Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Anim Breed Genet ; 134(1): 78-84, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27339108

ABSTRACT

In the last decades, several endangered breeds of livestock species have been re-established effectively. However, the successful revival of the Dutch and Danish Landrace goats involved crossing with exotic breeds and the ancestry of the current populations is therefore not clear. We have generated genotypes for 27 FAO-recommended microsatellites of these landraces and three phenotypically similar Nordic-type landraces and compared these breeds with central European, Mediterranean and south-west Asian goats. We found decreasing levels of genetic diversity with increasing distance from the south-west Asian domestication site with a south-east-to-north-west cline that is clearly steeper than the Mediterranean east-to-west cline. In terms of genetic diversity, the Dutch Landrace comes next to the isolated Icelandic breed, which has an extremely low diversity. The Norwegian coastal goat and the Finnish and Icelandic landraces are clearly related. It appears that by a combination of mixed origin and a population bottleneck, the Dutch and Danish Land-races are separated from the other breeds. However, the current Dutch and Danish populations with the multicoloured and long-horned appearance effectively substitute for the original breed, illustrating that for conservation of cultural heritage, the phenotype of a breed is more relevant than pure ancestry and the genetic diversity of the original breed. More in general, we propose that for conservation, the retention of genetic diversity of an original breed and of the visual phenotype by which the breed is recognized and defined needs to be considered separately.


Subject(s)
Goats/classification , Goats/genetics , Microsatellite Repeats , Animals , Conservation of Natural Resources , Female , Male , Phylogeography
2.
Oncogene ; 30(5): 548-60, 2011 Feb 03.
Article in English | MEDLINE | ID: mdl-20856199

ABSTRACT

Hypoxia-elicited adaptations of tumor cells are essential for tumor growth and cancer progression. Although ample evidence exists for a positive correlation between hypoxia-inducible factors (HIFs) and tumor formation, metastasis and bad prognosis, the function of the HIF-α protein stability regulating prolyl-4-hydroxylase domain enzyme PHD2 in carcinogenesis is less well understood. In this study, we demonstrate that downregulation of PHD2 leads to increased tumor growth in a hormone-dependent mammary carcinoma mouse model. Tissue microarray analysis of PHD2 protein expression in 281 clinical samples of human breast cancer showed significantly shorter survival times of patients with low-level PHD2 tumors over a period of 10 years. An angiogenesis-related antibody array identified, amongst others, amphiregulin to be increased in the absence of PHD2 and normalized after PHD2 reconstitution. Cultivation of endothelial cells in conditioned media derived from PHD2-downregulated cells resulted in enhanced tube formation that was blocked by the addition of neutralizing anti-amphiregulin antibodies. Functionally, amphiregulin was regulated on the transcriptional level specifically by HIF-2 but not HIF-1. Our data suggest that PHD2/HIF-2/amphiregulin signaling has a critical role in the regulation of breast tumor progression and propose PHD2 as a potential tumor suppressor in breast cancer.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Breast Neoplasms/metabolism , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Procollagen-Proline Dioxygenase/metabolism , Amphiregulin , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Disease Progression , EGF Family of Proteins , Female , Gene Expression Regulation, Neoplastic , Glycoproteins/genetics , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases , Immunoblotting , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Kaplan-Meier Estimate , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred Strains , Mice, Nude , Middle Aged , Procollagen-Proline Dioxygenase/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
3.
Oncogene ; 28(37): 3274-85, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19581925

ABSTRACT

The onconeuronal cerebellar degeneration-related antigen Cdr2 is associated with paraneoplastic syndromes. Neoplastic expression of Cdr2 in ovary and breast tumors triggers an autoimmune response that suppresses tumor growth by developing tumor immunity, but culminates in cerebellar degeneration when Cdr2-specific immune cells recognize neuronal Cdr2. We identified Cdr2 as a novel interactor of the hypoxia-inducible factor (HIF) prolyl-4-hydroxylase PHD1 and provide evidence that Cdr2 might represent a novel important tumor antigen in renal cancer. Strong Cdr2 protein expression was observed in 54.2% of papillary renal cell carcinoma (pRCC) compared with 7.8% of clear-cell RCC and no staining was observed in chromophobe RCC or oncocytoma. High Cdr2 protein levels correlated with attenuated HIF target gene expression in these solid tumors, and Cdr2 overexpression in tumor cell lines reduced HIF-dependent transcriptional regulation. This effect was because of both attenuation of hypoxic protein accumulation and suppression of the transactivation activity of HIF-1alpha. pRCC is known for its tendency to avascularity, usually associated with a lower pathological stage and higher survival rates. We provide evidence that Cdr2 protein strongly accumulates in pRCC, attenuates the HIF response to tumor hypoxia and may become of diagnostic importance as novel renal tumor marker.


