Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Clin Pharmacol Ther ; 87(5): 572-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20375999

ABSTRACT

Well-characterized genes that affect warfarin metabolism (cytochrome P450 (CYP) 2C9) and sensitivity (vitamin K epoxide reductase complex 1 (VKORC1)) explain one-third of the variability in therapeutic dose before the international normalized ratio (INR) is measured. To determine genotypic relevance after INR becomes available, we derived clinical and pharmacogenetic refinement algorithms on the basis of INR values (on day 4 or 5 of therapy), clinical factors, and genotype. After adjusting for INR, CYP2C9 and VKORC1 genotypes remained significant predictors (P < 0.001) of warfarin dose. The clinical algorithm had an R(2) of 48% (median absolute error (MAE): 7.0 mg/week) and the pharmacogenetic algorithm had an R(2) of 63% (MAE: 5.5 mg/week) in the derivation set (N = 969). In independent validation sets, the R(2) was 26-43% with the clinical algorithm and 42-58% when genotype was added (P = 0.002). After several days of therapy, a pharmacogenetic algorithm estimates the therapeutic warfarin dose more accurately than one using clinical factors and INR response alone.


Subject(s)
Genetic Variation/genetics , International Normalized Ratio/standards , Systems Integration , Warfarin/administration & dosage , Aged , Aryl Hydrocarbon Hydroxylases/genetics , Cohort Studies , Cytochrome P-450 CYP2C9 , Dose-Response Relationship, Drug , Female , Genotype , Humans , International Normalized Ratio/methods , Male , Middle Aged , Mixed Function Oxygenases/genetics , Pharmacogenetics/methods , Vitamin K Epoxide Reductases , Warfarin/pharmacokinetics
2.
Med Oncol ; 25(1): 40-9, 2008.
Article in English | MEDLINE | ID: mdl-18188713

ABSTRACT

GDEP (gene differentially expressed in prostate cancer aka. PCAN1), a newly discovered gene with remarkable tissue specificity, is a promising candidate for regulatory analysis because it exhibits a high level of expression that is limited to two tissues, the retina and the prostate. As these two tissues have different origins and disparate functions it is likely that the regulatory mechanisms responsible for expression are not shared in their entirety. In addition, both the retina and prostate are prime targets for gene therapy. To date there have been no functional studies of the GDEP promoter. Therefore to understand tissue-specific expression of GDEP we constructed promoter expression constructs. To further characterize functional regulatory regions within the GDEP gene, we investigated potential regulatory components for tissue-specific expression in the 40 kb intron of this gene. We have identified a 1.5 kb prostate-specific promoter from the proximal region of the GDEP gene. A smaller 0.5 kb promoter exhibited minimal activity in the retinoblastoma cell line Y79, but not in the prostate cells tested. In addition we have investigated three enhancer elements located in the 40 kb intron of the GDEP gene. We identified two enhancer elements that increase reporter gene expression in prostate cell line LNCaP and one additional enhancer element that increases expression in the Y79 cell line approximately 8-fold making it a strong retinal-specific enhancer.


Subject(s)
Enhancer Elements, Genetic , Neoplasm Proteins/genetics , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Retinoblastoma/genetics , Cell Line, Tumor , Humans , Male , Organ Specificity
3.
Med Oncol ; 25(3): 287-93, 2008.
Article in English | MEDLINE | ID: mdl-18163187

ABSTRACT

Homeobox (HOX) genes are crucial regulators of cell growth and differentiation. These genes initiate and control gene expression cascades that drive development. More recently, the absent or aberrant expression of HOX genes has been implicated in cancer development. Despite the observance of these expression changes, the regulation of the HOX genes in adult tissues and how these genes become deregulated in cancerous tissues still needs much investigation. We characterized the promoter region of the HOXB13 gene. A 3 kb region upstream of the HOXB13 gene, which included the 5'UTR, increased reporter gene expression in LNCaP cells by approximately 99 fold over the promoterless control construct. A highly conserved 179 base pair fragment containing only the 5'UTR of the HOXB13 gene constituted a minimal promoter in the LNCaP cell line. Strong promoter activity was seen in the presence or absence of testosterone, although testosterone exposure did decrease expression in LNCaP cells by 50%. In an androgen insensitive cell line Du145, no sensitivity to testosterone was detected and a consistent low basal level of expression was observed. Since HOXB13 expression is highly tissue specific, we investigated the ability of the promoter to drive expression in tissues other than prostate. We observed highest expression in LNCaP cells with low levels of expression in lung, retinoblastoma, and colon cancer cells and higher expression in MCF7 breast cancer cells.


