Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Eur Respir J ; 34(5): 1093-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19357154

ABSTRACT

Mutations in bone morphogenetic protein receptor type 2 (BMPR2) cause familial pulmonary arterial hypertension (FPAH), but the penetrance is reduced and females are significantly overrepresented. In addition, gene expression data implicating the oestrogen-metabolising enzyme CYP1B1 suggests a detrimental role of oestrogens or oestrogen metabolites. We examined genetic and metabolic markers of altered oestrogen metabolism in subjects with a BMPR2 mutation. Genotypes for CYP1B1 Asn453Ser (N453S) were determined for 140 BMPR2 mutation carriers (86 females and 54 males). Nested from those subjects, a case-control study of urinary oestrogen metabolite levels (2-hydroxyoestrogen (2-OHE) and 16alpha-hydroxyoestrone (16alpha-OHE(1))) was conducted in females (five affected mutation carriers versus six unaffected mutation carriers). Among females, there was four-fold higher penetrance among subjects homozygous for the wild-type genotype (N/N) than those with N/S or S/S genotypes (p = 0.005). Consistent with this finding, the 2-OHE/16alpha-OHE(1) ratio was 2.3-fold lower in affected mutation carriers compared to unaffected mutation carriers (p = 0.006). Our findings suggest that variations in oestrogens and oestrogen metabolism modify FPAH risk. Further investigation of the role of oestrogens in this disease with profound sex bias may yield new insights and, perhaps, therapeutic interventions.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/genetics , Estrogens/metabolism , Hypertension/diagnosis , Hypertension/epidemiology , Pulmonary Artery/physiopathology , Adult , Aged , Bone Morphogenetic Protein Receptors, Type II/metabolism , Case-Control Studies , Cohort Studies , Female , Genotype , Heterozygote , Humans , Hypertension/metabolism , Male , Middle Aged , Mutation , Polymorphism, Genetic , Sex Factors
2.
Free Radic Biol Med ; 106: 62-68, 2017 05.
Article in English | MEDLINE | ID: mdl-28189846

ABSTRACT

The cellular production of free radicals or reactive oxygen species (ROS) can lead to protein, lipid or DNA modifications and tumor formation. The cellular lipids undergo structural changes through the actions of enzymes (e.g. cyclooxygenases) or free radicals to form a class of compounds called Isolevuglandins (IsoLGs). The recruitment and continued exposure of tissue to ROS and IsoLGs causes increased cell proliferation, mutagenesis, loss of normal cell function and angiogenesis. The elevated concentration of ROS in cancerous tissues suggests that these mediators play an important role in cancer development. We hypothesized that tumors with elevated ROS levels would similarly possess an increased concentration of IsoLGs when compared with normal tissue. Using D11, an ScFv recombinant antibody specific for IsoLGs, we utilized immunohistochemistry to visualize the presence of IsoLG in human tumors compared to normal adjacent tissue (NAT) to the same tumor. We found that IsoLG concentrations were elevated in human breast, colon, kidney, liver, lung, pancreatic and tongue tumor cells when compared to NAT and believe that IsoLGs can be used as a gauge indicative of lipid peroxidation in tumors.


Subject(s)
Carcinogenesis/genetics , Neoplasms/metabolism , Oxidative Stress/genetics , Prostaglandins E/metabolism , Reactive Oxygen Species/metabolism , Antibodies/pharmacology , Cell Line, Tumor , Cell Proliferation/genetics , Free Radicals/metabolism , Humans , Lipid Peroxidation/genetics , Neoplasms/genetics , Neoplasms/pathology , Phospholipids/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism
3.
J Natl Cancer Inst ; 87(6): 446-51, 1995 Mar 15.
Article in English | MEDLINE | ID: mdl-7861463

