P-glycoprotein (SPGP) aka bile-salt export pump (BSEP), leads to a strong compensatory upregulation in MRP4 expression. Cumulatively, these studies reveal that the ATP-binding cassette (ABC) transporters MRP4 and MRP5 have a unique role in biology and in chemotherapeutic response.


Subject(s)
Drug Therapy , Multidrug Resistance-Associated Proteins/physiology , Animals , Biological Transport, Active/physiology , Drug Resistance, Neoplasm/physiology , Humans , Multidrug Resistance-Associated Proteins/genetics , Nucleotides, Cyclic/metabolism , T-Lymphocytes/chemistry , T-Lymphocytes/metabolism
10.
Mol Pharmacol ; 62(6): 1321-31, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12435799

ABSTRACT

Mercaptopurines have been used as anticancer agents for more than 40 years, and most acute lymphoblastic leukemias are treated with 6-mercaptopurine (6MP) or 6-thioguanine (TG). Overexpression of the two related multidrug resistance proteins MRP4 and MRP5 has been shown to confer some resistance against mercaptopurines, which has been attributed to extrusion of mercaptopurine metabolites by these transporters. We have analyzed the mercaptopurine metabolites formed in human embryonic kidney cells and determined which metabolites are extruded by MRP4 and MRP5. Incubation with 6MP led to the formation of thioinosine and thioxanthosine metabolites and we found that thio-IMP was transported by both MRP4 and MRP5; MRP5 showed the highest transport rate. In contrast, only MRP5 transported thioxanthosine monophosphate (tXMP). During incubation with TG, the monophosphorylated form of thioguanosine was transported by both MRP4 and MRP5; the highest transport rate was for MRP4. Similarly, only 6-methyl-thio-IMP was formed during incubation with 6-methyl mercaptopurine riboside. This compound was a substrate for both MRP4 and MRP5; MRP4 showed the highest transport rate. Our results show that all major thiopurine monophosphates important in the efficacy of mercaptopurine treatment are transported by MRP4 and MRP5, although the substrate specificity of the two transporters differs in detail.


Subject(s)
Mercaptopurine/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Ribosomal Proteins/metabolism , Thioguanine/metabolism , Biological Transport , Cells, Cultured , Chromatography, High Pressure Liquid , Drug Interactions , Humans , Kidney/cytology , Kidney/embryology , Kinetics , Mercaptopurine/pharmacology , Methylthioinosine/pharmacology , Multidrug Resistance-Associated Proteins/biosynthesis , Ribosomal Proteins/biosynthesis , Thioguanine/pharmacology , Transfection
11.
Biochem J ; 368(Pt 1): 325-32, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12133003

ABSTRACT

Cellular factors may contribute to the decreased efficacy of chemotherapy in HIV infection. Indeed, prolonged treatment with nucleoside analogues, such as azidothymidine (AZT), 2',3'-deoxycytidine or 9-(2-phosphonylmethoxyethyl)adenine, induces cellular resistance. We have developed a human T lymphoblastoid cell line (CEM 3TC) that is selectively resistant to the antiproliferative effect of 2',3'-dideoxy-3'-thiacytidine (3TC) because the CEM 3TC cells were equally sensitive to AZT, as well as the antimitotic agent, vinblastine. The anti-retroviral activity of 3TC against HIV-1 was also severely impaired in the CEM 3TC cells. Despite similar deoxycytidine kinase activity and unchanged uptake of nucleosides such as AZT and 2'-deoxycytidine, CEM 3TC had profoundly impaired 3TC accumulation. Further studies indicated that CEM 3TC retained much less 3TC. However, despite a small overexpression of multidrug resistance protein (MRP) 4, additional studies with cells specifically engineered to overexpress MRP4 demonstrated there was no impact on either 3TC accumulation or efflux. Finally, an increased expression of the MRP5 homologue, ATP-binding cassette C11 (ABCC11) was observed in the CEM 3TC cells. We speculate that the decreased 3TC accumulation in the CEM 3TC might be due to the upregulation of ABCC11.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Drug Resistance, Multiple/physiology , Lamivudine/pharmacokinetics , Multidrug Resistance-Associated Proteins/metabolism , T-Lymphocytes/metabolism , Anti-HIV Agents/pharmacology , Biological Transport , Deoxycytidine Kinase/metabolism , HIV/drug effects , Humans , Nucleosides/metabolism , Tumor Cells, Cultured , Zidovudine/pharmacology
12.
J Cardiovasc Surg (Torino) ; 43(4): 531-7, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12124569