Subject(s)
Genes, Homeobox , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , 5' Untranslated Regions , Base Sequence , Cell Line, Tumor , Conserved Sequence , Databases, Nucleic Acid , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Luciferases/metabolism , Male , Molecular Sequence Data , Promoter Regions, Genetic/drug effects , Prostatic Neoplasms/metabolism , Sequence Homology, Nucleic Acid , Testosterone/pharmacology
4.
Clin Genet ; 68(5): 448-53, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16207213

ABSTRACT

An analysis of PAX1 in the development of vertebral malformations. Due to the sporadic occurrence of congenital vertebral malformations, traditional linkage approaches to identify genes associated with human vertebral development are not possible. We therefore identified PAX1 as a candidate gene in vertebral malformations and congenital scoliosis due to its mutation in the undulated mouse. We performed DNA sequence analysis of the PAX1 gene in a series of 48 patients with congenital vertebral malformations, collectively spanning the entire vertebral column length. DNA sequence coding variants were identified in the heterozygous state in exon 4 in two male patients with thoracic vertebral malformations. One patient had T9 hypoplasia, T12 hemivertebrae and absent T10 pedicle, incomplete fusion of T7 posterior elements, ventricular septal defect, and polydactyly. This patient had a CCC (Pro)-->CTC (Leu) change at amino acid 410. This variant was not observed in 180 chromosomes tested in the National Institute of Environmental Health Sciences (NIEHS) single nucleotide polymorphism (SNP) database and occurred at a frequency of 0.3% in a diversity panel of 1066 human samples. The second patient had a T11 wedge vertebra and a missense mutation at amino acid 413 corresponding to CCA (Pro)-->CTA (Leu). This particular variant has been reported to occur in one of 164 chromosomes in the NIEHS SNP database and was found to occur with a similar frequency of 0.8% in a diversity panel of 1066 human samples. Although each patient's mother was clinically asymptomatic and heterozygous for the respective variant allele, the possibility that these sequence variants have clinical significance is not excluded.


Subject(s)
Mutation , Paired Box Transcription Factors/genetics , Spine/abnormalities , Base Sequence , DNA Mutational Analysis , Humans , Morphogenesis/genetics , Phenotype , Scoliosis/genetics , Spinal Diseases/genetics
5.
Med Oncol ; 21(2): 145-53, 2004.
Article in English | MEDLINE | ID: mdl-15299187

ABSTRACT

Prostate cancer is the most frequently diagnosed neoplasia in men and one of the leading causes of cancer-related deaths in men over 60. In an effort to understand the molecular events leading to prostate cancer, we have identified PCAN1 (prostate cancer gene 1) (also known as GDEP), a gene that is highly expressed in prostate epithelial tissue and frequently mutated in prostate tumors. Here we demonstrate its expression in neural retina, and retinoblastoma cell culture but not retinal pigment epithelial cell culture. We further characterize PCAN1 expression in the prostate cell lines RWPE1, RWPE2, and LnCAP FGC. We demonstrate an increase in expression when the cells are grown in the presence of Matrigel, an artificial extracellular basement membrane. Expression was time dependent, with expression observed on d 6 and little or no expression on d 12. Testosterone was not found to increase PCAN1 expression in this culture system. In addition, normal prostate epithelial cells co-cultured with normal prostate stromal cells did not exhibit PCAN1 expression at any time. To definitively locate the transcription initiation sites, we performed restriction-ligase-mediated 5' RACE, to selectively amplify only mRNA with a 5' cap. An initial characterization of the sequence upstream of the initiation sites determined six possible binding sites for the prostate specific regulatory protein NKX3.1 and four potential binding sites for the PPAR/RXR heterodimer that is involved in the control of cell differentiation and apoptosis.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Retina/physiology , Retinoblastoma/genetics , Retinoblastoma/pathology , Apoptosis , Cell Differentiation , Epithelial Cells , Humans , Male , Middle Aged , Promoter Regions, Genetic , Retina/cytology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
6.
Cytokines Cell Mol Ther ; 7(1): 25-30, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12171248