ABSTRACT

BACKGROUND: In breast cancer patients, about two thirds of the tumors are estrogen receptor (ER)-positive and one third are ER-negative. The molecular mechanisms leading to the ER-negative phenotype are poorly understood. Nearly all ER-negative and about 40% of ER-positive cancers are resistant to endocrine therapy. PURPOSE: In this study, we examined the entire coding region of the ER gene in ER-positive and ER-negative primary breast tumors to determine whether deletions/insertions or point mutations might account for the ER-negative phenotype. METHODS: We amplified exons 1 through 8 of the ER gene in 118 ER-positive and 70 ER-negative primary breast tumors and searched for mutations by single-strand conformation polymorphism analysis, denaturing gradient gel electrophoresis, and DNA sequencing. RESULTS: Both ER-negative and ER-positive tumors contained neutral polymorphisms in codons 10 [TCT-->TCC (Ser)], 87 [GCG-->GCC (Ala)], 243 [CGC-->CGT (Arg)], 325 [CCC-->CCG (Pro)], and 594 [ACA-->ACG (Thr)]. There was no correlation of any of the polymorphic alleles with the ER phenotype or other clinicopathologic parameters including tumor type, size, grade, or stage. However, the polymorphism in codon 325 showed a strong association with a family history of breast cancer (P = .0005). This association was observed both in premenopausal and postmenopausal patients. Despite extensive searching in exons 1 through 8, we found no deletions/insertions and only two missense mutations in codons 69 [AAC (Asn)-->AAG (Lys)] and 396 [ATG (Met)-->GTG (Val)] of the same ER-negative tumor. Thus, only 1% of the primary breast cancers had point mutations in the ER gene. CONCLUSIONS: In the majority of primary breast cancers, the ER-negative phenotype is not the result of mutations in the coding region of the ER gene, but is due to deficient ER expression at the transcriptional or post-transcriptional level. IMPLICATIONS: The correlation reported previously, as well as our current findings, suggest that further investigations are warranted to understand the possible linkage of the ER gene locus to hereditary breast cancer.


Subject(s)
Breast Neoplasms/genetics , Mutation/genetics , Receptors, Estrogen/genetics , Base Sequence , Breast Neoplasms/metabolism , Electrophoresis, Polyacrylamide Gel , Female , Gene Amplification , Gene Deletion , Humans , Molecular Sequence Data , Phenotype , Point Mutation , RNA, Messenger , RNA, Neoplasm , Transcription, Genetic
4.
Cancer Res ; 51(13): 3405-10, 1991 Jul 01.
Article in English | MEDLINE | ID: mdl-2054780

ABSTRACT

The estrogen receptor (ER) is a nuclear protein with a hormone- and a DNA-binding domain. We examined the DNA binding of ER in MCF-7 cells and 79 primary breast cancers by gel mobility shift assay using as a probe the estrogen response element (ERE). The mobility shift assay showed saturable, specific binding of ER to ERE in crude, high molar extracts containing greater than or equal to 4 mg/ml protein. Nonspecific binding was reduced by increasing concentrations of poly(deoxyinosidylate.deoxycytidylate) and shortening of the ERE probe from 35 to 15 base pairs. In the presence of Mg2+ the ER-ERE complex formation was hormone dependent at 22 degrees C but not at 37 degrees C. In the absence of Mg2+ estradiol was not necessary for ER-ERE complex formation. Correlation of the mobility shift assay with the hormone-binding (E2) assay showed agreement in 55 of the 79 tumors. Both assays were positive (E2 +/ERE+) in 35 cases and both were negative (E2-/ERE-) in 20 cases. In 11 tumors the hormone-binding assay was positive and the mobility shift assay negative (E2+/ERE-), suggesting an alteration of the DNA-binding domain. In 13 cancers the hormone-binding assay was negative and the mobility shift assay positive (E2-/ERE+) suggesting an alteration of the hormone-binding domain. By performing both hormone- and DNA-binding assays of ER and the hormone-binding assay of progesterone receptor (PR), we found the following subgroups of breast cancer: (a) E2+/ERE+/PR+, (b) E2+/ERE+/PR-, (c) E2+/ERE-/PR+, (d) E2+/ERE-/PR-, (e) E2-/ERE+/PR+, (f) E2-/ERE+/PR-, (g) E2-/ERE-/PR-. The simultaneous determination of 17 beta-estradiol and ERE binding may provide a better definition of the ER status of individual tumors and prove useful in refining endocrine therapy of patients with breast cancer.


Subject(s)
Breast Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Receptors, Estrogen/metabolism , Base Sequence , Breast Neoplasms/genetics , DNA, Neoplasm/metabolism , Estradiol/metabolism , Humans , In Vitro Techniques , Magnesium/pharmacology , Molecular Sequence Data , Oligonucleotides/chemistry , Oligonucleotides/metabolism , Regulatory Sequences, Nucleic Acid
5.
Cancer Res ; 54(7): 1641-4, 1994 Apr 01.
Article in English | MEDLINE | ID: mdl-8137273