ABSTRACT

BACKGROUND: Accurate pretreatment staging of esophageal cancer (EC) is important in the evaluation and comparison of results of different treatment modalities. Few studies using minimally invasive staging techniques for this purpose have been reported. We previously demonstrated the usefulness of the thoracoscopic/laparoscopic (Ts/Ls) technique in pretreatment staging of EC. This study was conducted to evaluate the impact of trimodality based on pretreatment Ts/Ls staging diagnosis on EC. METHODS: A retrospective study was performed on 2 groups of EC patients. Group A (44 patients) underwent pretreatment Ts/Ls staging and had trimodality treatment. Preoperative therapy consisted of concurrent chemotherapy (5-FU + cisplatinum) and radiotherapy. Group B (33 patients) underwent surgery alone. The study focused on stratified comparison of patterns of recurrence and survival in different pretreatment surgical T, N, and TNM stage categories. RESULTS: The 3-year disease free survival of Group A was 40.8% with a median survival of 32.0 months, it was 43.6% with a median survival of 23.6 months in Group B. The difference was not significant (p=0.87). There was no difference in recurrence pattern between the 2 groups. Patients with squamous cell carcinoma in Group A had no local recurrence during the follow-up period while those in Group B had a high local recurrence rate of 40% (p<0.005). When stratified by T factor, patients with locally advanced T stage (T3-4) in Group A had a lower distant recurrence rate than their counterpart patients in Group B (9.1 vs 38.5%, p=0.03), they had a better survival but the difference was not significant (3-year disease free survival: 41.7 vs 17.9%, p=0.14). There were no significant differences in recurrence pattern and survival in different N categories and TNM stages between 2 groups. Multivariate analysis showed that only pretreatment surgical N status was an independent prognostic factor for the whole group (p=0.02). CONCLUSIONS: Pretreatment Ts/Ls staging can provide accurate staging information for EC patients. Trimodality treatment was successful in local control for patients with squamous cell carcinoma. It was effective in reducing distant recurrence and might prolong survival in patients with advanced T stages. Pretreatment lymph node status was the most important prognosticator regardless of treatment modality. Pretreatment pathological staging should be included in the future clinical trials on multimodality treatments in EC patients.


Subject(s)
Adenocarcinoma/therapy , Carcinoma, Squamous Cell/therapy , Esophageal Neoplasms/therapy , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Aged , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Case-Control Studies , Combined Modality Therapy , Disease-Free Survival , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophagus/pathology , Female , Humans , Male , Middle Aged , Multivariate Analysis , Neoplasm Recurrence, Local/epidemiology , Neoplasm Staging , Retrospective Studies
13.
Mol Pharmacol ; 60(6): 1399-406, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11723248