Subject(s)
Carcinoma, Renal Cell/genetics , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Hypoxia/genetics , Cell Line, Tumor , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/immunology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Nerve Tissue Proteins/immunology , Oxygen , Procollagen-Proline Dioxygenase/metabolism , Protein Binding , Transcription, Genetic , Transcriptional Activation , p300-CBP Transcription Factors/metabolism
4.
Article in English | MEDLINE | ID: mdl-11867282

ABSTRACT

Despite the fact that the use of chicken as immunization host brings many advantages to the production of polyclonal antibodies, the generation of egg yolk immunoglobulins (IgY) is rarely chosen. In this review, we report on the fast and efficient method for generation and affinity purification of IgY, in this case raised against the alpha-subunit of hypoxia-inducible factor-1 (HIF-1). The IgY antibody was successfully applied in a variety of methods and a number of different species for HIF-1alpha detection. In electrophoretic mobility shift assays, the IgY antibody recognized the native HIF-1 complex. The IgY antibody also detected HIF-1alpha protein on Western blots with extracts derived from human, monkey, pig, dog and mouse cell lines grown under hypoxic conditions. Immunofluorescence and immunoprecipitation experiments using the IgY antibody allowed detection and subcellular localization of HIF-1alpha in the nuclei of hypoxic cells. Chicken antibody production brings great benefit concerning the welfare of the immunized animals, due to non-invasive antibody harvesting with the added convenience of simple egg collection. An additional advantage is the fast and simple IgY isolation from egg yolk. IgY technology is a great improvement and should be considered as a good alternative to conventional polyclonal antibody production in mammals.


Subject(s)
Antibodies/immunology , Egg Yolk/immunology , Immunoglobulins/immunology , Animals , Chickens
5.
Pflugers Arch ; 443(2): 240-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11713650

ABSTRACT

Hypoxia-inducible factors (HIFs) locate to HIF-binding sites (HBSs) within the hypoxia-response elements (HREs) of oxygen-regulated genes. Whereas HIF-1alpha is expressed ubiquitously, HIF-2alpha is found primarily in the endothelium, similar to endothelin-1 (ET-1) and fms-like tyrosine kinase 1 (Flt-1), the expression of which is controlled by HREs. We identified an unique sequence alteration in both ET-1 and Flt-1 HBSs not found in other HIF-1 target genes, implying that these HBSs might cause binding of HIF-2 rather than HIF-1. However, electrophoretic mobility shift assays showed HIF-1 and HIF-2 DNA complex formation with the unique ET-1 HBS to be about equal. Both DNA-binding and hypoxic activation of reporter genes using the ET-1 HBS was decreased compared with transferrin and erythropoietin HBSs. The Flt-1 HBS was non-functional when assayed in isolation, suggesting that additional factors are required for hypoxic up-regulation via the reported Flt-1 HRE. Interestingly, HIF-1 activity could be restored fully by point-mutating the ET-1 (but not the Flt-1) HBS, suggesting that the wild-type ET-1 HBS attenuated the full hypoxic response known from other oxygen-regulated genes. Such a mechanism might serve to limit the expression of this potent vasoconstrictor in hypoxia.


Subject(s)
DNA-Binding Proteins/physiology , Endothelin-1/metabolism , Nuclear Proteins/physiology , Base Sequence/genetics , Basic Helix-Loop-Helix Transcription Factors , Binding Sites/genetics , Binding, Competitive , Cell Hypoxia/genetics , DNA-Binding Proteins/metabolism , Endothelin-1/genetics , Erythropoietin/metabolism , Gene Expression , Genes, Reporter , HeLa Cells , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Nuclear Proteins/metabolism , Protein Biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Response Elements/physiology , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , Vascular Endothelial Growth Factor Receptor-1
6.
Bioorg Med Chem Lett ; 11(12): 1549-52, 2001 Jun 18.
Article in English | MEDLINE | ID: mdl-11412978

ABSTRACT

Based on the inverse relationship between polar surface area and cell permeability and capitalizing on the properties of pyrrolopyrimidines 1 as protein tyrosine kinase inhibitors, pyrrolopyridones 2 were designed and synthesized as potential leads for the development of novel inhibitors with improved cell permeability properties.


