Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
J Phys Condens Matter ; 29(22): 225901, 2017 Jun 07.
Article in English | MEDLINE | ID: mdl-28452343

ABSTRACT

Interatomic potentials for Pt and Au are developed within the third generation charge optimized many-body (COMB3) formalism. The potentials are capable of reproducing phase order, lattice constants, and elastic constants of Pt and Au systems as experimentally measured or calculated by density functional theory. We also fit defect formation energies, surface energies and stacking fault energies for Pt and Au metals. The resulting potentials are used to map a 2D contour of the gamma surface and simulate the tensile test of 16-grain polycrystalline Pt and Au structures at 300 K. The stress-strain behaviour is investigated and the primary slip systems {1 1 1}〈1 [Formula: see text] 0〉 are identified. In addition, we perform high temperature (1800 K for Au and 2300 K for Pt) molecular dynamics simulations of 30 nm Pt and Au truncated octahedron nanoparticles and examine morphological changes of each particle. We further calculate the activation energy barrier for surface diffusion during simulations of several nanoseconds and report energies of [Formula: see text] eV for Pt and [Formula: see text] eV for Au. This initial parameterization and application of the Pt and Au potentials demonstrates a starting point for the extension of these potentials to multicomponent systems within the COMB3 framework.

2.
Cancer Gene Ther ; 20(7): 403-12, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23764899

ABSTRACT

Colorectal cancer cells exhibit limited cytotoxicity towards Tiazofurin, a pro-drug metabolized by cytosolic nicotinamide mononucleotide adenylyltransferase2 (NMNAT2) to thiazole-4-carboxamide adenine dinucleotide, a potent inhibitor of inosine 5'-monophosphate dehydrogenase required for cellular guanylate synthesis. We tested the hypothesis that colorectal cancer cells that exhibit low levels of NMNAT2 and are refractory to Tiazofurin can be rendered sensitive to Tiazofurin by overexpressing NMNAT2. Transfection of hNMNAT2 resulted in a six- and threefold cytoplasmic overexpression in Caco2 and HT29 cell lines correlating with Tiazofurin-induced enhanced cell-kill. Folate receptors expressed on the cell surface of 30-50% colorectal carcinomas were exploited for cellular targeting with Tiazofurin encapsulated in folate-tethered nanoparticles. Our results indicated that in wild-type colorectal cancer cells, free Tiazofurin-induced EC50 cell-kill was 1500-2000 µM, which was reduced to 66-156 µM in hNMNAT2-overexpressed cells treated with Tiazofurin encapsulated in non-targeted nanoparticles. This efficacy was improved threefold by encapsulating Tiazofurin in folate-tethered nanoparticles to obtain an EC(50) cell-kill of 22-59 µM, an equivalent of 100-300 mg m(-2) (one-tenth of the approved dose of Tiazofurin in humans), which will result in minimal toxicity leading to cancer cell-kill. This proof-of-principle study suggests that resistance of colorectal cancer cell-kill to Tiazofurin can be overcome by sequentially overexpressing hNMNAT2 and then facilitating the uptake of Tiazofurin by folate-tethered nanoparticles, which enter cells via folate receptors.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Gene Expression , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Ribavirin/analogs & derivatives , Antimetabolites, Antineoplastic/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms , Drug Carriers/metabolism , Drug Resistance, Neoplasm , Fluoresceins/metabolism , Folic Acid Transporters/metabolism , Gene Knockdown Techniques , Guanosine Triphosphate/metabolism , Humans , IMP Dehydrogenase/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Nanoparticles/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Protein Transport , RNA, Small Interfering/genetics , Ribavirin/metabolism , Ribavirin/pharmacology , Transfection
3.
J Clin Pathol ; 56(8): 569-74, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12890803