ABSTRACT

It was previously shown that CYP3A4 is induced in the human intestinal Caco-2 cell model by treatment with 1alpha,25-dihydroxy vitamin D3 (1,25-D3). We demonstrate the vitamin D analog, 19-nor-1alpha,25-dihydroxy vitamin D2, is also an effective inducer of CYP3A4 in Caco-2 cells, but with half the potency of 1,25-D3. We report that treatment of LS180 cells, a human intestinal cell line, with 1 to 10 nM 1,25-D3 dose dependently increased CYP3A4 protein and CYP3A4 mRNA expression. CYP3A4- and CYP3A23-promoter-Luciferase reporter constructs transiently transfected into LS180 cells were transcriptionally activated in a dose-dependent manner by 1,25-D3, whereas mutation of the nuclear hormone receptor binding motif (ER6) in the CYP3A4 promoter abrogated 1,25-D3 activation of CYP3A4. Although the CYP3A4 ER6 promoter element has been shown to bind the pregnane X receptor (PXR), this receptor does not mediate 1,25-D3 induction of CYP3A4 because a) PXR is not expressed in Caco-2 cells; b) PXR mRNA expression is not induced by 1,25-D3 treatment of LS180 cells; and c) the ligand binding domain of human PXR was not activated by 1,25-D3. 1,25-D3 uses the vitamin D receptor to induce CYP3A4 because a) the vitamin D receptor (VDR)-retinoid X receptor (RXR) heterodimer binds specifically to the CYP3A4 ER6; b) selective mutation of the CYP3A4 ER6 disrupted the binding of VDR-RXR; and c) reporter constructs containing only three copies of the CYP3A4 ER6 linked to a TK-CAT reporter were activated by 1,25-D3 only in cells cotransfected with a human VDR expression plasmid. These data support the hypothesis that 1,25-D3 and VDR induce expression of intestinal CYP3A by binding of the activated VDR-RXR heterodimer to the CYP3A PXR response element and promoting gene transcription.


Subject(s)
Calcitriol/pharmacology , Cytochrome P-450 Enzyme System/biosynthesis , Intestines/drug effects , Mixed Function Oxygenases/biosynthesis , Transcription, Genetic/drug effects , Animals , COS Cells , Caco-2 Cells , Cells, Cultured , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , Dimerization , Dose-Response Relationship, Drug , Enzyme Induction , Ergocalciferols/pharmacology , Haplorhini , Humans , Intestines/enzymology , Luciferases/genetics , Mixed Function Oxygenases/genetics , Pregnane X Receptor , Protein Binding , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Retinoic Acid/metabolism , Receptors, Steroid/metabolism , Retinoid X Receptors , Time Factors , Transcription Factors/metabolism , Transcriptional Activation
14.
Nat Med ; 7(9): 1028-34, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11533706

ABSTRACT

Stem cells from bone marrow, skeletal muscle and possibly other tissues can be identified by the 'side-population' (SP) phenotype. Although it has been assumed that expression of ABC transporters is responsible for this phenotype, the specific molecules involved have not been defined. Here we show that expression of the Bcrp1 (also known as Abcg2 murine/ABCG2 human) gene is a conserved feature of stem cells from a wide variety of sources. Bcrp1 mRNA was expressed at high levels in primitive murine hematopoietic stem cells, and was sharply downregulated with differentiation. Enforced expression of the ABCG2 cDNA directly conferred the SP phenotype to bone-marrow cells and caused a reduction in maturing progeny both in vitro and in transplantation-based assays. These results show that expression of the Bcrp1/ABCG2 gene is an important determinant of the SP phenotype, and that it might serve as a marker for stem cells from various sources.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Macrophage Inflammatory Proteins , Membrane Glycoproteins , Multidrug Resistance-Associated Proteins , Neoplasm Proteins , Stem Cells/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Animals , Antigens, CD/metabolism , Antigens, CD34/metabolism , Biomarkers , Bone Marrow Cells/physiology , Cells, Cultured , Chemokines, CC , Cytokines/metabolism , Female , Humans , Male , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Ribosomal Proteins/metabolism , Stem Cells/cytology , Tetraspanin 29 , Transplants
15.
Clin Pharmacol Ther ; 70(2): 189-99, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11503014