Subject(s)
Cell Membrane Permeability , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyridones/pharmacokinetics , Animals , Biological Transport , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Humans , Pyridones/chemical synthesis , Static Electricity , Structure-Activity Relationship
8.
Biochim Biophys Acta ; 1493(1-2): 125-34, 2000 Sep 07.
Article in English | MEDLINE | ID: mdl-10978514

ABSTRACT

The heterodimeric hypoxia-inducible factor-1 (HIF-1), consisting of the subunits HIF-1alpha and HIF-1beta/ARNT, is a master transcriptional regulator of oxygen homeostasis. Under hypoxic conditions, HIF-1alpha levels very rapidly increase, mostly due to protein stabilization. However, translational regulation of HIF-1alpha has not been directly analyzed so far. Mouse HIF-1alpha exists as two mRNA isoforms (termed mHIF-1alphaI.1 and mHIF-1alphaI. 2) containing structurally different 5'-termini which might modulate translation initiation. Whereas the in vitro translation efficiency of these two mRNA isoforms was about equal, the mHIF-1alphaI.2 5'-untranslated region (5'-UTR) conferred significantly higher in vivo luciferase reporter gene activity than the mHIF-1alphaI.1 5'-UTR. Similar corresponding luciferase mRNA levels indicate translational rather than transcriptional alterations. Reporter gene expression was not affected upon exposure of transiently transfected cells to hypoxia (1% oxygen). Direct assessment of translational regulation by polysomal profile analysis of HeLaS3 cells showed that HIF-1alpha (and to a lower extent ARNT) mRNA was found mainly in the translationally active polyribosomal fractions under both normoxic and hypoxic conditions. In contrast, the association of mRNAs for beta-actin and ribosomal protein L28 with the polyribosomal fractions was substantially reduced under hypoxic conditions, suggesting decreased overall protein synthesis. Thus, efficient translation of mouse HIF-1alpha in a situation where the general translation efficiency is reduced represents a prerequisite for the very rapid accumulation of HIF-1alpha protein upon exposure to hypoxia.


Subject(s)
DNA-Binding Proteins/biosynthesis , Nuclear Proteins/biosynthesis , Receptors, Aryl Hydrocarbon , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator , Cell Hypoxia , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Dimerization , Exons , Gene Expression Regulation , Genes, Reporter , HeLa Cells , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Mice , Nuclear Proteins/genetics , Plasmids , Protein Biosynthesis , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , RNA, Messenger/biosynthesis , Transcription Factors/genetics , Transfection
9.
Blood ; 96(4): 1558-65, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10942406

ABSTRACT

Induction of erythropoietin (Epo) expression under hypoxic conditions is mediated by the heterodimeric hypoxia-inducible factor (HIF)-1. Following binding to the 3' hypoxia-response element (HRE) of the Epo gene, HIF-1 markedly enhances Epo transcription. To facilitate the search for HIF-1 (ant)agonists, a hypoxia-reporter cell line (termed HRCHO5) was constructed containing a stably integrated luciferase gene under the control of triplicated heterologous HREs. Among various agents tested, we identified a class of substances called epolones, which induced HRE-dependent reporter gene activity in HRCHO5 cells. Epolones are fungal products known to induce Epo expression in hepatoma cells. We found that epolones (optimal concentration 4-8 micromol/L) potently induce HIF-1 alpha protein accumulation and nuclear translocation as well as HIF-1 DNA binding and reporter gene transactivation. Interestingly, the activity of a compound related to the fungal epolones, ciclopirox olamine (CPX), was blocked after addition of ferrous iron. This suggests that CPX might interfere with the putative heme oxygen sensor, as has been proposed for the iron chelator deferoxamine mesylate (DFX). However, about 10-fold higher concentrations of DFX (50-100 micromol/L) than CPX were required to maximally induce reporter gene activity in HRCHO5 cells. Moreover, structural, functional, and spectrophotometric data imply a chelator:iron stoichiometry of 1:1 for DFX but 3:1 for CPX. Because the iron concentration in the cell culture medium was determined to be 16 micromol/L, DFX but not CPX function can be explained by complete chelation of medium iron. These results suggest that the lipophilic epolones might induce HIF-1 alpha by intracellular iron chelation. (Blood. 2000;96:1558-1565)