ABSTRACT

Transforming growth factor-beta (TGF-beta) is a multifunctional growth and differentiation factor that affects almost all cells. Although equipotent in many cases, the three isoforms of TGF-beta (-beta1, -beta2, -beta3) have several important isoform specific activities. For example, TGF-beta2 binds with higher affinity to a 60 kDa cell-surface glycosyl phosphatidylinositol (GPI)-linked protein, expressed on vascular endothelial cells. We used chimeric TGF-beta proteins, in which selected regions of TGF-beta1 had been exchanged for the corresponding region of TGF-beta2, to demonstrate that amino acids 67 and 68 regulate binding of TGF-beta to this protein. Exchange of amino acids 67 and 68 of TGF-beta1 into TGF-beta2 resulted in a protein similar in affinity to TGF-beta1 for binding to the GPI-linked protein. In contrast, exchange of only amino acid 67 of TGF-beta1 into TGF-beta2, or exchange of only amino acid 68 of TGF-beta1 into TGF-beta2, resulted in a protein with affinity similar to that of TGF-beta2. This suggests that the coordinated change of Gln and His of TGF-beta1 to Thr and Ile at positions 67 and 68 alters the specificity of TGF-beta. Amino acids 67 and 68 are part of a surface-exposed alpha-helix that forms a projection away from the center of the TGF-beta molecule and is accessible for receptor binding.


Subject(s)
Endothelium, Vascular/cytology , Transforming Growth Factor beta/chemistry , Amino Acid Sequence , Animals , CHO Cells , Cattle , Cell Membrane/metabolism , Cells, Cultured , Cricetinae , Cross-Linking Reagents/pharmacology , Cysteine/chemistry , Endothelium, Vascular/metabolism , Isoleucine/chemistry , Ligands , Models, Molecular , Molecular Sequence Data , Mutation , Plasmids/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Swine , Threonine/chemistry , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Transforming Growth Factor beta2
7.
Cytokines Cell Mol Ther ; 7(1): 31-6, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12171249

ABSTRACT

Transforming growth factor-beta (TGF-beta) proteins are multifunctional regulators of cell growth and differentiation. The three isoforms, TGF-beta1, -beta2, -beta3 share approximately 70% identical amino acid sequence and are coded by three distinct genes. Growth and differentiation functions in which the isoforms have differential activity include: inhibition of colorectal cancer cell growth, migration of aortic endothelial cells, survival of ciliary ganglionic neurons, and binding to cell surface receptors. A previous paper reported that TGF-beta1 and TGF-beta2 had bimodal dose-dependent stimulatory and inhibitory effects on granulocyte-macrophage colony-stimulating factor induced Day 7 granulocyte-macrophage colony-forming units. The effects of TGF-beta3 were only inhibitory. At low concentrations, TGF-beta1 and -beta2 stimulated growth, whereas at higher concentrations both isoforms inhibited growth. We now report that TGF-beta1, TGF-beta2, and TGF-beta3 are similar to each other at low concentrations; at higher concentrations TGF-beta1 and TGF-beta3 inhibit growth, but TGF-beta2 stimulates growth. Our results are consistent with the known affinities of the TGF-beta isoforms with the Type II TGF-beta signaling receptor, which has greater affinity for TGF-beta1 and TGF-beta3 than TGF-beta2.


Subject(s)
Colony-Stimulating Factors/physiology , Myelopoiesis , Transforming Growth Factor beta/chemistry , Amino Acid Sequence , Cell Cycle , Cell Differentiation , Cell Division , Cell Movement , Colony-Stimulating Factors/metabolism , Cytokines/metabolism , Dimerization , Dose-Response Relationship, Drug , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Protein Isoforms , Protein Structure, Secondary , RNA/metabolism , Sequence Homology, Amino Acid , Time Factors , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Transforming Growth Factor beta2 , Transforming Growth Factor beta3
8.
Med Oncol ; 18(3): 179-87, 2001.
Article in English | MEDLINE | ID: mdl-11917942

ABSTRACT

Although prostate cancer is the second leading cause of cancer death for men in the United States, the genetics of tumor development are poorly understood. Several expressed sequence tagged genes (ESTs) that are expressed predominantly in the prostate have recently been identified, although their role in the development and maintenance of the prostate is unknown. Here, we demonstrate that the gene identified as UNIGENE cluster Hs. 104215, which codes for a message found predominantly in the prostate, may be important in tumor development. We name this gene PCan1 for Prostate Cancer gene 1. Northern blot experiments were performed using RNA isolated from tumor-derived cell lines and human prostate to determine the expression pattern of the gene. DNA sequencing was used to identify mutations that occurred in tumor tissue. By Northern blot analysis, this gene product was not detectable in LNCaP, DU 145, or PC-3 prostate cancer cell lines, although it was readily observed in RNA isolated from total prostate and from dissected central and peripheral regions of prostate. Sequence analysis of genomic DNA from LNCaP, DU 145, or PC-3 cells demonstrated a G/A polymorphism at position 193. Analysis of matched tumor-derived DNA and blood-derived DNA samples from 11 of 13 patients who had undergone a radical prostatectomy and who were homozygous for A in blood-derived DNA demonstrated mutation of position 193 in matched tumor samples resulting in G/A polymorphism. Sixteen additional patient samples were G/A polymorphic in both blood-derived DNA and tumor-derived DNA and two samples were GG in both blood-derived and tumor-derived DNA. Our results suggest that this gene may be a hot spot for mutation in prostate cancer, especially because our radiation hybrid mapping located this gene within a region identified in linkage mapping studies of affected families with prostate cancer. Loss of heterozygosity in prostate tumors has also been reported at the location of PCan1. Further studies to determine the functional role of this candidate tumor suppressor gene are warranted.