ABSTRACT

AIMS: Folate receptors (FRs) mediate cellular uptake of folates in many cancer cells and in folate deficiency heterogeneous nuclear ribonucleoprotein E1 (hnRNP-E1) mediates translational upregulation of FR in cultured cervical cancer cells. hnRNP-E1 can also interfere with human papillomavirus 16 (HPV-16) viral capsid protein synthesis (and thereby HPV proliferation) in vitro. This study aimed to evaluate prospectively the relevance of FR and hnRNP-E1 expression in the normal cervix, cervical dysplasia, and cancer. METHODS: Cervical tissues from 12 women with normal histology and 69 consecutive women with varying grades of cervical dysplasia and cancer were prospectively evaluated for immunohistochemical expression of FR, hnRNP-E1, proliferating cell nuclear antigen (PCNA), and HPV. There were 22 women with low grade squamous intraepithelial lesions (LGSIL), 22 with high grade squamous intraepithelial lesions (HGSIL), and 25 with invasive cervical carcinoma. RESULTS: Among normal subjects, 100% and 92% expressed hnRNP-E1 and FR, respectively. FR expression decreased from 91% in LGSIL to 68% and 64% in women with HGSIL and cancer, respectively. Similarly, hnRNP-E1 expression decreased from 86% in LGSIL to 68% and 40% in HGSIL and cancer, respectively. There was a highly significant positive correlation between the extent of FR and hnRNP-E1 expression, and an inverse correlation between HPV infection and hnRNP-E1 expression during progression of cervical dysplasia to cancer. CONCLUSION: These results are consistent with a hypothesis that reduced hnRNP-E1 expression may be permissive for HPV proliferation and progression to cervical cancer, and support the need for prospective longitudinal studies of hnRNP-E1 expression in HPV-16 infected women.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/chemistry , Carrier Proteins/analysis , Heterogeneous-Nuclear Ribonucleoproteins/analysis , Papillomaviridae , Receptors, Cell Surface , Repressor Proteins , Uterine Cervical Neoplasms/chemistry , Biopsy , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/virology , Case-Control Studies , Cell Transformation, Viral , Cervix Uteri/pathology , Cervix Uteri/virology , Chi-Square Distribution , DNA-Binding Proteins , Female , Folate Receptors, GPI-Anchored , Humans , Immunohistochemistry/methods , Oncogene Proteins, Viral/analysis , Papillomavirus Infections/diagnosis , Proliferating Cell Nuclear Antigen/analysis , Prospective Studies , RNA-Binding Proteins , Statistics, Nonparametric , Tumor Virus Infections/diagnosis , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/virology , Uterine Cervical Dysplasia/diagnosis , Uterine Cervical Dysplasia/pathology
4.
J Biol Chem ; 276(44): 41510-7, 2001 Nov 02.
Article in English | MEDLINE | ID: mdl-11527973

ABSTRACT

The interaction of an 18-base cis-element in the 5'-untranslated region of human folate receptor (FR)-alpha mRNA with a cytosolic trans-factor protein is critical for the translation of FR (Sun, X.-L., and Antony, A. C. (1996) J. Biol. Chem. 271, 25539-25547). This trans-factor was isolated to apparent homogeneity as a 43- and 38-kDa doublet from human placenta using poly(U)-Sepharose, followed by preparative sodium dodecyl sulfate-polyacrylamide gel electrophoresis and electro-elution as major purification steps. Amino acid microsequencing of two cyanogen bromide-generated peptide fragments of the 43-kDa trans-factor revealed complete identity with 43-kDa heterogeneous nuclear ribonucleoprotein E1 (hnRNP E1). Purified specific rabbit anti-hnRNP E1 peptide antibodies (generated against a synthetic oligopeptide that was not represented in microsequenced peptides of the trans-factor) also recognized the purified trans-factor on Western blots. Conversely, the 18-base FR RNA cis-element also bound hnRNP E1 protein on Northwestern blots. Moreover, a 19-base RNA cis-element in the 3'-untranslated region of 15-lipoxygenase mRNA that is known to bind hnRNP E1 also interacted with placental 43-kDa trans-factor. In addition, several murine tissues containing a hnRNP E1-related protein (also known as alphaCP-1) readily interacted with the 18-base FR RNA cis-element. Finally, anti-hnRNP E1 antibodies specifically inhibited translation of FR in vitro in a dose-dependent manner, and the antibody effect could be reversed in a dose-dependent manner by either purified trans-factor or hnRNP E1. Collectively, the data favor identity of the FR mRNA-binding trans-factor and hnRNP E1, confirm its critical role in the translation of FR, and highlight yet another role of multifunctional hnRNP E1 in eukaryotic mRNA regulation.


Subject(s)
Carrier Proteins/genetics , Placenta/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/isolation & purification , Receptors, Cell Surface , Amino Acid Sequence , Base Sequence , DNA Primers , Female , Folate Receptors, GPI-Anchored , Heterogeneous-Nuclear Ribonucleoproteins , Humans , Molecular Sequence Data , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism
6.
Teratology ; 62(1): 42-50, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10861632

ABSTRACT

BACKGROUND: What accounts for the wide spectrum of folate-responsive dysmorphogeneses? Both embryonic and fetal cells are entirely dependent on maternal folate to support their requirement for precisely timed proliferative bursts during gestation. Folate receptors (FRs) mediate transport into cells and are central to transplacental maternal-to-fetal folate transport. FRs are also critical for neural tube and neural crest development because recent murine "knock-out" and "knock-down" of FRs results in a high percentage of folate-responsive neural tube defects (NTDs) and neurocristopathies. HYPOTHESIS: Central to our hypothesis is the fact that folate deficiency is accompanied by a reduction in the proliferative capacity of highly mitotic neural tube or neural crest cells. Therefore, depending on when in pregnancy various cohorts of highly proliferative cells are deprived of folate, and the origin of the affected cells will determine the type of developmental dysmorphogenesis. Thus, selective folate deficiency in early pregnancy of only highly proliferative neural tube or neural crest cells predisposes to NTDs or gross dysmorphogenesis, respectively. Folate deficiency that compromises placental development will predispose to small-for-date babies due to an overall nutrient deficiency, and the development of folate insufficiency later in pregnancy could predispose to more subtle midline birth defects involving atresia of neural crest cell-derived structures. Finally, a congenital folate transport defect would only be corrected by suprapharmacological doses of folate, which ensures passive diffusion. CONCLUSION: This hypothesis can explain the results of several earlier and more recent clinical trials on folate supplementation in pregnancy, but it also raises the possibility that there may be several as yet undiscovered neurocristopathies that are folate responsive. Teratology 62:42-50, 2000. Published 2000 Wiley-Liss, Inc.