Subject(s)
DNA-Binding Proteins/genetics , Erythropoietin/genetics , Gene Expression Regulation/drug effects , Nuclear Proteins/genetics , Pyridones/pharmacology , Animals , CHO Cells , Cell Hypoxia , Cricetinae , DNA-Binding Proteins/metabolism , Erythropoietin/biosynthesis , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
10.
J Control Release ; 65(1-2): 245-51, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10699284

ABSTRACT

In recent years, major progress has been made in the design and synthesis of fibrinogen antagonists, which are peptidomimetic Arg-Gly-Asp (RGD) analogs. These RGD analogs are very promising antiplatelet agents. However, the clinical development of orally active RGD analogs has been hindered by the low oral bioavailability of many such RGD analogs. Aimed at enhancing their oral bioavailability, we have synthesized several coumarin-based cyclic prodrugs of RGD analogs, which have the two most polar functional groups, a carboxyl and an amino group, masked as an ester and an amide, respectively. As expected, these cyclic prodrugs have higher membrane interaction potentials as estimated by determining their partitioning between aqueous buffer and an immobilized artificial membrane than the corresponding RGD analogs. Consequently, these cyclic prodrugs are 5-6-fold more able to permeate monolayers of Caco-2 cells, an in vitro cell culture model of the intestinal mucosa barrier. Preliminary studies using dog also indicate the promising potential of using this coumarin-based prodrug strategy to improve the oral bioavailability of such RGD analogs.


Subject(s)
Anticoagulants/administration & dosage , Coumarins/administration & dosage , Mouth Mucosa/metabolism , Oligopeptides/pharmacokinetics , Prodrugs , Receptors, Immunologic/metabolism , Animals , Anticoagulants/pharmacokinetics , Anticoagulants/pharmacology , Biotransformation , Cell Line , Chemical Phenomena , Chemistry, Physical , Coumarins/pharmacokinetics , Coumarins/pharmacology , Dogs , Half-Life , Humans , Mouth Mucosa/cytology , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacology , Tirofiban , Tyrosine/analogs & derivatives , Tyrosine/pharmacokinetics , Tyrosine/pharmacology
11.
J Cell Sci ; 112 ( Pt 8): 1203-12, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10085255

ABSTRACT

Hypoxia-inducible factor-1 (HIF-1) is a master regulator of mammalian oxygen homeostasis. HIF-1 consists of two subunits, HIF-1alpha and the aryl hydrocarbon receptor nuclear translocator (ARNT). Whereas hypoxia prevents proteasomal degradation of HIF-1alpha, ARNT expression is thought to be oxygen-independent. We and others previously showed that ARNT is indispensable for HIF-1 DNA-binding and transactivation function. Here, we have used ARNT-mutant mouse hepatoma and embryonic stem cells to examine the requirement of ARNT for accumulation and nuclear translocation of HIF-1alpha in hypoxia. As shown by immunofluorescence, HIF-1alpha accumulation in the nucleus of hypoxic cells was independent of the presence of ARNT, suggesting that nuclear translocation is intrinsic to HIF-1alpha. Co-immunoprecipitation of HIF-1alpha together with ARNT could be performed in nuclear extracts but not in cytosolic fractions, implying that formation of the HIF-1 complex occurs in the nucleus. A proteasome inhibitor and a thiol-reducing agent could mimic hypoxia by inducing HIF-1alpha in the nucleus, indicating that escape from proteolytic degradation is sufficient for accumulation and nuclear translocation of HIF-1alpha. During biochemical separation, both HIF-1alpha and ARNT tend to leak from the nuclei in the absence of either subunit, suggesting that heterodimerization is required for stable association within the nuclear compartment. Nuclear stabilization of the heterodimer might also explain the hypoxically increased total cellular ARNT levels observed in some of the cell lines examined.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Receptors, Aryl Hydrocarbon , Transcription Factors/physiology , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator , Carcinoma, Hepatocellular/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/physiology , Electrophoresis, Polyacrylamide Gel , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Fluorescent Antibody Technique, Indirect , HeLa Cells , Humans , Hypoxia , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Immunoblotting , Mice , Nuclear Localization Signals/physiology , Nuclear Proteins/chemistry , Nuclear Proteins/physiology , Precipitin Tests , Stem Cells , Transcription Factors/metabolism , Tumor Cells, Cultured
12.
Chem Pharm Bull (Tokyo) ; 47(1): 90-5, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9987829