Subject(s)
DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Polymorphism, Genetic , Prostatic Neoplasms/genetics , Sequence Tagged Sites , Base Sequence , Blotting, Northern , Cell Transformation, Neoplastic , DNA Mutational Analysis , Humans , Loss of Heterozygosity , Male , Molecular Sequence Data , Prostatic Neoplasms/pathology , RNA/genetics , Tumor Cells, Cultured
9.
Eur J Hum Genet ; 8(11): 875-83, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11093278

ABSTRACT

Gilles de la Tourette syndrome (GTS) is a common, heritable neurological disorder manifested by chronic motor and vocal tics with childhood onset. Previous extensive linkage analysis failed to identify a GTS gene based on an autosomal dominant pattern of inheritance. Recently, a family was reported with a balanced chromosomal translocation t(1;8)(q21.1;q22.1) in family members with GTS or tics. Chromosome 8q22.1 was previously implicated in GTS by both association and linkage results. We therefore cloned and sequenced both translocation breakpoints from this family. The CBFA2T1 gene was identified 11 kb distal to the 8q22.1 breakpoint. Sequencing of CBFA2TI exons within 37 unrelated GTS patients failed to identify any mutations. However, it is possible that the translocation altered the expression of this gene or another nearby gene. Examination of the breakpoint sequences revealed a duplication of six nucleotides from chromosome 8 but no change in the chromosome 1 sequence. The sequences immediately flanking the breakpoints on the two chromosomes were modestly similar, but the breakpoints did not occur within known interspersed repeats. Our results add to our knowledge of the genetics of GTS and the mechanisms of balanced chromosomal translocations.


Subject(s)
Chromosome Breakage/genetics , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 8/genetics , Tourette Syndrome/genetics , Translocation, Genetic , Bacteriophages , Base Sequence , Chromosomes, Bacterial , Contig Mapping , Cosmids , DNA/chemistry , DNA/genetics , Family Health , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Nucleic Acid , Tourette Syndrome/pathology
10.
Prostate ; 45(2): 106-14, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11027409

ABSTRACT

BACKGROUND: Since the publication of the first genome screen for prostate cancer (CaP) 5 years ago, over a dozen linkage studies have appeared. Most attention has been directed to chromosome 1, where two separate regions have been identified as harboring a prostate cancer susceptibility locus: HPC1 in the 1q24-25 interval and PCaP in the 1q42.2-43 interval. Linkage analysis of chromosome 16 has also provided evidence of harboring two loci predisposing to CaP. METHODS: We report on a replication linkage study of chromosomes 1 and 16 in 45 new and 4 expanded multiplex CaP families. Multipoint Z-scores were obtained for 30 highly polymorphic short-sequence tandem repeat markers spanning chromosome 1, and 22 markers spanning chromosome 16. RESULTS: The replication sample gave no evidence for a CaP susceptibility locus in the 1q24-25 interval and equivocal evidence for such a locus at 1q42.2-43. With respect to chromosome 16, positive Z-scores were obtained over a contiguous interval covering the entire p arm and the proximal half of the q arm. CONCLUSIONS: The linkage analysis of our replication sample does not support the existence of HPC1, and the evidence for the existence of PCaP remains equivocal. Evidence of a susceptibility locus on 16p remains strong, but the evidence for a susceptibility locus on 16q is weakened.


Subject(s)
Chromosomes, Human, Pair 16 , Chromosomes, Human, Pair 1 , Genetic Linkage , Prostatic Neoplasms/genetics , Genetic Heterogeneity , Genetic Predisposition to Disease , Humans , Male
11.
Med Oncol ; 17(3): 211-7, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10962532

ABSTRACT

Recent reports have demonstrated that the HIV-1 transactivator protein, tat, induces apoptosis in T-lymphocyte cell lines, as well as in peripheral blood mononuclear cells, and stimulates a cascade of events resulting in up-regulation of the potent immunosuppressive cytokine, transforming growth factor-beta (TGF-beta). In this study we evaluated the ability of TGF-beta to mediate tat induced apoptosis in T-lymphocyte cell lines. T-cells treated exogenously with either TGF-beta1 or a combination of tat and pan-specific TGF-beta neutralizing antibodies showed little change in the amount of apoptosis. When treated with pan-specific TGF-beta neutralizing antibodies, Jurkat cells that stably express tat protein (Jurkat-tat ) showed only a modest decrease in apoptosis, while CEM-TART cells (CEM T-cells expressing both HIV-1 tat and rev ) demonstrated little change in the amount of apoptosis. In conclusion, we have demonstrated that TGF-beta does not play a significant role in mediating tat induced T-cell apoptosis.