ABSTRACT

Microsatellite instability (MSI) has been described in colorectal and other cancers. The purpose of this study was to determine the presence of MSI in breast cancer and to correlate its occurrence with clinicopathological parameters. For microsatellite markers we examined mono-, di-, tri-, and tetranucleotide repeats that, due to their polymorphic nature, may also be used to investigate loss of heterozygosity. In 20 paired breast cancer-peripheral blood DNA samples we identified four tumors (20%) with somatic MSI. All four tumors were stage I or II, grade 1 or 2, and estrogen receptor positive. To study MSI in relation to tumor progression we also examined paired DNA samples from two ipsilateral and three contralateral breast cancers, as well as two matched tumor-metastatic lymph node specimens. None of these seven cases showed MSI, but two of the contralateral tumors revealed allelic loss of polymorphic repeats. These data suggest that MSI is an early event in mammary tumorigenesis while loss of heterozygosity may occur at a later stage.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Chromosome Deletion , DNA, Neoplasm/genetics , DNA, Satellite/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Base Sequence , Biopsy , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/genetics , Carcinoma, Lobular/pathology , DNA Primers , DNA, Neoplasm/isolation & purification , DNA, Satellite/blood , DNA, Satellite/isolation & purification , Female , Humans , Lymphatic Metastasis , Middle Aged , Molecular Sequence Data , Neoplasm Invasiveness , Neoplasm Staging , Polymerase Chain Reaction , Repetitive Sequences, Nucleic Acid
6.
Cancer Res ; 58(22): 5038-41, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9823305

ABSTRACT

A key enzyme involved in the production of potentially carcinogenic estrogen metabolites and the activation of environmental carcinogens is cytochrome P450 1B1 (CYP1B1), the predominant member of the CYP1 family expressed in normal breast tissue and breast cancer. Because of the preeminent role of CYP1B1 in mammary estrogen/carcinogen metabolism, we examined the CYP1B1 gene to determine whether genetic differences could account for interindividual differences in breast cancer risk. We focused on exon 3, because it encodes the catalytically important heme binding domain of the enzyme, and discovered three polymorphisms of which two are associated with amino acid substitutions in codons 432 (Val-->Leu) and 453 (Asn-->Ser), designated as m1 and m2, respectively. Approximately 40% of Caucasian women have the m1 Val allele compared with nearly 70% of African-American women (P < 0.0001). The allele frequency also differs significantly in m2, with the rare Ser allele being present in 17.4% of Caucasians but only in 3.4% of African Americans (P < 0.0003). To determine whether the polymorphic CYP1B1 alleles hold implications as potential breast cancer risk factors, we compared the CYP1B1 genotypes in 164 Caucasian and 59 African-American breast cancer cases with those in age-, race-, and frequency-matched controls. Odds ratio calculations failed to show a significant association between any of the genotypes and breast cancer. Because CYP1B1 is known to be involved in mammary estrogen metabolism, we investigated whether the estrogen receptor status is influenced by the CYP1B1 genotypes. Caucasian patients with the m1 Val/Val genotype have a significantly higher percentage of estrogen receptor-positive (P = 0.02) and progesterone receptor-positive breast cancers (P = 0.003). There was no correlation with the m2 genotypes. These data suggest that the CYP1B1 polymorphisms in exon 3 are not associated with increased breast cancer risk but that the m1 polymorphism may be functionally important for steroid receptor expression in breast cancer of Caucasian patients.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cytochrome P-450 Enzyme System/genetics , Polymorphism, Genetic , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Black People/genetics , Breast Neoplasms/chemistry , Breast Neoplasms/ethnology , Case-Control Studies , Cytochrome P-450 CYP1B1 , Cytochrome P-450 Enzyme System/metabolism , Estradiol/metabolism , Exons/genetics , Female , Genotype , Humans , White People/genetics
7.
Cancer Res ; 52(1): 77-83, 1992 Jan 01.
Article in English | MEDLINE | ID: mdl-1345763

ABSTRACT

The presence of estrogen receptor (ER) is a well-known predictor of clinical outcome in human breast cancer. We examined the ER gene in 26 primary breast cancers (14 ER-positive, 12 ER-negative) to determine if alterations of the gene are associated with the ER-negative status. In tumor biopsies and peripheral blood DNA obtained from the same patients we analyzed the ER exon structure using polymerase chain reaction amplification, restriction endonuclease digestion, and agarose gel electrophoresis. All blood and tumor samples, regardless of ER status, showed a complete set of eight exons of normal sizes, ruling out deletions or rearrangements of the ER gene in excess of +/- 20 nucleotides. Previous reports indicate that the two-allele ER PvuII polymorphism could be associated with ER expression in breast cancer (Hill et al., Cancer Res., 49: 145-148, 1989) as well as with patient age at time of tumor diagnosis (Parl et al., Breast Cancer Res. Treat., 14: 57-64, 1989). We localized the PvuII polymorphism in intron 1, 0.4 kilobase upstream of exon 2. Sequence analysis showed the polymorphism to result from a point mutation (T----C) at the fifth position of the restriction site (CATCTG). We determined the PvuII restriction fragment-length polymorphism genotype in 257 primary breast cancers and 140 peripheral blood DNA samples obtained from women without breast cancer. The results indicate that the PvuII polymorphism is not associated with ER content or patient age at tumor diagnosis.