ABSTRACT

Earlier, we reported the development of a coumarin-based prodrug system that could be used for the preparation of cyclic prodrugs of opioid peptides. These cyclic prodrugs exhibited excellent membrane permeability characteristics. Therefore, it was of interest to determine the effects of this prodrug strategy on the membrane permeabilities of peptidomimetics which also have low membrane permeabilities. For this study, we have chosen two RGD (Arg-Gly-Asp) peptidomimetics, which have the potentials to be developed clinically as orally active antithrombotic agents. However, the clinical development of oral dosage forms of these RGD analogs has been hindered by their low intestinal mucosal permeability. Therefore, we have synthesized the corresponding coumarin-based cyclic prodrugs of these RGD peptidomimetics, which have the two most polar functional groups, a carboxyl and an amino group, masked as an ester and an amide, respectively. These cyclic prodrugs were shown to have higher membrane interaction potentials, as estimated by their partitioning between aqueous buffer and an immobilized artificial membrane, than the corresponding RGD analogs suggesting that they should exhibit good membrane permeation characteristics. Subsequently, in a separate study these cyclic prodrugs were shown to be 5 to 6-fold more able to permeate monolayers of Caco-2 cells, an in vitro cell culture model of the intestinal mucosa barrier, than the corresponding RGD peptidomimetics.


Subject(s)
Coumarins/chemical synthesis , Coumarins/pharmacokinetics , Esterases/metabolism , Oligopeptides/chemical synthesis , Oligopeptides/pharmacokinetics , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/pharmacokinetics , Platelet Glycoprotein GPIb-IX Complex , Platelet Membrane Glycoproteins , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Cell Membrane Permeability , Chemical Phenomena , Chemistry, Physical , Coumarins/pharmacology , Drug Stability , Humans , Membrane Potentials , Oligopeptides/pharmacology , Peptides, Cyclic/pharmacology , Prodrugs/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Sensitivity and Specificity , Structure-Activity Relationship
13.
FASEB J ; 13(2): 285-95, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9973316

ABSTRACT

Little is known about the factors influencing the hemoglobin switch in vertebrates during development. Inasmuch as the mammalian conceptus is exposed to changing oxygen tensions in utero, we examined the effect of different oxygen concentrations on beta-globin switching. We used an in vitro model of mouse embryogenesis based on the differentiation of blastocyst-derived embryonic stem cells to embryoid bodies (EBs). Cultivation of EBs at increasing oxygen concentrations (starting at 1% O2) did not influence the temporal expression pattern of embryonic (betaH1) globin compared to the normoxic controls (20% O2). In contrast, when compared to normoxically grown EBs, expression of fetal/adult (betamaj) globin in EBs cultured at varying oxygen concentrations was delayed by about 2 days and persisted throughout differentiation. Quantitation of hemoglobin in EBs using a 2,7-diaminofluorene-based colorimetric assay revealed the appearence of hemoglobin in two waves, an early and a late one. This observation was verified by spectrophotometric analysis of hemoglobin within single EBs. These two waves might reflect the switch of erythropoiesis from yolk sac to fetal liver. Reduced oxygenation is known to activate the hypoxia-inducible factor-1 (HIF-1), which in turn specifically induces expression of a variety of genes among them erythropoietin (EPO). Although EBs increased EPO expression upon hypoxic exposure, the altered beta-globin appearance was not related to EPO levels as determined in EBs overexpressing EPO. Since mRNA from both mouse HIF-1alpha isoforms was detected in all EBs tested at different differentiation stages, we propose that HIF-1 modulates beta-globin expression during development.