Subject(s)
Apoptosis , Gene Products, tat/pharmacology , T-Lymphocytes/drug effects , Transforming Growth Factor beta/pharmacology , HIV-1/chemistry , Humans , Leukemia, T-Cell/pathology , T-Lymphocytes/immunology , Transforming Growth Factor beta/immunology , Tumor Cells, Cultured , tat Gene Products, Human Immunodeficiency Virus
12.
Am J Hum Genet ; 67(1): 92-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10825281

ABSTRACT

The aggressiveness of prostate cancer (PCa) varies widely: some tumors progress to invasive, potentially life-threatening disease, whereas others stay latent for the remainder of an individual's lifetime. The mechanisms resulting in this variability are not yet understood, but they are likely to involve both genetic and environmental influences. To investigate genetic factors, we conducted a genomewide linkage analysis of 513 brothers with PCa, using the Gleason score, which reflects tumor histology, as a quantitative measure of PCa aggressiveness. To our knowledge, this is the first time that a measure of PCa aggressiveness has been directly investigated as a quantitative trait in a genomewide scan. We employed a generalized multipoint Haseman-Elston linkage-analysis approach that regresses the mean-corrected cross product between the brothers' Gleason scores on the estimated proportion of alleles shared by brothers identical by descent at each marker location. Our results suggest that candidate regions on chromosomes 5q, 7q, and 19q give evidence for linkage to PCa-aggressiveness genes. In particular, the strongest signals detected in these regions were at the following markers (with corresponding P values): for chromosome 5q31-33, between markers D5S1480 and D5S820 (P=.0002); for chromosome 7q32, between markers D7S3061 and D7S1804 (P=.0007); and, for chromosome 19q12, at D19S433 (P=.0004). This indicates that one or more of these candidate regions may contain genes that influence the progression of PCa from latent to invasive disease. Identification of such genes would be extremely valuable for elucidation of the mechanism underlying PCa progression and for determination of treatment in men in whom this disease has been diagnosed.


Subject(s)
Genetic Linkage/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Chromosome Mapping , Chromosomes, Human/genetics , Genetic Markers/genetics , Genetic Testing , Genome, Human , Humans , Male , Matched-Pair Analysis , Middle Aged , Neoplasm Invasiveness , Nuclear Family , Phenotype
13.
Am J Hum Genet ; 66(3): 933-44, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10712208

ABSTRACT

Analysis of a genome screen of 504 brothers with prostate cancer (CaP) who were from 230 multiplex sibships identified five regions with nominally positive linkage signals, on chromosomes 2q, 12p, 15q, 16p, and 16q. The strongest signal in these data is found on chromosome 16q, between markers D16S515 and D16S3040, a region suspected to contain a tumor-suppressor gene. On the basis of findings from previous genome screens of families with CaP, three preplanned subanalyses were carried out, in the hope of increasing the subgroup homogeneity. Subgroups were formed by dividing the sibships into a group with a positive family history (FH+) that met criteria for "hereditary" CaP (n=111) versus those which did not meet the criteria (n=119) and by dividing the families into those with a mean onset age below the median (n=115) versus those with a mean onset age above the median (n=115). A separate subanalysis was carried out for families with a history of breast cancer (CaB+ [n=53]). Analyses of these subgroups revealed a number of potentially important differences in regions that were nonsignificant when all the families were analyzed together. In particular, the subgroup without a positive family history (FH-) had a signal in a region that is proximal to the putative site of the HPC1 locus on chromosome 1, whereas the late-age-at-onset group had a signal on 4q. The CaB+ subgroup revealed a strong linkage signal at 1p35.1.