ABSTRACT

MDR1 (P-glycoprotein) is an important factor in the disposition of many drugs, and the involved processes often exhibit considerable interindividual variability that may be genetically determined. Single-strand conformational polymorphism analysis and direct sequencing of exonic MDR1 deoxyribonucleic acid from 37 healthy European American and 23 healthy African American subjects identified 10 single nucleotide polymorphisms (SNPs), including 6 nonsynonymous variants, occurring in various allelic combinations. Population frequencies of the 15 identified alleles varied according to racial background. Two synonymous SNPs (C1236T in exon 12 and C3435T in exon 26) and a nonsynonymous SNP (G2677T, Ala893Ser) in exon 21 were found to be linked (MDR1*2 ) and occurred in 62% of European Americans and 13% of African Americans. In vitro expression of MDR1 encoding Ala893 (MDR1*1 ) or a site-directed Ser893 mutation (MDR1*2 ) indicated enhanced efflux of digoxin by cells expressing the MDR1-Ser893 variant. In vivo functional relevance of this SNP was assessed with the known P-glycoprotein drug substrate fexofenadine as a probe of the transporter's activity. In humans, MDR1*1 and MDR1*2 variants were associated with differences in fexofenadine levels, consistent with the in vitro data, with the area under the plasma level-time curve being almost 40% greater in the *1/*1 genotype compared with the *2/*2 and the *1/*2 heterozygotes having an intermediate value, suggesting enhanced in vivo P-glycoprotein activity among subjects with the MDR1*2 allele. Thus allelic variation in MDR1 is more common than previously recognized and involves multiple SNPs whose allelic frequencies vary between populations, and some of these SNPs are associated with altered P-glycoprotein function.


Subject(s)
Black People/genetics , Genes, MDR/genetics , Polymorphism, Single Nucleotide , Terfenadine/pharmacokinetics , White People/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Africa/ethnology , Alleles , Anti-Allergic Agents/pharmacokinetics , Area Under Curve , Cloning, Molecular , DNA Primers , Digoxin/pharmacokinetics , Enzyme Inhibitors/pharmacokinetics , Europe/ethnology , Genetic Variation , Genotype , Haplotypes , Humans , Polymerase Chain Reaction , Sequence Analysis, DNA , Terfenadine/analogs & derivatives , Time Factors
16.
J Biol Chem ; 276(42): 39411-8, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11509573

ABSTRACT

Sister of P-glycoprotein (SPGP) is the major hepatic bile salt export pump (BSEP). BSEP/SPGP expression varies dramatically among human livers. The potency and hierarchy of bile acids as ligands for the farnesyl/bile acid receptor (FXR/BAR) paralleled their ability to induce BSEP in human hepatocyte cultures. FXR:RXR heterodimers bound to IR1 elements and enhanced bile acid transcriptional activation of the mouse and human BSEP/SPGP promoters. In FXR/BAR nullizygous mice, which have dramatically reduced BSEP/SPGP levels, hepatic CYP3A11 and CYP2B10 were strongly but unexpectedly induced. Notably, the rank order of bile acids as CYP3A4 inducers and activators of pregnane X receptor/steroid and xenobiotic receptor (PXR/SXR) closely paralleled each other but was markedly different from their hierarchy and potency as inducers of BSEP in human hepatocytes. Moreover, the hepatoprotective bile acid ursodeoxycholic acid, which reverses hydrophobic bile acid hepatotoxicity, activates PXR and efficaciously induces CYP3A4 (a bile-metabolizing enzyme) in primary human hepatocytes thus providing one mechanism for its hepatoprotection. Because serum and urinary bile acids increased in FXR/BAR -/- mice, we evaluated hepatic transporters for compensatory changes that might circumvent the profound decrease in BSEP/SPGP. We found weak MRP3 up-regulation. In contrast, MRP4 was substantially increased in the FXR/BAR nullizygous mice and was further elevated by cholic acid. Thus, enhanced hepatocellular concentrations of bile acids, due to the down-regulation of BSEP/SPGP-mediated efflux in FXR nullizygous mice, result in an alternate but apparent compensatory up-regulation of CYP3A, CYP2B, and some ABC transporters that is consistent with activation of PXR/SXR by bile acids.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Cell Nucleus/metabolism , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/genetics , Amino Acid Sequence , Animals , Base Sequence , Bile Acids and Salts/metabolism , Cell Line , Cells, Cultured , Cytochrome P-450 CYP3A , Dimerization , Dose-Response Relationship, Drug , Down-Regulation , Genes, Reporter , Hepatocytes/metabolism , Humans , Immunoblotting , Ligands , Liver/metabolism , Luciferases/metabolism , Mice , Mice, Inbred BALB C , Mice, Transgenic , Mixed Function Oxygenases/metabolism , Molecular Sequence Data , Multidrug Resistance-Associated Proteins/metabolism , Plasmids/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Proteins/metabolism , Sequence Homology, Nucleic Acid , Transfection , Up-Regulation , Ursodeoxycholic Acid/pharmacology
17.
Gene ; 273(1): 89-96, 2001 Jul 25.
Article in English | MEDLINE | ID: mdl-11483364