Subject(s)
Embryonic and Fetal Development/genetics , Gene Expression Regulation, Developmental , Globins/genetics , Oxygen/metabolism , Transcription Factors , Aerobiosis , Anaerobiosis , Animals , DNA-Binding Proteins/genetics , Embryonic and Fetal Development/drug effects , Gene Expression Regulation, Developmental/drug effects , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Mice , Nuclear Proteins/genetics , Oxygen/pharmacology
14.
FASEB J ; 13(1): 81-8, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9872932

ABSTRACT

Avian embryos and neonates acquire passive immunity by transferring maternal immunoglobulins from serum to egg yolk. Despite being a convenient source of antibodies, egg yolk immunoglobulins (IgY) from immunized hens have so far received scant attention in research. Here we report the generation and rapid isolation of IgY from the egg yolk of hens immunized against the alpha subunit of the human hypoxia-inducible factor 1 (HIF-1alpha). Anti-HIF-1alpha IgY antibodies were affinity purified and tested for their performance in various applications. Abundant HIF-1alpha protein was detected by Western blot analysis in nuclear extracts derived from hypoxic cells of human, mouse, monkey, swine, and dog origin whereas in hypoxic quail and frog cells, the HIF-1alpha signal was weak or absent, respectively. In electrophoretic mobility shift assays, affinity-purified IgY antibody was shown to recognize the native HIF-1 (but not the related HIF-2) complex that specifically binds an oligonucleotide containing the HIF-1 DNA binding site. Furthermore, IgY antibody immunoprecipitated HIF-1alpha from hypoxic cell extracts. Immunofluorescence experiments using IgY antibody allowed the detection of HIF-1alpha in the nucleus of hypoxic COS-7 cells. For comparison, the application of a mouse monoclonal antibody raised against the identical HIF-1alpha fragment was more restricted. Because chicken housing is inexpensive, egg collection is noninvasive, isolation and affinity purification of IgY antibodies are fast and simple, and the applicability of IgY is widespread, immunization of hens represents an excellent alternative for the generation of polyclonal antibodies.


Subject(s)
DNA-Binding Proteins/immunology , Egg Yolk/immunology , Immunoglobulins/immunology , Nuclear Proteins/immunology , Transcription Factors , Animals , COS Cells , Cell Line , Chickens , Dogs , Haplorhini , HeLa Cells , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Immunoglobulins/biosynthesis , Immunoglobulins/isolation & purification , Mice , Quail , Swine , Tumor Cells, Cultured , Xenopus laevis
15.
Cancer Res ; 58(24): 5678-80, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9865721

ABSTRACT

Oxygen-deprived regions of a solid tumor can induce tumor suppressor p53 expression and hence select for p53-mutant tumor cells with diminished apoptotic potential. It has been proposed that the hypoxia-inducible factor-1 (HIF-1) alpha subunit binds to p53 and protects it from proteasomal degradation. However, we found that hypoxic conditions that strongly induce HIF-1-dependent endogenous gene expression as well as HIF-1alpha protein neither induce p53-dependent gene expression nor p53 protein. The iron chelator deferoxamine induced both HIF-1alpha and p53, but p53 up-regulation could still be detected in HIF-1alpha-deficient cells, suggesting that mechanisms other than HIF-1alpha activation contribute to oxygen-regulated p53 induction.


Subject(s)
Cell Hypoxia , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Genes, p53 , Nuclear Proteins/metabolism , Receptors, Aryl Hydrocarbon , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator , Deferoxamine/pharmacology , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Mice , Transcription Factors/metabolism , Tumor Cells, Cultured , Up-Regulation
16.
Eur J Pharm Sci ; 6(4): 317-24, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9795088

ABSTRACT

A recently developed, new theoretical absorption model for passive diffusion through biological membranes describing the dependency of membrane permeability on lipophilicity and molecular size, predicts different sigmoid-hyperbolic permeability-lipophilicity relationships for different molecular weight ranges. This model has been tested with experimental in vitro cultured epithelial cell (Caco-2) permeability data for structurally diverse drugs differing in lipophilicity, ionization state and molecular size. These data were pooled with literature values. Using this simple physicochemical approach, the permeability of a compound through Caco-2 cells by passive diffusion can be predicted from the compounds' distribution coefficient in 1-octanol/water (log D(oct)) and its molecular weight (MW). Deviations from this expected behaviour may point to the involvement of biological components in the transport process, which may require further investigations.