Subject(s)
Genetic Heterogeneity , Genetic Linkage/genetics , Genetic Testing , Genome, Human , Nuclear Family , Prostatic Neoplasms/genetics , Age of Onset , Alleles , Breast Neoplasms/epidemiology , Breast Neoplasms/genetics , Female , Genetic Markers/genetics , Genetic Predisposition to Disease/genetics , Genotype , Humans , Male , Phenotype , Prostatic Neoplasms/epidemiology
14.
Growth Factors ; 17(1): 63-73, 1999.
Article in English | MEDLINE | ID: mdl-10495963

ABSTRACT

Transforming growth factor-beta (TGF-beta) is a multi-functional regulator of cell growth and differentiation. Three distinct isoforms of TGF-beta exist having similar, but not identical actions. TGF-beta 1, but not TGF-beta 2, binds to T beta RII and also to endoglin, a cell surface protein abundant on endothelial cells. In contrast, the affinity constant of TGF-beta 2 for alpha 2-macroglobulin is 10-fold greater than that of TGF-beta 1. TGF-beta 2 also binds better than TGF-beta 1 to a glycosyl phosphatidylinositol (GPI)-linked binding protein expressed on vascular endothelial cells. Using chimeric TGF-beta molecules, in which selected regions of TGF-beta 1 have been exchanged for the corresponding region of TGF-beta 2, we demonstrate here that amino acids 92-95 or 94-98 of TGF-beta determine isoform specific binding to endoglin. In contrast, exchange of only amino acids 95 and 98 did not alter TGF-beta specificity. Isoform specific binding to a GPI-linked protein on EJG endothelial cells was modulated by a region containing amino acids 40-68, although exchange of only amino acids 40-47 did not confer isoform specific binding. Significantly, the 92-98 region also modulates binding of TGF-beta to the type II receptor whereas isoform specific binding to alpha 2-macroglobulin requires concerted exchange of amino acids 45 and 47. Taken together, these results show that at least three different functional domains are important modulators of TGF-beta interaction with binding proteins and receptors.


Subject(s)
Endothelium, Vascular/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , alpha-Macroglobulins/metabolism , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Cells, Cultured , Cricetinae , Endothelium, Vascular/cytology , Humans , Models, Molecular , Molecular Sequence Data , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases , Protein Structure, Tertiary , Receptor, Transforming Growth Factor-beta Type II , Sequence Homology, Amino Acid , Structure-Activity Relationship , Transforming Growth Factor beta/chemistry
15.
Med Oncol ; 16(2): 110-8, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10456659

ABSTRACT

Patients with chronic lymphocytic leukemia (CLL) frequently respond to initial treatment, but then become resistant to chemotherapy. Studies have shown one important cause of chemotherapeutic resistance to be multidrug resistance (MDR). To investigate the potential role of MDR and transforming growth factor-beta (TFG-beta), a potent growth inhibitor of B lymphocytes, in the development of chemotherapeutic resistance in CLL, we evaluated 22 CLL patients for loss or mutation of TGF-beta receptors (TbetaR), plasma TGF-beta1 levels, and expression of MDR1 mRNA. Receptor crosslinking and immunoprecipitation experiments did not demonstrate loss of TbetaRs in any patients studied. No relationship between plasma TGF-beta1 levels and expression of MDR1 mRNA was seen. Correlation of plasma TGF-beta1 levels to disease stage revealed a consistent decline in plasma TGF-beta1 levels with advancing disease stage (P = 0.031).


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Multiple , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Receptors, Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/blood , Adult , Aged , Aged, 80 and over , DNA Mutational Analysis , Disease Progression , Drug Resistance, Multiple/genetics , Female , Gene Expression , Genes, MDR , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Male , Middle Aged , Precipitin Tests , Receptors, Transforming Growth Factor beta/blood , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/genetics
16.
Am J Rhinol ; 13(3): 221-3, 1999.
Article in English | MEDLINE | ID: mdl-10392242

ABSTRACT

Nasal polyps are the most common mass lesions found in the nose and their etiology is unknown. Nasal polyps from cystic fibrosis (CF) patients are histologically distinct from nasal polyps from patients without CF. It has been suggested that a mutation (G551D) of the cystic fibrosis transmembrane conductance regulator (CFTR) gene may play a role in nasal polyp formation in patients without CF. To investigate the possibility that this or other CFTR gene exon mutations are required for nasal polyp formation, the CFTR gene exons were sequenced from peripheral blood DNA derived from an adult patient with edematous eosinophilic nasal polyps and no personal or family history of CF. No mutations or deletions were identified in any of the CFTR exons. A single polymorphism (A or G) was found in exon 10, base pair 1540, amino acid 470. This polymorphism was detected in 11 of 16 subjects (69%) with edematous eosinophilic nasal polyps and 10 of 21 normal subjects (48%) without nasal polyps and was not statistically significant (p = 0.316). These results demonstrate that mutations of the CFTR coding region are not a prerequisite for the formation of edematous eosinophilic nasal polyps.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Nasal Polyps/genetics , Sequence Analysis, DNA , Edema/pathology , Eosinophilia/pathology , Exons/genetics , Humans , Male , Middle Aged , Mutation , Nasal Polyps/pathology , Polymorphism, Genetic
17.
Growth Factors ; 15(3): 231-42, 1998.
Article in English | MEDLINE | ID: mdl-9570043