Subject(s)
Folic Acid Deficiency/complications , Models, Neurological , Neural Crest/abnormalities , Neural Tube Defects/etiology , Receptors, Cell Surface , Animals , Base Sequence , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Division , DNA, Antisense/administration & dosage , DNA, Antisense/genetics , Female , Folate Receptors, GPI-Anchored , Folic Acid/metabolism , Folic Acid Deficiency/metabolism , Humans , Infant, Newborn , Mice , Mice, Knockout , Neural Crest/embryology , Neural Crest/metabolism , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Pregnancy
7.
Blood ; 93(11): 3940-8, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10339503

ABSTRACT

We have investigated the expression and functional competence of folate receptor (FR) isoforms on human hematopoietic cells. Using immunofluorescence and reverse transcriptase-polymerase chain reaction (RT-PCR) methodology, we find that a substantial fraction of low-density mononuclear and CD34(+) cells express both the beta and gamma isoforms of FR. The alpha isoform of FR (the form most commonly found on cancer cells) was surprisingly absent from all hematopoietic cells examined. Compared with KB cells (a human cell line known for its elevated expression of FR-alpha), the abundance of FR-beta on CD34(+) cell surfaces was relatively low (approximately 8% of KB cell levels). Because many antifolates and folic acid-linked chemotherapeutic agents enter malignant cells at least partially via FR endocytosis, it was important to evaluate the ability of FR on CD34(+) cells to bind folic acid (FA). Based on three FR binding assays, freshly isolated CD34(+) cells were found to display no affinity for FA. Thus, regardless of whether steps were taken to remove endogenous folates before receptor binding assays, FR on primitive hematopoietic cells failed to bind 3H-FA, fluorescein isothiocyanate (FITC)-linked FA, or FA-derivatized liposomes. In contrast, analogous studies on KB cells showed high levels of receptor binding for all three FR probes. These studies show that although multipotent hematopoietic progenitor cells express FR, the receptor does not transport significant amounts of FA. Consequently, antifolates and FA-linked chemotherapeutic agents that can be engineered to enter malignant cells exclusively through the FR should not harm progenitor/stem cell function.


Subject(s)
Carrier Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Antigens, CD34 , Cell Line , Flow Cytometry , Folate Receptors, GPI-Anchored , Humans , Protein Isoforms/metabolism , Receptors, Cell Surface/metabolism
8.
Cancer Res ; 59(4): 940-6, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-10029088

ABSTRACT

Cervical carcinoma is an AIDS-defining illness. The expression of folate receptors (FRs) in cervical carcinoma (HeLa-IU1) cells was modulated by stable transduction of FR cDNA encapsidated in recombinant adeno-associated virus-2 in the sense and antisense orientation (sense and antisense cells, respectively). Although sense cells proliferated slower than antisense or untransduced cells in vivo and in vitro in 2% (but not 10%) FCS, [methyl-3H]thymidine incorporation into DNA was significantly increased in sense cells in 10% serum; therefore, the basis for this discrepancy was investigated. The activity of thymidine kinase (TK) was subsequently directly correlated with the extent of FR expression in single cell-derived clones of transduced cells. This elevated TK activity was not a result of recruitment of the salvage pathway based on the presence of adequate dTTP pools, normal thymidylate synthase (TS) activity, persistence of increased thymidine incorporation despite the exogenous provision of excess 5,10-methylene-tetrahydrofolate, and documentation of adequate folates in sense cells. The increase in TK activity conferred significant biological properties to sense cells (but not antisense or untransduced cells) as demonstrated by augmented phosphorylation of 3'-azido-3'-deoxythymidine (AZT) and concomitantly greater sensitivity to the cytotoxic effects of AZT. Conversely, sense cells were highly resistant to methotrexate, but this was reversed by the addition of AZT. The direct correlation of FR expression and TK activity indicates a previously unrecognized consequence of FR overexpression.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Carrier Proteins/genetics , Gene Transfer Techniques , Methotrexate/pharmacology , Receptors, Cell Surface , Thymidine Kinase/metabolism , Uterine Cervical Neoplasms/drug therapy , Zidovudine/pharmacology , Carrier Proteins/analysis , Drug Resistance, Neoplasm , Female , Folate Receptors, GPI-Anchored , Humans , Thymidine/metabolism , Uterine Cervical Neoplasms/metabolism
9.
J Biol Chem ; 271(41): 25539-47, 1996 Oct 11.
Article in English | MEDLINE | ID: mdl-8810326