Subject(s)
Cell Membrane Permeability/drug effects , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , ATP Binding Cassette Transporter, Subfamily B/chemistry , Algorithms , Caco-2 Cells , Chemical Phenomena , Chemistry, Physical , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Diffusion , Humans , Lipids/chemistry , Molecular Weight , Structure-Activity Relationship
17.
Eur J Pharm Sci ; 6(4): 325-29, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9795090

ABSTRACT

The objective of this study was to rationalize the shape of membrane permeability-lipophilicity curves, when considering, in addition to the usual transcellular route, a parallel diffusion pathway through aqueous pores as present in biological membranes. The theoretical influence of different pH in donor and acceptor compartment and the molecular weight on the permeability curves was studied. We combined and extended two previously proposed absorption models, namely one describing diffusion through a simple membrane (two stagnant aqueous and two organic layers in series, no pores) as the sum of the two distribution steps at both membrane interfaces, and a second theoretical model considering the sum of different diffusional resistances through stagnant layers and membrane, respectively. Under certain conditions the equivalence of the two-step distribution model and the diffusional resistance model can be demonstrated. Incorporation of an aqueous diffusion pathway leads to an extended two-step distribution model. This theoretical membrane permeation model will permit a more physicochemical-based interpretation of permeation data and shows that combined log D values and molecular weight are important determinants for membrane transport processes through, e.g. Caco-2 monolayers and the mucosal GI membranes. We have demonstrated that the well-known sigmoidal permeability-lipophilicity relationship should be considered as a molecular weight-dependent set of sigmoidal relationships.


Subject(s)
Cell Membrane Permeability , Lipids/chemistry , Pharmaceutical Preparations/chemistry , Algorithms , Chemical Phenomena , Chemistry, Physical , Diffusion , Hydrogen-Ion Concentration , Models, Theoretical , Molecular Weight , Pharmacokinetics , Porosity , Water/metabolism
18.
Pharm Res ; 15(8): 1174-81, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9706046

ABSTRACT

PURPOSE: To compare the bioconversion rates in various biological media and the Caco-2 cell permeation characteristics of coumarin based cyclic prodrugs (3a, 3b) and methylester-based linear prodrugs (1b, 2b) of two RGD peptidomimetics (1a, 2a). METHODS: Bioconversion rates of the prodrugs to the RGD peptidomimetics were determined in Hank balances salt solution (HBSS), pH 7,4, at 37 degrees C and in various biological media (human blood plasma, rat liver homogenate, Caco-2 cell homogenate) known to have esterase activity. Transport rates of the prodrugs and the RGD peptidomimetics were determined using Caco-2 cell monolayers, an in vitro cell culture model of the intestinal mucosa. RESULTS. In HBSS, pH 7,4, the coumarin-based cyclic prodrugs 3a and 3b degraded slowly and quantitatively to the RGD peptidomimetics 1a and 2a, respectively (3a, t1/2 = 630+2-14 min; 3b, t1/2 = 301 +/-12 min). The methylester-based linear prodrugs 1b and 2b were more stable to chemical hydrolysis (1b and 2b, t1/2 > 2000 min). Both the coumarin-based cyclic prodrugs and the methylester-based linear prodrugs degraded more rapidly in biological media containing esterase activity (e.g., 90% human blood plasma: 1b, t1/2 < 5 min; 2b, t1/2 < 5 min; 3a, t1/2 < 91+/-1 min; 3b, 1/2 < 57+/-2 min). When the apical (AP)-to-basolateral (BL) permeation characteristics were determined using Caco-2 cell monolayers, it was found that the methylester prodrugs 1b and 2b underwent esterase bioconversion (>80%) to the RGD peptidomimetics 1a and 2a, respectively, In contrast, the cyclic prodrugs 3a and 3b permeated the cell monolayers intact. Considering the appearance of both the prodrug and the RGD peptidomimetic on the BL side, the methylester prodrugs 1b and 2b were approximately 12-fold more able to permeate than were the RGD peptidomimetics 1a and 2a. When similar analysis of the transport data for the coumarin prodrugs 3a and 3b was performed, they were shown to be approximately 6-fold and 5-fold more able to permeate than were the RGD peptidomimetics 1a and 12a, respectively. CONCLUSION: The coumarin-based cyclic prodrugs 3a and 3b were chemically less stable, but metabolically more stable, then the methylester based linear prodrugs. The esterase stability of the cyclic prodrugs 3a and 3b means that they are transported intact across the Caco-2 cell monolayer in contrast to the methylester prodrugs 1b and 2b, which undergo facile bioconversion during their transport to the RGD peptidomimetics. However, both prodrug systems successfully delivered more (5-12-fold) of the RGD peptidomimetic and/or the precursor (prodrug) than did the RGD peptidomimetics themselves.