ABSTRACT

Transforming growth factor-beta s (TGF-beta 1, -beta 2, -beta 3) are important regulators of cell growth and differentiation which share approximately 70% identical amino acids. Using LS513 colorectal cells, which are growth inhibited by TGF-beta 1 (ED50 of 100 pM), but are refractory to TGF-beta 2 (ED50 of 50,000 to 100,000 pM), we have determined that amino acids 92-98 of TGF-beta specify growth inhibition. The chimeric protein TGF-beta 1/beta 2(92-98), in which amino acids 92-98 of TGF-beta 1 were exchanged for the corresponding amino acids of TGF-beta 2, was indistinguishable from TGF-beta 2 at inhibiting growth of LS513 cells. In contrast, both TGF-beta 1/beta 2(92-95) and TGF-beta 1/beta 2(94-98) inhibited the growth of LS513 cells with an ED50 of approximately 1000 pM. TGF-beta 1/beta 2(95-98), in which amino acids 95-98 of TGF-beta 1 have been replaced with the corresponding amino acids of TGF-beta 2, had full activity and was indistinguishable from TGF-beta 1. Receptor cross-linking experiments demonstrated that binding of the chimeras to the type I and type II receptors of LS513 cells was consistent with their biological activity. TGF-beta 1/beta 2(95-98), TGF-beta 1/beta 2(92-95) and TGF-beta 1/beta 2(94-98) were each similar to TGF-beta 2 in that they failed to bind to the soluble Type II receptor in a solid-phase assay. These results demonstrate that amino acids 92-98 are involved in the interaction between TGF-beta and its signaling receptors and they show that modest changes within this region can substantially alter biological response.


Subject(s)
DNA Mutational Analysis , Receptors, Transforming Growth Factor beta/genetics , Amino Acid Sequence , Amino Acid Substitution , Binding Sites/genetics , Cell Line , Colorectal Neoplasms/pathology , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Receptors, Transforming Growth Factor beta/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
18.
J Biol Chem ; 271(48): 30656-62, 1996 Nov 29.
Article in English | MEDLINE | ID: mdl-8940041

ABSTRACT

Transforming growth factor-beta (TGF-beta) isoforms have differential binding affinities for the TGF-beta type II receptor (TbetaRII). In most cells, TGF-beta1 and TGF-beta3 bind to TbetaRII with much higher affinity than TGF-beta2. Here, we report an analysis of the effect of TGF-beta structure on its binding to TbetaRII by using TGF-beta mutants with domain deletions, amino acid replacements, and isoform chimeras. Examination of the binding of TGF-beta mutants to the recombinant extracellular domain of TbetaRII by a solid-phase TGF-beta/TbetaRII assay demonstrated that only those TGF-beta mutants containing the C terminus of TGF-beta1 (TGF-beta1-(Delta69-73), TGF-beta1-(Trp71), and TGF-beta2/beta1-(83-112)) bind with high affinity to TbetaRII, similar to native TGF-beta1. Moreover, replacement of only 6 amino acids in the C terminus of TGF-beta1 with the corresponding sequence of TGF-beta2 (TGF-beta1/beta2-(91-96)) completely eliminated the high affinity binding of TGF-beta1. Proliferation of fetal bovine heart endothelial (FBHE) cells was inhibited to a similar degree by all of the TGF-beta mutants. However, recombinant soluble TbetaRII blocked the inhibition of FBHE cell proliferation induced by TGF-beta mutants retaining the C terminus of TGF-beta1, consistent with the high binding affinity between these TGF-beta molecules and TbetaRII. It was further confirmed that the TGF-beta2 mutant with its C terminus replaced by that of TGF-beta1 (TGF-beta2/beta1-(83-112)) competed as effectively as TGF-beta1 with 125I-TGF-beta1 for binding to membrane TbetaRI and TbetaRII on FBHE cells. These observations clearly indicate that the domain in TGF-beta1 responsible for its high affinity binding to TbetaRII, both the soluble and membrane-bound forms, is located at C terminus of the molecule.