ABSTRACT

Folate receptors (FR) are inversely regulated by the extracellular folate concentration at the translational level in cervical carcinoma cells. Accordingly, the potential for interaction of cis-elements in FR-alpha mRNA and trans-factors in these cells was determined. Gel-shift assays identified two signals that were specifically derived from the interaction of cytosolic proteins with the 5'-untranslated region of FR-alpha mRNA. RNase T1 mapping revealed that the RNA sequences interacting with these proteins were located between nucleotides -133 to -116 (18-bases) and -158 to -116 (43-bases), upstream of the translation start site. However, selective RNase H cleavage indicated that the 18-base RNA sequence was the cis-element. The RNA-protein interaction was competed by poly(C), but not by poly(U), homopolymers. UV cross-linking and Northwestern blot analysis confirmed that the trans-factors were 46-kDa proteins. An 18-base antisense oligodeoxynucleotide complementary to the cis-element specifically quenched the RNA-protein interaction and also completely inhibited translation of FR-alpha mRNA without changing its stability. Thus, the interaction of the 18-base cis-element and the 46-kDa trans-factors likely have an important role in translational regulation of FR. In addition, because the 46-kDa proteins were widely distributed in cells expressing little to no FR-alpha, these species probably have additional functions that are unrelated to translation of FR.


Subject(s)
Carrier Proteins/biosynthesis , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Receptors, Cell Surface , Base Composition , Base Sequence , Carrier Proteins/genetics , Cloning, Molecular , Cytosol/metabolism , Female , Folate Receptors, GPI-Anchored , Gene Expression Regulation, Neoplastic , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Protein Biosynthesis , RNA, Messenger/isolation & purification , RNA-Binding Proteins/isolation & purification , Recombinant Proteins/biosynthesis , Regulatory Sequences, Nucleic Acid , Ribonuclease H , Ribonuclease T1 , Uterine Cervical Neoplasms/metabolism
10.
J Biol Chem ; 271(19): 11493-9, 1996 May 10.
Article in English | MEDLINE | ID: mdl-8626708

ABSTRACT

Glycosyl-phosphatidylinositol-anchored hydrophobic placental folate receptors (PFRs), which have an important functional role in maternal-to-fetal transplacental folate transport, can be converted to soluble hydrophilic forms by a placental metalloprotease. Using a Triton X-114 temperature-induced phase separation assay to monitor enzyme-mediated conversion of radiolabeled hydrophobic PFR into hydrophilic PFR, a metalloenzyme was isolated to apparent homogeneity from Triton X-114-solubilized human placenta using concanavalin A-Sepharose and reverse-phase high performance liquid chromatography (HPLC) as major purification steps. The purified hydrophobic enzyme eluted as a single protein peak on reverse-phase HPLC and SDS-polyacrylamide gel electrophoresis revealed a single 63,000 M(r) species, which was reduced to 58,000 M(r) following deglycosylation, findings comparable with amino acid analysis (M(r) approximately 59,000). The metalloenzyme was activated by Mg2+, Zn2+, Mn2+, and Ca2+, optimally at physiologic pH; it also exhibited EDTA-sensitive endoproteolytic cleavage of [3H]leucine-labeled full-length nascent PFR polypeptide generated in vitro in the absence of microsomes. Rabbit polyclonal anti-metalloprotease antiserum specifically immunoprecipitated 125I-metalloprotease and recognized cross-reacting moieties on plasma membranes of normal human hematopoietic progenitor cells and human cervical carcinoma cells, both of which also express FR.


Subject(s)
Carrier Proteins/metabolism , Metalloendopeptidases/isolation & purification , Metalloendopeptidases/metabolism , Placenta/enzymology , Receptors, Cell Surface , Amino Acids/analysis , Animals , Antibodies , Cations, Divalent/pharmacology , Chromatography, Affinity , Chromatography, Gel , Chromatography, High Pressure Liquid , Concanavalin A , Cross Reactions , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Female , Folate Receptors, GPI-Anchored , Humans , Kinetics , Metalloendopeptidases/chemistry , Molecular Weight , Pregnancy , Rabbits , Tumor Cells, Cultured , Uterine Cervical Neoplasms/enzymology
11.
Annu Rev Nutr ; 16: 501-21, 1996.
Article in English | MEDLINE | ID: mdl-8839936

ABSTRACT

Glycosyl-phosphatidylinositol-anchored folate receptors (FR) have physiologic and pharmacologic relevance in mediating cellular and transcellular folate/antifolate transport. Three FR isoforms with differing relative affinities for folates and expression patterns in normal and malignant cells/tissues are recognized, but the precise mechanism of cellular entry of folate via FR remains controversial. Although FR expression allows previously FR-deficient cells to survive a reduced folate milieu, an inverse relationship between FR expression and cell proliferation has been established in some cells. The inverse regulation of FR expression by the extracellular folate concentration suggests heterogeneity in underlying mechanisms. Whereas reduced FR expression is yet another mechanism for acquiring antifolate resistance, overexpression of FR does not invariably render cells more sensitive to antifolates. The exploitation of FRs as Trojan horses to deliver folate-tagged liposomes bearing diverse cargo represents a novel therapeutic strategy to target FR-expressing cells. Finally, a critically important role of human placental FR in mediating maternal-to-fetal transplacental transport of folates has been established. Thus, FR appear to have a major impact on several aspects of human physiology and medicine.