Subject(s)
Alleles , Breast Neoplasms/blood , Breast Neoplasms/genetics , Exons/genetics , Receptors, Estrogen/genetics , Adult , Base Sequence , Deoxyribonucleases, Type II Site-Specific , Female , Humans , Middle Aged , Molecular Sequence Data , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length
8.
Cancer Res ; 61(18): 6716-22, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11559542

ABSTRACT

The oxidative metabolism of 17beta-estradiol (E2) and estrone (E1) to catechol estrogens (2-OHE2, 4-OHE2, 2-OHE1, and 4-OHE1) and estrogen quinones has been postulated to be a factor in mammary carcinogenesis. Catechol-O-methyltransferase (COMT) catalyzes the methylation of catechol estrogens to methoxy estrogens, which simultaneously lowers the potential for DNA damage and increases the concentration of 2-methoxyestradiol (2-MeOE2), an antiproliferative metabolite. We expressed two recombinant forms of COMT, the wild-type (108Val) and a common variant (108Met), to determine whether their catalytic efficiencies differ with respect to catechol estrogen inactivation. The His-tagged proteins were purified by nickel-nitrilo-triacetic acid chromatography and analyzed by electrophoresis and Western immunoblot. COMT activity was assessed by determining the methylation of 2-OHE2, 4-OHE2, 2-OHE1, and 4-OHE1, using gas chromatography/mass spectrometry for quantitation of the respective methoxy products. In the case of 2-OHE2 and 2-OHE1, methylation occurred at 2-OH and 3-OH groups, resulting in the formation of 2-MeOE2 and 2-OH-3-MeOE2, and 2-MeOE1 and 2-OH-3-MeOE1, respectively. In contrast, in the case of 4-OHE2 and 4-OHE1, methylation occurred only at the 4-OH group, yielding 4-MeOE2 and 4-MeOE1, respectively. Individual and competition experiments revealed the following order of product formation: 4-MeOE2 > 4-MeOE1 >> 2-MeOE2 > 2-MeOE1 > 2-OH-3-MeOE1 > 2-OH-3-MeOE2. The variant isoform differed from wild-type COMT by being thermolabile, leading to 2-3-fold lower levels of product formation. MCF-7 breast cancer cells with the variant COMT 108Met/Met genotype also displayed 2-3-fold lower catalytic activity than ZR-75 breast cancer cells with the wild-type COMT 108Val/Val genotype. Thus, inherited alterations in COMT catalytic activity are associated with significant differences in catechol estrogen and methoxy estrogen levels and, thereby, may contribute to interindividual differences in breast cancer risk associated with estrogen-mediated carcinogenicity.


Subject(s)
Catechol O-Methyltransferase/metabolism , Estrogens, Catechol/metabolism , Alleles , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Catechol O-Methyltransferase/genetics , Cloning, Molecular , Enzyme Activation , Enzyme Stability , Enzyme-Linked Immunosorbent Assay , Gas Chromatography-Mass Spectrometry , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Methylation , Polymerase Chain Reaction , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tumor Cells, Cultured
9.
Cancer Res ; 44(1): 415-21, 1984 Jan.
Article in English | MEDLINE | ID: mdl-6690057

ABSTRACT

Two human breast cancer lines, MCF-7 and T47D cells, were investigated for the presence of estrogen receptor (ER) by biochemical and histochemical techniques. Using the dextran-coated charcoal technique and isoelectric focusing, MCF-7 cells were ER positive, and T47D cells were ER negative. Fluorescein conjugates to 17 beta-estradiol by the sixth carbon (17 beta-estradiol-6-carboxymethyloxime:bovine serum albumin: fluorescein isothiocyanate and 17 beta-estradiol-6-iminooxyacetylfluoresceinamine) and by the 17th carbon [N-fluoresceino-N'-[17 beta-(estradiol hemisuccinamide)ethyl]thiourea, 17-FE] were prepared for cytochemical evaluation of the ER status of the two cell lines. The binding affinity of the estradiol conjugates for ER varied, the 17-FE conjugate having the highest affinity of 0.08 relative to 17 beta-estradiol. Following incubation with 10 nM 17-FE, both MCF-7 and T47D cells displayed cytoplasmic and nuclear fluorescent staining. Isoelectric focusing of MCF-7 cytosol incubated in the presence of 10 nM 17-FE revealed binding of the fluorescein conjugate to a protein species which did not bind 17 beta-[3H]-estradiol. Isoelectric focusing of T47D cytosol revealed binding of 17-FE to two protein components, neither one of which showed specific binding of 17 beta-[3H]estradiol. The results suggest different protein binding species for fluoresceinated estradiol conjugates and [3H]estradiol and help to explain reported differences in histochemical and biochemical ER analyses.