Subject(s)
Receptors, Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/chemistry , Amino Acid Sequence , Animals , Binding, Competitive , Biological Assay , Cattle , Cell Division , Cells, Cultured , Epitope Mapping , Humans , Molecular Sequence Data , Protein Binding , Protein Serine-Threonine Kinases , Protein Structure, Tertiary , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Recombinant Proteins , Sequence Alignment , Solubility , Structure-Activity Relationship , Transforming Growth Factor beta/metabolism
19.
J Biol Chem ; 270(49): 29460-8, 1995 Dec 08.
Article in English | MEDLINE | ID: mdl-7493985

ABSTRACT

Diminished cellular responsiveness to transforming growth factor-beta (TGF-beta) is frequently correlated with decreased transcription of the type II receptor for TGF-beta (TGF-beta RII). We have cloned and characterized the human TGF-beta RII promoter and, using S1 nuclease mapping and 5' rapid amplification of cDNA ends polymerase chain reaction, have identified five alternative transcription start sites within the region -33 to +57. DNA transfection experiments and electrophoretic mobility shift assays have revealed the existence of five distinct regulatory regions including two positive regulatory elements and two negative regulatory elements in addition to the core promoter region. The first positive regulatory element (-219 to -172) interacts with two distinct nuclear protein complexes, at least one of which appears to be a previously unidentified transcription factor. The second positive regulatory element (+1 to +35) also interacts with two separate protein complexes, both of which appear to be novel transcription factors. Deletion of either positive regulatory element markedly decreased expression of the target gene, suggesting that both positive regulatory elements are necessary for basal expression levels of TGF-beta RII.


Subject(s)
Promoter Regions, Genetic , Receptors, Transforming Growth Factor beta/genetics , Base Sequence , DNA, Complementary/isolation & purification , Genes, Regulator , Humans , Molecular Sequence Data , Transcription, Genetic
20.
J Biol Chem ; 269(48): 30402-6, 1994 Dec 02.
Article in English | MEDLINE | ID: mdl-7527030

ABSTRACT

Transforming growth factor beta 2 (TGF-beta 2) is less potent than TGF-beta 1 in some endothelial cell proliferation assays due to the greater tendency of TGF-beta 2 to bind alpha 2-macroglobulin (alpha 2M). Substitution of TGF-beta 1 residues 40-47 into the TGF-beta 2 sequence yields a chimeric molecule that, like TGF-beta 1, expresses activity that is not substantially affected by serum alpha 2M (Burmester, J. K., Qian, S. W., Roberts, A. B., Huang, A., Amatayakul-Chantler, S., Suardet, L., Odartchenko, N., Madri, J. A., and Sporn, M. B. (1993) Proc. Natl. Acad. Sci. U.S.A. 90, 8628-8632). In this investigation, we studied the binding of TGF-beta chimeras, which contain TGF-beta 1 residues 40-47, to both major conformations of human alpha 2M under apparent equilibrium conditions. Native alpha 2M, the primary form of this protein in serum, bound TGF-beta 2/beta 1 (40-82) and TGF-beta 2/beta 1 (40-47) with low affinity. The apparent KD values for the two chimeras and native alpha 2M were 310 and 330 nM, respectively. These values were much higher than the KD determined for TGF-beta 2 and native alpha 2M (11 nM) and equivalent to the KD determined for TGF-beta 1 and native alpha 2M. By contrast, both TGF-beta chimeras bound alpha 2M-methylamine, an altered conformation of alpha 2M, with high affinity (16 and 19 nM), which is characteristic of TGF-beta 2 and not TGF-beta 1. Fetal bovine heart endothelial cell DNA synthesis was inhibited to a similar degree by TGF-beta 1, TGF-beta 2, TGF-beta 2/beta 1 (40-82), and TGF-beta 2/beta 1 (40-47) in the presence of dilute (0.2%) fetal bovine serum. When 0.07 microM alpha 2M-methylamine was added, the activities of TGF-beta 2, TGF-beta 2/beta 1 (40-82), and TGF-beta 2/beta 1 (40-47) were significantly counteracted while the activity of TGF-beta 1 was unchanged, as would be predicted by the equilibrium binding analyses. These studies indicate that the TGF-beta structural elements, which mediate binding to native alpha 2M and conformationally transformed alpha 2M, are not equivalent. Residues 40-47 are critical in determining affinity for native alpha 2M but are less important in determining affinity for alpha 2M-methylamine.


Subject(s)
Transforming Growth Factor beta/metabolism , alpha-Macroglobulins/metabolism , Animals , Autoradiography , Blood , Cattle , Electrophoresis, Polyacrylamide Gel , Fetus , Humans , Iodine Radioisotopes , Kinetics , Protein Binding , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/isolation & purification , alpha-Macroglobulins/chemistry , alpha-Macroglobulins/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...