ABSTRACT

Several years ago, we initiated a long-term project of cloning new human ATP-binding cassette (ABC) transporters and linking them to various disease phenotypes. As one of the results of this project, we present two new members of the human ABCC subfamily, ABCC11 and ABCC12. These two new human ABC transporters were fully characterized and mapped to the human chromosome 16q12. With the addition of these two genes, the complete human ABCC subfamily has 12 identified members (ABCC1-12), nine from the multidrug resistance-like subgroup, two from the sulfonylurea receptor subgroup, and the CFTR gene. Phylogenetic analysis determined that ABCC11 and ABCC12 are derived by duplication, and are most closely related to the ABCC5 gene. Genetic variation in some ABCC subfamily members is associated with human inherited diseases, including cystic fibrosis (CFTR/ABCC7), Dubin-Johnson syndrome (ABCC2), pseudoxanthoma elasticum (ABCC6) and familial persistent hyperinsulinemic hypoglycemia of infancy (ABCC8). Since ABCC11 and ABCC12 were mapped to a region harboring gene(s) for paroxysmal kinesigenic choreoathetosis, the two genes represent positional candidates for this disorder.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Chromosomes, Human, Pair 16 , Amino Acid Sequence , Base Sequence , Cell Line , Chromosome Mapping , Cloning, Molecular , Humans , Molecular Sequence Data , Multidrug Resistance-Associated Protein 2 , Phylogeny
18.
J Biol Chem ; 276(42): 39359-67, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11483599

ABSTRACT

The most frequently expressed drug resistance genes, MDR1 and MRP1, occur in human tumors with mutant p53. However, it was unknown if mutant p53 transcriptionally regulated both MDR1 and MRP1. We demonstrated that mutant p53 did not activate either the MRP1 promoter or the endogenous gene. In contrast, mutant p53 strongly up-regulated the MDR1 promoter and expression of the endogenous MDR1 gene. Notably, cells that expressed either a transcriptionally inactive mutant p53 or the empty vector showed no endogenous MDR1 up-regulation. Transcriptional activation of the MDR1 promoter by mutant p53 required an Ets binding site, and mutant p53 and Ets-1 synergistically activated MDR1 transcription. Biochemical analysis revealed that Ets-1 interacted exclusively with mutant p53s in vivo but not with wild-type p53. These findings are the first to demonstrate the induction of endogenous MDR1 by mutant p53 and provide insight into the mechanism.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Genes, MDR/genetics , Genes, p53/genetics , Multidrug Resistance-Associated Proteins/metabolism , Mutation , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Up-Regulation , Base Sequence , Binding Sites , Blotting, Western , Caspases/metabolism , Cell Line , DNA/metabolism , Flow Cytometry , Genetic Vectors , Glutathione Transferase/metabolism , Humans , Luciferases/metabolism , Molecular Sequence Data , Promoter Regions, Genetic , Protein Binding , Protein Biosynthesis , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins c-ets , Ribonucleases/metabolism , Transcription, Genetic , Transcriptional Activation , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
19.
Hepatology ; 33(6): 1425-31, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11391531