Subject(s)
Breast Neoplasms/metabolism , Receptors, Estrogen/metabolism , Breast Neoplasms/pathology , Cell Line , Estradiol/metabolism , Female , Fluorescent Dyes , Humans , Isoelectric Focusing , Receptors, Estrogen/isolation & purification , Tritium
10.
Cancer Res ; 60(13): 3440-4, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10910054

ABSTRACT

Activation of 17beta-estradiol (E2) through the formation of catechol estrogen metabolites, 2-OH-E2 and 4-OH-E2, and the C-16alpha hydroxylation product, 16alpha-OH-E2, has been postulated to be a factor in mammary carcinogenesis. Cytochrome P450 1B1 (CYP1B1) exceeds other P450 enzymes in both estrogen hydroxylation activity and expression level in breast tissue. To determine whether inherited variants of CYP1B1 differ from wild-type CYP1B1 in estrogen hydroxylase activity, we expressed recombinant wild-type and five polymorphic variants of CYP1B1: variant 1 (codon 48Arg-->Gly), variant 2 (codon 119Ala-->Ser), variant 3 (codon 432Val-->Leu), variant 4 (codon453Asn-->Ser), variant 5 (48Gly, 119Ser, 432Leu, 453Ser). The His-tagged proteins were purified by nickel-nitrilotriacetic acid (Ni-NTA) chromatography and analyzed by electrophoresis and spectrophotometry. We performed assays of E2 hydroxylation activity and quantitated production of 2-OH-E2, 4-OH-E2, and 16alpha-OH-E2 by gas chromatography/mass spectrometry. Wild-type CYP1B1 formed 4-OH-E2 as main product (Km, 40+/-8 microM; k(cat) 4.4+/-0.4, min(-1); k(cat)/Km, 110 mM(-1) min(-1)), followed by 2-OH-E2 (Km, 34+/-4 microM; k(cat), 1.9+/-0.1 min(-1); k(cat)/Km, 55 mM(-1)min(-1)) and 16alpha-OH-E2 (Km, 39+/-5.7 microM; k(cat), 0.30+/-0.02 min(-1); k(cat)/Km, 7.6 mM(-1)min(-1)). The CYP1B1 variants also formed 4-OH-E2 as the main product but displayed 2.4- to 3.4-fold higher catalytic efficiencies k(cat)/Km than the wild-type enzyme, ranging from 270 mM(-1)min(-1) for variant 4, to 370 mM(-1)min(-1) for variant 2. The variant enzymes also exceeded wild-type CYP1B1 with respect to 2- and 16alpha-hydroxylation activity. Thus, inherited alterations in CYP1B1 estrogen hydroxylation activity may be associated with significant changes in estrogen metabolism and, thereby, may possibly explain interindividual differences in breast cancer risk associated with estrogen-mediated carcinogenicity.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Estradiol/metabolism , Polymorphism, Genetic , Amino Acid Substitution , Cloning, Molecular , Codon/genetics , Cytochrome P-450 CYP1B1 , Cytochrome P-450 Enzyme System/chemistry , Escherichia coli , Estradiol/analogs & derivatives , Gas Chromatography-Mass Spectrometry , Genetic Variation , Humans , Hydroxylation , Kinetics , Mutagenesis, Site-Directed , Pharmacogenetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Steroid 16-alpha-Hydroxylase
11.
Cancer Res ; 58(1): 65-70, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9426059

ABSTRACT

Genetically based differences in carcinogen metabolism have been related to polymorphisms of the cytochrome P450IA1 gene (CYPIA1) and the null genotypes of glutathione S-transferase classes mu and theta (GSTM1 and GSTT1). By PCR we examined the genotypes of CYPIA1, GSTM1, and GSTT1 in relation to breast cancer risk in Caucasian and African-American women. The study included 164 Caucasian and 59 African-American women with primary invasive breast cancer and age-matched female controls. Enzyme polymorphisms included in this study were the null deletions of GSTM1 and GSTT1 and the m1 (MspI), m2 (codon 462: isoleucine-->valine), m3 (MspI-AA), and m4 (codon 461: threonine-->asparagine) polymorphisms of CYPIA1. Contrary to previous reports by other investigators, none of the enzyme genotypes, individually or combined, appear to associate with an increased risk for breast cancer in Caucasian or African-American women. We also report that the recently described m4 allele occurs at a lower frequency in African-Americans than Caucasians and is not linked with breast cancer in either race. Thus, it is unlikely that polymorphisms of GSTM1, GSTT1, or CYPIA1 represent susceptibility factors for breast cancer in Caucasians or African-Americans.