Subject(s)
Carrier Proteins , Receptors, Cell Surface , Animals , Biological Transport , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/physiology , Cell Division , Folate Receptors, GPI-Anchored , Folic Acid/metabolism , Gene Expression Regulation , Humans
12.
J Clin Invest ; 96(3): 1535-47, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7657824

ABSTRACT

Although folate receptors (FRs) mediate folate uptake into cells, the independent role of FRs in cell proliferation remains unclear. We tested the hypothesis that transduction of FR cDNA in sense or antisense orientation using recombinant adeno-associated virus modulated FR expression and altered proliferation of cervical carcinoma cells (which constitutively overexpress FR genes). We determined that the integration of recombinant adeno-associated virions was not site specific. When compared with untransduced cells, sense and antisense FR cDNA-transduced cells exhibited an increase and decrease in FR mRNA and FR expression on the cell surface, respectively. However, when compared with antisense FR cDNA-transduced and untransduced cells, sense FR cDNA-transduced cells exhibited statistically significant (a) increased in total FRs, (b) smaller colonies, (c) lowered cell proliferation in vitro, and (d) less tumor volume with dramatic prolongation of tumor doubling times (225.6 h vs. 96 h) after transplantation into nude mice. Finally, (f) using single cell-derived transduced clones, an inverse relationship between cell proliferation and FR expression was established (r = 0.90, P < 0.001). Thus, transduction of sense/antisense FR cDNA into cervical carcinoma cells modulated expression of FRs and had an impact on cell proliferation in vitro and in vivo.


Subject(s)
Carrier Proteins/biosynthesis , Uterine Cervical Neoplasms/pathology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Division , DNA, Antisense , DNA, Complementary , Dependovirus , Female , Folate Receptors, GPI-Anchored , Genetic Vectors , HeLa Cells , Humans , KB Cells , Mice , Mice, Nude , Receptors, Cell Surface/biosynthesis , Regression Analysis , Transfection , Transplantation, Heterologous , Uterine Cervical Neoplasms/metabolism , Virion
13.
J Lab Clin Med ; 126(2): 184-203, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7636392

ABSTRACT

Folates play a vital role in cellular processes that are essential for fetal growth and viability. Thus the human placenta, which contains high-affinity membrane-associated placental folate receptors (PFRs), maintains a concentrative maternal-to-fetal flux of the vitamin under conditions of minimal dependence on variations of maternal dietary intake. To define transplacental folate transport and the role of PFRs in this mechanism, we utilized the isolated perfused human placental cotyledon. In closed system perfusions with 10 nmol/L 5-methyltetrahydrofolate, placental binding was rapid and extensive (47%), with a gradual maternal-to-fetal transfer of 5-methyltetrahydrofolate. Although hydrophilic PFRs were released into the fetal perfusate, PFR-bound folates constituted only a fraction of net transplacental folate transport. Transfer was bidirectional, not saturable, not inhibited by anion channel blockers, and dependent on perfusate levels. Placental binding far exceeded transfer, and pulsing the maternal circuit with tritiated 5-methyltetrahydrofolate, followed by washout of unbound radiolabel and rechallenge with unlabeled 5-methyltetrahydrofolate or folate, led to release of bound tritiated 5-methyltetrahydrofolate, illustrating reversible binding. Perfusion with the N-hydroxysuccinimide ester of folic acid eliminated essentially all 5-methyltetrahydrofolate binding to PFRs, while increasing net maternal-to-fetal transfer of the vitamin. Finally, because it has been suggested that impaired placental transport of folate may be linked to the fetotoxic effects of ethanol, the effect of this compound on the above processes was examined. An acute 6-hour exposure to ethanol (2.5 to 3.1 mg/ml) had no effect (p > 0.05) on net maternal-to-fetal transfer of 5-methyltetrahydrofolate. These studies suggest that net maternal-to-fetal transfer is a process consisting of two steps. First is the concentrative component in which circulating 5-methyltetrahydrofolate is bound to (captured by) PFRs on the maternally facing chorionic surface. Although kinetics favor binding, there is a dynamic state wherein a gradual release of 5-methyltetrahydrofolate from this pool can add to incoming circulating folates to generate an intervillous blood level approximately 3 times that in the maternal blood. In the second step, folates are passively transferred to the fetal circulation along a downhill concentration gradient. This unique mechanism for transplacental folate transport may be applicable to other small relative molecular mass ligand nutrients that bind to high-affinity placental receptors.