ABSTRACT

The multidrug resistance protein Mdr1b in rats is up-regulated during liver regeneration after partial hepatectomy or after endotoxin treatment. We hypothesize that up-regulation of Mdr1b in these models is TNF-alpha-dependent. The mechanism of Mdr1b activation by TNF-alpha is unknown as TNF-alpha can signal through various pathways, including NF-kappaB and p53, transcription factors for which binding sites in the Mdr1b promoter have been identified. We aimed to elucidate the mechanism of up-regulation of Mdr1b by TNF-alpha. We selectively used constructs expressing dominant negative Fas-associated death domain protein (FADD), TNF receptor associated factor-2 (TRAF2) or IkappaB to inhibit pathways downstream of the TNF receptor. Further, the proteasome inhibitor MG-132 was used, which prevents the breakdown of IkappaB. We show a critical role for NF-kappaB in activation of Mdr1b gene expression both in primary rat hepatocytes and in rat hepatoma H-4-II-E cells. Because p53 is up-regulated by TNF-alpha in an NF-kappaB-dependent manner and the Mdr1b promoter contains a p53 binding site, we used liver cells expressing a dominant negative p53 to show that TNF-alpha up-regulation of Mdr1b is independent of functional p53. Using transient transfection assays, we show that Mdr1b up-regulation correlates with activation of the promoter. Mutation of the NF-kappaB site in the Mdr1b promoter prevents its induction by TNF-alpha. In conclusion our results show that activation of the rat Mdr1b gene by TNF-alpha is a result of NF-kappaB signaling and independent of p53.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , Gene Expression Regulation/physiology , Liver/physiology , NF-kappa B/physiology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/physiology , Tumor Suppressor Protein p53/physiology , Animals , Binding Sites/physiology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Hepatocytes/physiology , I-kappa B Proteins/pharmacology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Promoter Regions, Genetic/physiology , Rats , Transfection , Tumor Cells, Cultured , ATP-Binding Cassette Sub-Family B Member 4
20.
Oncogene ; 20(3): 303-13, 2001 Jan 18.
Article in English | MEDLINE | ID: mdl-11313959

ABSTRACT

The mdr1b gene is thought to be a "stress-responsive" gene, however it is unknown if this gene is regulated by p53 in the whole animal. Moreover, it is unknown if overexpression of mdr1b affects cell survival. The dependence of mdr1b upon p53 for upregulation was evaluated in p53 knockout mice. Wild-type (wt) or p53-/- mice were treated singly or in combination with gamma irradiation (IR) and/or the potent DNA damaging agent, diethylnitrosoamine (DEN). Both IR and DEN induced mdr1b in wild-type animals, but not in the p53-/- mice. IR also upregulated endogenous mdr1b in the H35 liver cell line, and the mdr1b promoter was activated by IR and activation correlated with p53 levels; moreover activation required an intact p53 binding site. Colony survival studies revealed that co-transfection of both mdr1b and p53 dramatically reduced colony numbers compared to cells transfected with either p53 or mdr1b alone and cells microinjected with both mdr1b and p53 had a more dramatic loss in viability compared to cells injected with either expression vector alone. Further studies using acridine orange and ethidium bromide to measure apoptosis revealed that mdr1b caused apoptosis and this was enhanced by p53, however the increased apoptosis required a functional p53 transactivation domain. These studies indicate that mdr1b is a downstream target of p53 in the whole animal and expression of mdr1b facilitates p53-mediated cell death.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Tumor Suppressor Protein p53/metabolism , ATP Binding Cassette Transporter, Subfamily B/radiation effects , Alkylating Agents/pharmacology , Animals , Base Sequence , Binding Sites , Cell Death/physiology , Cells, Cultured , Conserved Sequence , DNA Damage/drug effects , DNA Damage/genetics , Diethylnitrosamine/pharmacology , Gamma Rays , Liver/drug effects , Liver/metabolism , Liver/radiation effects , Male , Mice , Mice, Knockout , Molecular Sequence Data , Promoter Regions, Genetic , Radiation, Ionizing , Rats , Sequence Homology, Nucleic Acid , Tumor Suppressor Protein p53/genetics , Up-Regulation , Whole-Body Irradiation , ATP-Binding Cassette Sub-Family B Member 4
SELECTION OF CITATIONS
SEARCH DETAIL
...