Subject(s)
Black People/genetics , Breast Neoplasms/enzymology , Cytochrome P-450 CYP1A1/genetics , Glutathione Transferase/genetics , Isoenzymes/genetics , Polymorphism, Genetic , White People/genetics , Alleles , Breast Neoplasms/genetics , Female , Gene Frequency , Genotype , Humans
12.
Cancer Res ; 58(12): 2515-9, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9635570

ABSTRACT

Southern analysis has shown that DNA from 25% of primary estrogen receptor (ER) alpha-negative breast tumors displays aberrant methylation at one site within the ER gene CpG island. To examine more sites and increase sensitivity, we developed a methylation-specific PCR assay to map methylation of the entire ER CpG island. The island was unmethylated in normal breast tissue and ER-positive breast cancer cell lines, but extensively methylated in all ER-negative cell lines and breast tumors examined. In addition, some of the ER-positive/progesterone receptor-negative and ER-positive/progesterone receptor-positive tumors (about 70% and 35%, respectively) displayed methylation of the ER CpG island, suggesting that this heterogeneity within tumor cell populations could potentially shed light on the etiology of ER-negative recurrent tumors arising from ER-positive tumors.


Subject(s)
Breast Neoplasms/genetics , CpG Islands/genetics , DNA Methylation , DNA, Neoplasm/genetics , Receptors, Estrogen/genetics , Adult , Aged , Aged, 80 and over , DNA, Neoplasm/metabolism , Female , Genetic Markers/genetics , Humans , Middle Aged , Polymerase Chain Reaction/methods , Tumor Cells, Cultured
13.
Cancer Res ; 54(10): 2552-5, 1994 May 15.
Article in English | MEDLINE | ID: mdl-8168078

ABSTRACT

Breast cancer is the most common malignancy in women and hormone resistance is a challenging problem in its treatment. Loss of estrogen receptor expression is an important means of hormone resistance, but the mechanisms involved are poorly understood. We now demonstrate a potential role for abnormal DNA methylation in transcriptional inactivation of the estrogen receptor gene. Estrogen receptor-negative human breast cancer cells growing in culture lack estrogen receptor mRNA, have a higher capacity to methylate DNA, and display extensive methylation of the CpG island in the 5' promoter region of the estrogen receptor gene, which would correlate with silencing of expression. These results suggest that abnormal methylation could account for transcriptional inactivation of the estrogen receptor gene and subsequent hormone resistance in some human breast cancers.


Subject(s)
Breast Neoplasms/genetics , DNA, Neoplasm/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Breast Neoplasms/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Female , Humans , Methylation , RNA, Messenger/metabolism , Tumor Cells, Cultured
14.
Cancer Res ; 60(16): 4346-8, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10969774

ABSTRACT

Loss of expression for both the estrogen receptor-alpha and E-cadherin genes has been linked to disease progression in human ductal breast carcinomas and has been associated with aberrant 5' CpG island methylation. To assess when, during malignant progression, such methylation begins and whether such methylation increases with advancing disease, we have surveyed 111 ductal carcinomas of the breast for aberrant methylation of the estrogen receptor-alpha and E-cadherin 5' CpG islands. Hypermethylation of either CpG island was evident prior to invasion in approximately 30% of ductal carcinoma in situ lesions and increased significantly to nearly 60% in metastatic lesions. Coincident methylation of both CpG islands also increased significantly from approximately 20% in ductal carcinoma in situ to nearly 50% in metastatic lesions. Furthermore, in all cases, the pattern of methylation displayed substantial heterogeneity, reflecting the well-established, heterogeneous loss of expression for these genes in ductal carcinomas of the breast.