Subject(s)
Carrier Proteins/physiology , Fetus/metabolism , Maternal-Fetal Exchange/physiology , Placenta/metabolism , Tetrahydrofolates/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , 4-Acetamido-4'-isothiocyanatostilbene-2,2'-disulfonic Acid/pharmacology , Carrier Proteins/analysis , Carrier Proteins/metabolism , Ethanol/pharmacology , Female , Fetus/physiology , Folate Receptors, GPI-Anchored , Folic Acid/metabolism , Humans , Maternal-Fetal Exchange/drug effects , Perfusion/methods , Placenta/chemistry , Placenta/physiology , Pregnancy , Probenecid/pharmacology , Receptors, Cell Surface/analysis , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/physiology , Time Factors
14.
Genes Chromosomes Cancer ; 9(4): 234-43, 1994 Apr.
Article in English | MEDLINE | ID: mdl-7519045

ABSTRACT

The primary cytogenetic abnormality in acute promyelocytic leukemia (APL; FAB M3) is a reciprocal translocation, t(15;17)(q22;q12), which serves to fuse the PML gene on chromosome 15 to the retinoic acid receptor alpha (RARA) gene on chromosome 17. A PML-RARA fusion message transcribed from the der(15) is thought to mediate leukemogenesis. Two APL patients with simple variants of this translocation, t(3;15)(q21;q22) and t(X;15)(p11;q22), have previously been reported who lack cytogenetic involvement of chromosome 17, although their breakpoint positions on chromosome 15 still suggest the involvement of the PML gene. Here we report on a combined analysis by molecular genetics and in situ hybridization of these two patients, in which we wanted to determine whether the PML gene has alternative fusion partners or whether cryptic rearrangement of the RARA locus has occurred instead. A cryptic involvement of RARA was demonstrated in both patients by a combination of Southern analysis, reverse transcription coupled to PCR (RT-PCR), and fluorescence in situ hybridization. The results indicate an absolute requirement for the rearrangement of the RARA gene in the pathogenesis of APL and underline the importance of RARA during normal myeloid differentiation.


Subject(s)
Chromosomes, Human, Pair 15/ultrastructure , Chromosomes, Human, Pair 17/ultrastructure , DNA, Neoplasm/genetics , Leukemia, Promyelocytic, Acute/genetics , Neoplasm Proteins/genetics , Nuclear Proteins , Oncogene Proteins, Fusion/genetics , Receptors, Retinoic Acid/genetics , Transcription Factors/genetics , Translocation, Genetic , Adolescent , Adult , Aged , Child, Preschool , Chromosomes, Human, Pair 1/ultrastructure , Chromosomes, Human, Pair 11/ultrastructure , Chromosomes, Human, Pair 3/ultrastructure , Chromosomes, Human, Pair 8/ultrastructure , Female , Humans , In Situ Hybridization, Fluorescence , Male , Polymerase Chain Reaction , Promyelocytic Leukemia Protein , Retinoic Acid Receptor alpha , Tumor Suppressor Proteins , X Chromosome/ultrastructure
15.
Biochem J ; 298 ( Pt 1): 9-16, 1994 Feb 15.
Article in English | MEDLINE | ID: mdl-8129735

ABSTRACT

Existing methods of identifying the cleavage site of the nascent polypeptide and the C-terminal residue to which the glycosylphosphatidylinositol (GPI) anchor is attached in mature GPI-anchored proteins are technically difficult and labour-intensive. We tested the hypothesis that it was possible to predict this locus using data from the cDNA-deduced amino acid sequence and amino acid composition of GPI-anchored proteins. We employed a statistical approach which allowed repeated chi 2 comparisons between the proportions of residual amino acids in the major body of the cDNA-deduced polypeptide (minus the N-terminal signal peptide) after repeated computer-generated progressive exoproteolysis from its C-terminus one amino acid at a time and the fixed proportion of amino acids obtained from amino acid analysis of the mature GPI-anchored protein. Initial comparison of the two parameters invariably revealed a relatively high chi 2 statistic which progressively lowered to a minimum point at which the amino acid proportions of progressively exoproteolysed polypeptide and fixed endoproteolysed polypeptides of the mature GPI-anchored protein were in closest agreement. This objectively defined and unique minimum point of closest agreement accurately identified the locus of post-translational endoproteolytic cleavage of the nascent polypeptide in several tissue-specific single-gene-encoded GPI-anchored proteins. Thus the C-terminal amino acid to which the GPI anchor is attached can be rapidly identified using data from the cDNA sequence and the amino acid composition of proteins suspected to be GPI-anchored.