Subject(s)
Coumarins/pharmacokinetics , Oligopeptides/pharmacokinetics , Prodrugs/pharmacokinetics , Animals , Caco-2 Cells , Coumarins/chemistry , Drug Stability , Esters , Humans , Male , Molecular Mimicry , Oligopeptides/chemistry , Permeability , Rats , Rats, Sprague-Dawley
19.
Eur J Biochem ; 253(3): 771-7, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9654078

ABSTRACT

The hypoxia-inducible factor-1 (HIF-1) is a transcriptional activator involved in the expression of oxygen-regulated genes such as that for erythropoietin. Following exposure to low oxygen partial pressure (hypoxia), HIF-1 binds to an hypoxia-response element located 3' to the erythropoietin gene and confers activation of erythropoietin expression. The conserved core HIF-1 binding site (HBS) of the erythropoietin 3' enhancer (CGTG) contains a CpG dinucleotide known to be a potential target of cytosine methylation. We found that methylation of the HBS abolishes HIF-1 DNA binding as well as hypoxic reporter gene activation, suggesting that a methylation-free HBS is mandatory for HIF-1 function. The in vivo methylation pattern of the erythropoietin 3' HBS in various human cell lines and mouse organs was assessed by genomic Southern blotting using a methylation-sensitive restriction enzyme. Whereas this site was essentially methylation-free in the erythropoietin-producing cell line Hep3B, a direct correlation between erythropoietin protein expression and the degree of erythropoietin 3' HBS methylation was found in different HepG2 sublines. However, the finding that this site is partially methylation-free in human cell lines and mouse tissues that do not express erythropoietin suggests that there might be a general selective pressure to keep this site methylation-free, independent of erythropoietin expression.


Subject(s)
Cell Hypoxia , DNA-Binding Proteins/metabolism , Dinucleoside Phosphates/metabolism , Erythropoietin/biosynthesis , Gene Expression Regulation , Nuclear Proteins/metabolism , Animals , Binding Sites , Carcinoma, Hepatocellular , Cell Nucleus/metabolism , DNA Methylation , Gene Expression Regulation, Neoplastic , Genes, Reporter , HeLa Cells , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Kidney/embryology , Kidney/growth & development , Kidney/metabolism , L Cells , Leukemia , Liver/embryology , Liver/growth & development , Liver/metabolism , Liver Neoplasms , Luciferases/biosynthesis , Mice , Neuroblastoma , Organ Specificity , Recombinant Fusion Proteins/biosynthesis , Transcription Factors/metabolism , Transcriptional Activation , Transfection , Tumor Cells, Cultured
20.
J Drug Target ; 6(2): 151-65, 1998.
Article in English | MEDLINE | ID: mdl-9886238

ABSTRACT

The influence of physicochemical properties, including lipophilicity, H-bonding capacity and molecular size and shape descriptors on brain uptake has been investigated using a selection of marketed CNS and CNS-inactive drugs. It is demonstrated that the polar surface area of a drug can be used as a suitable descriptor for the drugs' H-bonding potential. A combination of a H-bonding and a molecular size descriptor, i.e., the major components of lipophilicity and permeability, avoiding knowledge of distribution coefficients, is proposed to estimate brain penetration potential of new drug candidates. Previously reported experimental surface activity data appear to be strongly correlated to molecular size of the drug compounds. Present analysis offers a modern basis for property-based design and targeting of CNS drugs.


Subject(s)
Blood-Brain Barrier/physiology , Drug Delivery Systems/methods , Hydrogen Bonding , Molecular Structure , Pharmacokinetics , Humans , Molecular Weight , Solubility , Statistics as Topic
SELECTION OF CITATIONS
SEARCH DETAIL
...