Subject(s)
Breast Neoplasms/genetics , Cadherins/genetics , Carcinoma, Ductal, Breast/genetics , CpG Islands/physiology , DNA Methylation , Receptors, Estrogen/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma in Situ/genetics , Carcinoma in Situ/metabolism , Carcinoma in Situ/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic/physiology , Gene Silencing/physiology , Humans , Neoplasm Metastasis , Tumor Cells, Cultured
15.
Clin Cancer Res ; 2(5): 805-10, 1996 May.
Article in English | MEDLINE | ID: mdl-9816234

ABSTRACT

Hormonal factors have a profound influence on the development, treatment, and outcome of breast cancer. The absence of steroid hormone receptors is highly correlated with resistance to antihormonal treatments. Work in cultured human breast cancer cell lines has shown that the absence of estrogen receptor (ER) gene expression in ER- cells is associated with extensive methylation of the ER gene 5' CpG island, and treatment with agents that demethylate the ER gene CpG island results in the production of functional ER protein. The current study shows that CpG islands in the 5' region of the ER and progesterone receptor (PR) genes are methylated in a significant fraction of primary human breast cancer tissues. The ER CpG island is methylated at the methylation-sensitive NotI restriction site in 9 of 39 (25%) of primary ER- breast cancers but remains unmethylated in 53 ER+ breast cancers and 9 normal breast specimens. Three methylation-sensitive restriction sites in the PR gene CpG island are not methylated in normal breast specimens and PR+ human breast cancers but are hypermethylated in 40% of PR- human breast tumors. These data demonstrate that methylation of the ER and PR gene CpG islands is associated with the lack of ER and PR gene expression in a significant fraction of human breast cancers.


Subject(s)
Breast Neoplasms/genetics , CpG Islands , DNA Methylation , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Female , Gene Expression , Humans , Tumor Cells, Cultured
16.
Mol Endocrinol ; 11(8): 1009-19, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9212049

ABSTRACT

The estrogen receptor (ER) belongs to a family of ligand-inducible nuclear receptors that exert their effects by binding to cis-acting DNA elements in the regulatory region of target genes. The detailed mechanisms by which ER interacts with the estrogen response element (ERE) and affects transcription still remain to be elucidated. To study the ER-ERE interaction and transcription initiation, we employed purified recombinant ER expressed in both the baculovirus-Sf9 and his-tagged bacterial systems. The effect of high-mobility group (HMG) protein HMG-1 and purified recombinant TATA-binding protein-associated factor TAF(II)30 on ER-ERE binding and transcription initiation were assessed by electrophoretic mobility shift assay and in vitro transcription from an ERE-containing template (pERE2LovTATA), respectively. We find that purified, recombinant ER fails to bind to ERE in spite of high ligand-binding activity and electrophoretic and immunological properties identical to ER in MCF-7 breast cancer cells. HMG-1 interacts with ER and promotes ER-ERE binding in a concentration- and time-dependent manner. The effectiveness of HMG-1 to stimulate ER-ERE binding in the electrophoretic mobility shift assay depends on the sequence flanking the ERE consensus as well as the position of the latter in the oligonucleotide. We find that TAF(II)30 has no effect on ER-ERE binding either alone or in combination with ER and HMG-1. Although HMG-1 promotes ER-ERE binding, it fails to stimulate transcription initiation either in the presence or absence of hormone. In contrast, TAF(II)30, while not affecting ER-ERE binding, stimulates transcription initiation 20-fold in the presence of HMG-1. These results indicate that HMG-1 and TAF(II)30 act in sequence, the former acting to promote ER-ERE binding followed by the latter to stimulate transcription initiation.


Subject(s)
Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Receptors, Estrogen/metabolism , TATA-Binding Protein Associated Factors , Transcription Factor TFIID , Transcription Factors/metabolism , Transcription, Genetic , Animals , Carrier Proteins/genetics , DNA-Binding Proteins/genetics , Estrogens/metabolism , HMGB1 Protein , High Mobility Group Proteins/genetics , Humans , Oligonucleotides/chemistry , Oligonucleotides/genetics , Oligonucleotides/metabolism , Receptors, Estrogen/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription Factors/genetics
17.
Cancer Epidemiol Biomarkers Prev ; 10(12): 1239-48, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11751440

ABSTRACT

Using the International Project on Genetic Susceptibility to Environmental Carcinogens (GSEC) database containing information on over 15,000 control (noncancer) subjects, the allele and genotype frequencies for many of the more commonly studied metabolic genes (CYP1A1, CYP2E1, CYP2D6, GSTM1, GSTT1, NAT2, GSTP, and EPHX) in the human population were determined. Major and significant differences in these frequencies were observed between Caucasians (n = 12,525), Asians (n = 2,136), and Africans and African Americans (n = 996), and some, but much less, heterogeneity was observed within Caucasian populations from different countries. No differences in allele frequencies were seen by age, sex, or type of controls (hospital patients versus population controls). No examples of linkage disequilibrium between the different loci were detected based on comparison of observed and expected frequencies for combinations of specific alleles.