Subject(s)
Glycosylphosphatidylinositols/metabolism , Membrane Proteins/metabolism , Peptides/metabolism , Amino Acid Sequence , Animals , Binding Sites , Chi-Square Distribution , DNA, Complementary , Humans , Hydrolysis , Membrane Proteins/chemistry , Molecular Sequence Data , Peptides/chemistry
16.
Biochem J ; 286 ( Pt 3): 707-15, 1992 Sep 15.
Article in English | MEDLINE | ID: mdl-1417729

ABSTRACT

Human saliva contains a single 72,000-M(r) species which specifically reacted with rabbit anti-[human placental folate receptor (PFR)] serum on SDS/PAGE and Western blots. Although a specific radioimmunoassay for human PFR and related folate-binding proteins (FBPs) identified 55 ng of cross-reacting material (CRM) per mg of crude salivary proteins, only a minor fraction (1.6 ng) specifically bound radiolabelled folate. The major fraction of CRM did not contain bound endogenous folate and did not bind radiolabelled folates. On the basis of folate binding, salivary CRM species to PFR were designated as either functional (f-FBP) or non-functional (nf-FBP) species respectively. nf-FBPs and f-FBPs were isolated by different purification schemes. Both purified f-FBPs and nf-FBPs migrated as a single apparent 72,000-M(r) species on SDS/PAGE, but on Sephacryl S-200 gel filtration and sucrose-density-gradient ultracentrifugation they were eluted/sedimented with 40,000-M(r) markers. Each microgram of purified f-FBP and nf-FBP was measured in the radioimmunoassay for PFR as being equivalent to 18 ng and 24 ng of CRM respectively, indicating low epitope-relatedness to PFR. The Kd of f-FBPs was 50 pM and 0.94 mol of folate was bound/mol of protein. f-FBPs exhibited an unusual dependence on Triton X-100 for optimal ligand binding, despite the fact that Triton X-100 micelle binding was not demonstrated. The relative order of affinity of f-FBPs for pteroylglutamate greater than methotrexate greater than 5-formyltetrahydrofolate greater than 5-methyltetrahydrofolate was also distinct from that of purified PFR. Whereas amino acid and carbohydrate analysis revealed that nf-FBP (M(r) 51,400) and f-FBP (M(r) 39,200) were distinct glycoproteins with 8 and 13% carbohydrate respectively, isoelectric focusing and immunological studies suggested some structural identity. The presence of f-FBP and nf-FBP in normal saliva raises new questions about their possible role in vivo.


Subject(s)
Carrier Proteins/metabolism , Folic Acid/metabolism , Receptors, Cell Surface , Saliva/chemistry , Adult , Blotting, Western , Carrier Proteins/immunology , Carrier Proteins/isolation & purification , Chromatography, Gel , Chromatography, High Pressure Liquid , Cross Reactions , Electrophoresis, Polyacrylamide Gel , Folate Receptors, GPI-Anchored , Humans , Immune Sera , Radioimmunoassay
18.
J Biol Chem ; 267(6): 4119-27, 1992 Feb 25.
Article in English | MEDLINE | ID: mdl-1310989

ABSTRACT

Although normal human chorionic villi-associated hydrophobic placental folate receptors (PFR) are converted to hydrophilic forms by an endogenous, EDTA-sensitive, Mg(2+)-dependent protease under serum-free conditions (Verma, R. S., and Antony, A. C. (1991) J. Biol. Chem. 266, 12522-12535), it is not known whether hydrophobic PFR are also susceptible to conversion by endogenous phospholipases. We isolated and characterized hydrophobic PFR, and tested the hypothesis that purified, in situ, and de novo-synthesized native PFR were covalently linked to glycosyl-phosphatidylinositol (GPI) anchors. 125I-hydrophobic PFR, but not 125I-hydrophilic PFR, (i) separated into the Triton X-114 micellar phase at 30 degrees C, (ii) efficiently incorporated into phosphatidylcholine-cholesterol liposomes, and (iii) were covalently labeled by the hydrophobic probe 3-(trifluoromethyl)-3-(meta[125I]iodophenyl)diazirine, [125I]TID. (iv) [125I]TID-labeled- and [phenyl-3H]Triton X-100-bound hydrophobic PFR, as well as native PFR in situ, were released as hydrophilic forms by recombinant (r) GPI-specific phospholipase(PL) C (GPI-PLC), and GPI-PLD (but not by PLC), in the absence and presence of a concentration of EDTA known to inhibit endogenous Mg(2+)-dependent protease. (v) Nitrous acid deamination of [125I]TID-labeled hydrophobic PFR as well as (r)GPI-PLC cleavage of [phenyl-3H]Triton-X-100- and [125I] TID-labeled hydrophobic PFR, released hydrophobic radiolabeled moieties which comigrated on thin layer chromatography distinct from free radiolabel. Finally, (vi) biosynthetic studies on chorionic villi cultured in vitro revealed incorporation of radiolabeled precursors into the GPI anchor of hydrophobic PFR. We conclude that native hydrophobic PFR are linked to GPI anchors and are therefore potential substrates for three distinct endogenous enzymes (GPI-PLC, GPI-PLD, and specific Mg(2+)-dependent metalloprotease) in maternal serum and placenta in vivo.


Subject(s)
Carrier Proteins/metabolism , Folic Acid/metabolism , Glycolipids/metabolism , Phosphatidylinositols/metabolism , Placenta/metabolism , Receptors, Cell Surface/metabolism , Affinity Labels , Cell Membrane/metabolism , Chromatography, Gel , Chromatography, Thin Layer , Culture Media, Serum-Free , Electrophoresis, Polyacrylamide Gel , Female , Folate Receptors, GPI-Anchored , Glycosylphosphatidylinositols , Humans , Magnesium/metabolism , Phospholipases/metabolism , Photochemistry , Pregnancy , Protease Inhibitors/metabolism
19.
J Biol Chem ; 266(19): 12522-35, 1991 Jul 05.
Article in English | MEDLINE | ID: mdl-2061325

ABSTRACT

Full term placental chorionic villi cultured for 7 days in serum-free medium released hydrophilic folate-binding proteins (S-FBP(PCM) into the conditioned medium; in contrast, hydrophobic FBPs were the only form recovered from chorionic villi. Kinetic studies revealed that (i) S-FBP(PCM) was maximally released by the 3rd day, and this was associated with a proportionate decrease in hydrophobic FBPs; (ii) although cycloheximide inhibited de novo synthesis of [35S]methionine-labeled hydrophobic FBPs and S-FBP(PCM) by greater than 90%, unlabeled net S-FBP(PCM) release was only inhibited by 50%; (iii) EDTA markedly inhibited release of S-FBP(PCM) which was accompanied by a proportionate increase in hydrophobic FBPs; (iv) EDTA effects were completely reversed by 5-fold molar excesses of Mg2+ which led to a 50-fold greater release of S-FBP(PCM) compared to EDTA alone; (v) whereas Mg2+ alone stimulated S-FBP(PCM) release 4-fold greater than basal conditions, addition of cycloheximide to Mg2+ suppressed (by 4-fold) the expected increase observed with Mg2+ alone. Biochemical analyses of isolated S-FBP(PCM) revealed similarities to hydrophobic FBPs with respect to the ligand-binding domain and epitopes but differed in detergent-binding characteristics; furthermore, amino acid and carbohydrate analysis revealed a lower Mr = 25,500 with 12% carbohydrate. Based on kinetic analysis of S-FBP(PCM) release from chorionic villi-associated hydrophobic FBPs as well as structural analysis of S-FBP(PCM), these data continue to support the hypothesis that (a) a significant amount of maternal and probably fetal serum hydrophilic FBPs originate from placental hydrophobic FBPs, and (b) the endogenous hydrophobic FBP-directed Mg(2+)-dependent placental protease plays a major role in their release.


Subject(s)
Carrier Proteins/metabolism , Chorionic Villi/chemistry , Receptors, Cell Surface , Carrier Proteins/isolation & purification , Cells, Cultured , Chorionic Villi/drug effects , Cycloheximide/pharmacology , Electrophoresis, Polyacrylamide Gel , Female , Folate Receptors, GPI-Anchored , Humans , Isoelectric Focusing , Kinetics , Methionine/metabolism , Precipitin Tests
20.
J Clin Invest ; 87(1): 313-25, 1991 Jan.
Article in English | MEDLINE | ID: mdl-1702099

ABSTRACT

We tested the hypothesis that anti-placental folate receptor (PFR) antiserum-mediated effects on hematopoietic progenitor cells in vitro of increased cell proliferation and megaloblastic morphology were independent responses. We determined that (a) purified IgG from anti-PFR antiserum reacted with purified apo- and holo-PFR and specifically immunoprecipitated a single (44-kD) iodinated moiety on cell surfaces of low density mononuclear cells (LDMNC); (b) when retained in culture during in vitro hematopoiesis, anti-PFR IgG (in contrast to PFR-neutralized anti-PFR IgG and nonimmune IgG) consistently led to increased cloning efficiency of colony forming unit-erythroid (CFU-E), burst forming unit-E (BFU-E), CFU-granulocyte macrophage (CFU-GM), and CFU-GEM megakaryocyte (CFU-GEMM), and objectively defined megaloblastic changes in orthochromatic normoblasts from CFU-E- and BFU-E-derived colonies; (c) when anti-PFR antiserum was removed after initial (less than 1 h) incubation with LDMNC, a cell proliferation response was induced, but megaloblastic changes were not evident. (d) Conversely, delay at 4 degrees C for 24 h before long-term plating with antiserum resulted in megaloblastosis without increased cell proliferation; (e) however, 500-fold molar excess extracellular folate concentrations completely abrogated the expected anti-PFR antiserum-induced megaloblastic changes, without altering cell proliferative responses. Thus, although cell proliferative and megaloblastic changes are induced after short-term and prolonged interaction of antibody with folate receptors on hematopoietic progenitors, respectively, they are independent effects.


Subject(s)
Carrier Proteins/physiology , Folic Acid/metabolism , Hematopoiesis , Hematopoietic Stem Cells/physiology , Megaloblasts/physiology , Receptors, Cell Surface , Animals , Carrier Proteins/immunology , Cell Division , DNA/analysis , Folate Receptors, GPI-Anchored , Humans , Immune Sera/immunology , Immunoglobulin G/immunology , Macrophages/physiology , RNA/analysis , Rabbits , T-Lymphocytes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...