Subject(s)
Black People/genetics , Gene Frequency , Genetic Predisposition to Disease , Neoplasms/genetics , Polymorphism, Genetic , White People/genetics , Cytochrome P-450 Enzyme System/genetics , Databases, Factual , Genetic Linkage , Humans
18.
J Histochem Cytochem ; 31(5): 608-12, 1983 May.
Article in English | MEDLINE | ID: mdl-6341455

ABSTRACT

Cerebrospinal fluid (CSF) is generally considered to be derived from plasma through a combined process of ultrafiltration and secretion by the choroid plexus. However, the mechanisms ultimately responsible for achieving the final protein composition of CSF are uncertain. Some proteins, in particular prealbumin, are present in quantities not easily explained by usual physicochemical considerations. To investigate the possibility of de novo synthesis by choroid epithelium, we have examined human choroid plexus an ependyma for the presence of prealbumin. Using the peroxidase-antiperoxidase method at the light and electron microscopic level, as well as immunofluorescence, we localized prealbumin in choroid epithelial cytoplasm on the endoplasmic reticulum and in association with the Golgi apparatus. Ependymal cells and stroma did not reveal immunocytochemical labeling. These findings indicate that the choroid plexus epithelium contributes to the final CSF composition by de novo protein synthesis.


Subject(s)
Choroid Plexus/analysis , Prealbumin/analysis , Serum Albumin/analysis , Animals , Choroid Plexus/anatomy & histology , Choroid Plexus/ultrastructure , Epithelium/analysis , Epithelium/anatomy & histology , Histocytochemistry , Humans , Immunoenzyme Techniques , Microscopy, Electron , Microscopy, Fluorescence , Prealbumin/biosynthesis , Prealbumin/cerebrospinal fluid , Rabbits
19.
Mol Cell Endocrinol ; 52(3): 235-42, 1987 Aug.
Article in English | MEDLINE | ID: mdl-3653505

ABSTRACT

We transcribed a cDNA clone of the human estrogen receptor (ER) with T7 RNA polymerase. The 32P-cRNA transcript complementary to ER mRNA was hybridized to poly(A)+ RNA from human uterus and revealed a single band of approximately 4.2 kilobases. No hybridization was seen with the cRNA probe of the opposite orientation. Hybridization of total RNA from calf and rat uterus yielded a single band at approximately 3.8 kilobases for both species. Total RNA from rat spleen did not hybridize. A 35S-labeled cRNA probe was prepared for in situ hybridization of ER mRNA in human uterus and spleen. Autoradiographic signal was present over endometrial epithelium, stromal cells of the lamina propria, and smooth muscle cells of the myometrium but was absent from sections of spleen. The ER mRNA hybridization label was located over cytoplasm and nuclei of uterine target cells.


Subject(s)
RNA, Messenger/metabolism , Receptors, Estrogen/metabolism , Uterus/metabolism , DNA/genetics , Female , Humans , Nucleic Acid Hybridization , RNA, Messenger/genetics , Receptors, Estrogen/genetics , Tissue Distribution
20.
Hum Pathol ; 19(8): 960-6, 1988 Aug.
Article in English | MEDLINE | ID: mdl-3402985

ABSTRACT

We examined the estrogen receptor (ER) content of 124 primary breast cancers by hormone binding and immunohistochemical (ER-ICA) assays. Both assays were in agreement in 110 tumors (89%; P less than .0001); 68 tumors were positive and 42 were negative. In 14 cases (11%), the assays yielded discordant results. Three tumors showed hormone binding in the absence of immunohistochemically detectable ER; the false positive hormone binding resulted from the presence of normal epithelium adjacent to ER-ICA negative malignant cells. Eleven tumors failed to show hormone binding but were ER-ICA positive. Four of these were from premenopausal patients whose circulating endogenous estrogen may occupy the receptor, giving rise to false negative hormone binding assays. In four cases, the discrepancy of negative hormone binding assay and positive ER-ICA assay was attributed to scant tumor cells in the tissue sample. The remaining three discrepancies could not be resolved with certainty, but possibly resulted from alteration of the hormone binding site with preservation of the immunoreactive epitope on the ER molecule. These results indicate that the ER-ICA assay is more accurate than the hormone binding assay in identifying the presence of ER in cancer cells. The heterogeneous immunostaining of ER in tumor sections, which may reflect mosaicism of tumor cells, rate of cell proliferation, or phase of cell cycle, remains unexplained.


Subject(s)
Breast Neoplasms/analysis , Receptors, Estrogen/analysis , Biological Assay , Breast Neoplasms/pathology , Estradiol/metabolism , Female , Humans , Immunohistochemistry , Progesterone/metabolism , Receptors, Estrogen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL