Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Int J Hyperthermia ; 39(1): 48-56, 2022.
Article in English | MEDLINE | ID: mdl-34937522

ABSTRACT

Aim: In oncology, thermal therapy is the application of external heat to fight cancer cells. The goal of whole-body thermal treatment (WBTT) is to raise the patient's core temperature to 39-42 °C, and represents the only thermal treatment modality that can act on both the primary tumor and distant metastases. However, WBTT carries potential risks for toxicity when applied without accurate thermometry and monitoring.Methods: ElmediX has developed a medical device, HyperTherm, to deliver long-term controlled and accurate WBTT (41.5 °C, up to 8 h). The safety of the device and thermal treatment protocol was initially evaluated in minipigs, and we present the confirmation of tolerability of WBTT in dogs with advanced cancer, in combination with a reduced dose of radiotherapy or chemotherapy.Results: Thermometry in liver, rectum, and tumor confirmed a homogeneous heating of these body parts. Monitoring of clinical parameters showed acceptable and reversible changes in liver, cardiac, muscle and coagulation parameters, as was expected. Combination of WBTT with both radiotherapy and chemotherapy only caused some low-grade adverse events.Conclusion: We conclude that our findings support the safe use of HyperTherm-mediated WBTT for canine patients with advanced malignancies. They also tend to support a genuine therapeutic potential for long-term WBTT which needs to be confirmed on a larger dog patient population. Combined with previously reported safety results in minipigs, these contribute to support the ongoing clinical evaluation of WBTT in advanced human cancer patients.


Subject(s)
Hyperthermia, Induced , Neoplasms , Animals , Combined Modality Therapy , Dogs , Human Body , Humans , Hyperthermia, Induced/methods , Neoplasms/radiotherapy , Swine , Swine, Miniature , Temperature
2.
Ann Oncol ; 28(9): 2278-2285, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28911087

ABSTRACT

BACKGROUND: The transforming growth factor (TGF)-ß pathway is a well-described inducer of immunosuppression and can act as an oncogenic factor in advanced tumors. Several preclinical and clinical studies show that the TGF-ß pathway can be considered a promising molecular target for cancer therapy. The human genome has three TGF-ß isoforms and not much is known about the oncogenic response to each of the isoforms. Here, we studied the antitumor response to ISTH0047, a recently developed locked nucleic acid-modified antisense oligonucleotide targeting TGF-ß2. MATERIALS AND METHODS: We have studied the anticancer response to ISTH0047 using gymnotic delivery in tumor cell cultures and in in vivo preclinical orthotopic mouse models for primary tumors (breast and kidney tumors) and lung metastasis. RESULTS: We observed that ISTH0047 is able to significantly reduce TGF-ß2 mRNA and protein levels without altering the levels of TGF-ß1 and TGF-ß3. ISTH0047 prevented lung metastasis in syngeneic orthotopic renal cell carcinoma (RENCA) and breast cancer (4T1) tumor models. In addition, using an orthotopic xenograft model of a lung cancer cell line (CRL5807) that mainly expresses TGF-ß2, we observed that ISTH0047 had an important effect on the lung microenvironment inhibiting the growth of lung lesions. ISTH0047 treatment re-educated macrophages in the lung parenchyma to express the tumor-suppressive factor, CD86. CONCLUSION: Overall, our data point to TGF-ß2 as a therapeutic target and ISTH0047 as a novel anticancer drug to prevent lung metastasis by impacting on the tumor niche, in part, through the induction of CD86 in tumor-associated macrophages.


Subject(s)
B7-2 Antigen/immunology , Breast Neoplasms/pathology , Kidney Neoplasms/pathology , Lung Neoplasms/secondary , Macrophages/immunology , Oligonucleotides, Antisense/genetics , Transforming Growth Factor beta2/genetics , Animals , Enzyme-Linked Immunosorbent Assay , Female , Humans , Lung/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
3.
Pharmazie ; 63(3): 221-5, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18444511

ABSTRACT

The objective of the current study was to develop cellular delivery approaches for catalytic DNA enzymes (DNAzymes) which cleave targeted messenger RNA, using vectors based on colloidal gold. The model DNAzyme was a 32mer oligonucleotide designed to specifically interact with and cleave c-myc mRNA. Colloidal gold particles were prepared by reduction of tetrachlororauric [III] acid with sodium citrate. Particles could be produced in the 1-90 nm range. A cationic substrate linked to transferrin was electrostatically/hydrophobically bound to the gold particle. These vectors were then treated with the DNAzyme to yield the condensed DNA-cationic polymer-particulate product. The pH (4-11.5), the quantity of the DNAzymes (0.079-0.567 microg/probe), the cationic polymer (polylysine (PL) or polyethylenimine (PEI)) as well as the surfactant (PVP) concentration (0-0.5%) were varied to give stable constructs which decomplexed under the desired conditions (i.e., in lysosomes and at lower pH values). Cellular uptake of the FITC-labelled c-myc DNAzyme incorporated in this vector was measured using FACS analysis in human HT29 colon carcinoma cells. Data suggested that PEI gave better delivery efficiencies than PL. The use of PVP to stabilize the formed dispersions was detrimental to DNAzyme delivery when PL was used but had little effect in the PEI systems. In the best cases, delivery to 77% of the cells was possible using PEI with the PVP stabilizer and completing the DNA condensation at pH 5.5 with 0.118 microg of DNAzyme/probe. In contrast, the best conditions for PL gave only transfection to 43% of the cells (no PVP, condensed at pH 5.7 and with a loading of 0.079 microg DNAzyme/probe). The PL probe tended to be more toxic than the PEI-based systems (65% cell death in PL transfected cells compared to 22% for PEI). These results suggest that cellular targeting using colloidal gold appears feasible for DNAzyme delivery.


Subject(s)
DNA, Catalytic/administration & dosage , DNA, Catalytic/pharmacology , Gold Colloid/administration & dosage , Nanoparticles/administration & dosage , Proto-Oncogene Proteins c-myc/drug effects , Drug Carriers , Drug Delivery Systems , Drug Stability , Electrophoresis, Polyacrylamide Gel , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Gold Colloid/chemistry , HT29 Cells , Humans , Hydrogen-Ion Concentration , Nanoparticles/chemistry , Transfection
4.
Oncogene ; 25(50): 6672-7, 2006 Oct 26.
Article in English | MEDLINE | ID: mdl-16732328

ABSTRACT

The hdm-2 oncogene is overexpressed in several types of malignancies including osteosarcomas, soft tissue sarcomas and gliomas and hdm-2 has been associated with accelerated tumor formation in both hereditary and sporadic cancers. Among the other key binding partners, hdm-2 forms a complex with the tumor suppressor p53, resulting in a rapid proteasome-mediated degradation of the p53 protein. This positions the hdm-2-p53 complex as an attractive target for the development of anticancer therapy and recently the first small molecule hdm-2 antagonist has been reported. Development of hdm-2 antagonists is currently focused on malignancies containing a wild-type p53 genotype, which is the case in approximately half of human cancer indications. However, hdm-2 has also been implicated in oncogenesis in the absence of p53. We therefore studied the effect of hdm-2 antagonists in p53-deficient human H1299 lung carcinoma cells. The hdm-2 antagonistic peptide caused G1 cell cycle arrest, inhibited colony growth and induced expression of G1 checkpoint regulatory proteins, such as p21(waf1,cip1). These data demonstrate that hdm-2 regulates the G1 cell cycle checkpoint in a p53-independent manner, suggesting that hdm-2 antagonists represent a novel class of anticancer therapeutics with broad applicability towards tumors with different p53 genetic backgrounds.


Subject(s)
Cell Cycle/drug effects , Lung Neoplasms/genetics , Peptides/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Carcinoma/genetics , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Green Fluorescent Proteins/metabolism , Humans , Imidazoles/pharmacology , Peptides/metabolism , Peptides/therapeutic use , Piperazines/pharmacology , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured
5.
Cancer Res ; 59(20): 5239-44, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10537303

ABSTRACT

The expression of activated ras genes has been implicated as a contributing factor to the radioresistance of tumor cells. As a strategy for compromising Ras protein activity and potentially enhancing the radiosensitivity of tumor cells, we have investigated the application of the AV1Y28 adenovirus, which expresses a single-chain antibody fragment directed against p21 Ras proteins. The ability of AV1Y28 transduction to modulate radioresponse was investigated using four human tumor cell lines--U251 glioblastoma, MIA PaCa-2 pancreatic carcinoma, and the colon carcinomas SW620 and HT29. Cultures were exposed to sufficient levels of AV1Y28 to transduce more than 90% of the cells; 24 h later, cultures were exposed to ionizing radiation, and clonogenic cell survival was determined. Tumor cell survival was reduced by 40-50% when the tumor cell lines were exposed to AV1Y28 only. In addition, for each tumor cell line, AV1Y28 exposure enhanced the level of radiation-induced cell killing. Dose enhancement factors at a surviving fraction of 0.1 ranged from 1.3 to 1.5. Furthermore, for each of the cell lines, the surviving fraction at 2 Gy was significantly reduced by AV1Y28 exposure. In contrast to the results seen in tumor cells, the radiosensitivity of a normal human fibroblast cell line was not affected by AV1Y28. These data indicate that this anti-Ras adenovirus enhances the radiosensitivity of tumor cells but does not affect the radiosensitivity of normal cells.


Subject(s)
Adenoviridae/genetics , Immunoglobulin Fragments/therapeutic use , Neoplasms/radiotherapy , Radiation Tolerance , ras Proteins/antagonists & inhibitors , Apoptosis/radiation effects , Cell Cycle/radiation effects , Cell Survival/radiation effects , Fibroblasts/radiation effects , Humans , Immunoglobulin Fragments/genetics , Neoplasms/genetics , Neoplasms/pathology , Recombinant Proteins/therapeutic use , Tumor Cells, Cultured
6.
Diabetes Care ; 13(6): 565-75, 1990 Jun.
Article in English | MEDLINE | ID: mdl-2162754

ABSTRACT

We identified the earliest events in autophosphorylation of the insulin receptor after insulin addition. Insulin-stimulated autophosphorylation at specific sites in the tyrosine kinase domain of the receptor's beta-subunit is correlated kinetically with activation of kinase-catalyzed phosphorylation of a model substrate (reduced and carboxyamidomethylated lysozyme; RCAM-lysozyme). To identify these sites, the deduced amino acid sequence of the 3T3-L1 adipocyte insulin receptor of the mouse was determined. Insulin-induced activation of substrate phosphorylation was shown to require autophosphorylation of three neighboring tyrosines (Tyr1148, Tyr1152, and Tyr1153) in the mouse receptor. A search for cellular substrates of the receptor kinase revealed that insulin causes accumulation of a 15,000-Mr phosphorylated (on tyrosine) cytosolic protein (pp15) in 3T3-L1 adipocytes treated with oxophenylarsine (PAO). PAO blocks turnover of the phosphoryl group of pp15, causing its accumulation, and thereby appears to interrupt signal transmission from the receptor to the glucose-transport system. Two membrane-bound protein phosphotyrosine phosphatases that are inhibited by PAO and are apparently responsible for the turnover of the pp15 phosphoryl group have been purified from 3T3-L1 adipocytes and characterized. These and other results support the hypothesis that turnover of the phosphoryl group of pp15, a product of insulin-receptor tyrosine kinase action, couples signal transmission to the glucose-transport system. [32P]pp15 was purified to homogeneity from 3T3-L1 adipocytes. Amino acid and radiochemical sequence analysis of the purified tryptic [32P]phosphopeptide revealed that pp15 is the phosphorylation product of 422(aP2) protein, a 15,000-Mr adipocyte protein whose cDNA we previously cloned and sequenced. 422(aP2) protein was found to bind fatty acids. When exposed to a free fatty acid, notably oleic acid, 422(aP2) protein becomes an excellent substrate of the isolated insulin-receptor tyrosine kinase. Compelling evidence indicates that on binding fatty acid, 422(aP2) protein undergoes a conformational change whereby Tyr19 becomes accessible to the receptor tyrosine kinase and undergoes O-phosphorylation. Adipose tissue and skeletal and heart muscle, which exhibit insulin-stimulated glucose uptake, express a specific insulin-responsive glucose transporter. A cDNA (GT2) that encodes this protein was isolated from a mouse 3T3-L1 adipocyte library and sequenced. We also isolated and characterized the corresponding mouse gene GLUT4. DNase I footprinting with nuclear extracts from 3T3-L1 cells revealed that a differentiation-specific nuclear factor binds to the GLUT4 promoter. The purified transcription factor C/EBP binds at the same position.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Glucose/metabolism , Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Biological Transport, Active , Genes , Mice , Molecular Sequence Data , Monosaccharide Transport Proteins/genetics , Phosphorylation , Receptor, Insulin/metabolism , Signal Transduction
7.
Mol Cell Endocrinol ; 72(3): 175-85, 1990 Sep 10.
Article in English | MEDLINE | ID: mdl-2289630

ABSTRACT

The degradation products generated from A14 and B26 125I-labelled insulins in liver endosomes in vivo and in vitro have been isolated by high-performance liquid chromatography and cleavages in the B chain have been identified by automated radiosequence analysis. In rats sacrificed various times after injection of each of the 125I-labelled insulins, two major degradation products slightly less hydrophobic than intact iodoinsulins were identified; these accounted, at 8 min. for about 45% (A14 125I-labelled insulin) and 15% (B26 125I-labelled insulin) of the total radioactivity recovered, respectively. The products generated from A14 125I-labelled insulin contained an intact A chain, whereas those generated from B26 125I-labelled insulin contained a B chain cleaved at the B16-B17 bond. With B26 125I-labelled insulin, two minor products, with cleavages at the B23-B24 and B24-B25 bonds, were also observed. In vivo chloroquine treatment did not alter the nature but caused a decrease in the amount of insulin degradation products associated with endosomes. When endosomal fractions isolated from iodoinsulin injected rats were incubated at 30 degrees C in isotonic KCl, a rapid degradation of iodoinsulin, maximal at pH 6, was observed. With A14 125I-labelled insulin, the two major degradation products identified in vivo were generated along with monoiodotyrosine, but with B26 125I-labelled insulin monoiodotyrosine was the main product formed. Addition of ATP, presumably by decreasing the endosomal pH, shifted the medium pH for maximal iodoinsulin degradation to about 7-8. These studies have allowed a direct identification of two previously suggested cleavage sites in the B chain. They have also shown that the degradation products generated in cell-free endosomes under conditions that promote endosomal acidification are similar to those identified in vivo.


Subject(s)
Insulin/metabolism , Liver/metabolism , Animals , Chloroquine/pharmacology , Chromatography, High Pressure Liquid , Golgi Apparatus/metabolism , Insulysin/metabolism , Liver/drug effects , Male , Peptide Fragments/analysis , Rats , Rats, Inbred Strains , Subcellular Fractions/metabolism
8.
Drugs Exp Clin Res ; 30(3): 89-98, 2004.
Article in English | MEDLINE | ID: mdl-15366785

ABSTRACT

The p21(Waf1/Cip1) protein represents a broad-acting cyclin-dependent kinase inhibitor that plays a key role in cell cycle regulation. Furthermore, p21(Waf1/Cip1) protein has been described as a direct participant in regulating genes involved in growth arrest, senescence and aging. In response to genotoxic insults (e.g., following chemotherapeutic treatment), p21(Waf1/Cip1) protein accumulates mainly through p53-mediated transcriptional activation and is also regulated at the post-transcriptional level. In tumor cells, p53 is frequently mutated leading to reduced p21(Waf1/Cip1) protein induction that may contribute to resistance to treatment by DNA-damaging agents. In order to identify compounds capable of restoring p21(Waf1/Cip1) protein level, we have developed a 96-multi-well plate-based high throughput screening assay in intact cells using the Applied Biosystems Fluorometric Microvolume Assay Technology (FMAT) macro-confocal system. Briefly, following incubation with test compounds, human MCF7 breast carcinoma cells were fixed and p21(Waf1/Cip1) protein was detected using anti-p21(Waf1/Cip1) monoclonal antibody and anti-mouse IgG conjugated to the red fluorescent dye Alexafluor 633. FMAT provides a set of raw images at a high magnification, in which fluorescence concentrated in a cell is detected as a specific signal. The mean fluorescence of a population of cells is calculated independently of the number of cells as with a classical FACS analysis. This is of particular interest for screening anticancer drugs that may affect cell number and therefore may impact on the readout. This assay was validated using reference compounds such as camptothecin and actinomycin D, known inducers of p21(Waf1/Cip1) protein.


Subject(s)
Cell Cycle Proteins/analysis , Cell Cycle Proteins/genetics , Technology, Pharmaceutical/methods , Technology, Pharmaceutical/trends , Tumor Cells, Cultured , Belgium , Camptothecin/metabolism , Camptothecin/pharmacology , Cell Cycle Proteins/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Dactinomycin/metabolism , Dactinomycin/pharmacology , Fluorometry/instrumentation , Fluorometry/methods , Gene Expression/drug effects , Gene Expression/genetics , Humans , Microchemistry/instrumentation , Microchemistry/methods , Reproducibility of Results , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
12.
Br J Cancer ; 97(10): 1344-53, 2007 Nov 19.
Article in English | MEDLINE | ID: mdl-18000499

ABSTRACT

R306465 is a novel hydroxamate-based histone deacetylase (HDAC) inhibitor with broad-spectrum antitumour activity against solid and haematological malignancies in preclinical models. R306465 was found to be a potent inhibitor of HDAC1 and -8 (class I) in vitro. It rapidly induced histone 3 (H3) acetylation and strongly upregulated expression of p21waf1,cip1, a downstream component of HDAC1 signalling, in A2780 ovarian carcinoma cells. R306465 showed class I HDAC isotype selectivity as evidenced by poor inhibition of HDAC6 (class IIb) confirmed by the absence of downregulation of Hsp90 chaperone c-raf protein expression and tubulin acetylation. This distinguished it from other HDAC inhibitors currently in clinical development that were either more potent towards HDAC6 (e.g. vorinostat) or had a broader HDAC inhibition spectrum (e.g. panobinostat). R306465 potently inhibited cell proliferation of all main solid tumour indications, including ovarian, lung, colon, breast and prostate cancer cell lines, with IC50 values ranging from 30 to 300 nM. Haematological cell lines, including acute lymphoblastic leukaemia, acute myeloid leukaemia, chronic lymphoblastic leukaemia, chronic myeloid leukaemia, lymphoma and myeloma, were potently inhibited at a similar concentration range. R306465 induced apoptosis and inhibited angiogenesis in cell-based assays and had potent oral in vivo antitumoral activity in xenograft models. Once-daily oral administration of R306465 at well-tolerated doses inhibited the growth of A2780 ovarian, H460 lung and HCT116 colon carcinomas in immunodeficient mice. The high activity of R306465 in cell-based assays and in vivo after oral administration makes R306465 a promising novel antitumoral agent with potential applicability in a broad spectrum of human malignancies.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Neoplasms/drug therapy , Sulfones/pharmacology , Administration, Oral , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Histones/drug effects , Histones/metabolism , Humans , Hydroxamic Acids/chemistry , Immunohistochemistry , Isoenzymes/antagonists & inhibitors , Male , Mice , Mice, Nude , Neoplasms/pathology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Rats , Rats, Sprague-Dawley , Sulfones/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Eur J Biochem ; 163(2): 433-42, 1987 Mar 02.
Article in English | MEDLINE | ID: mdl-3816813

ABSTRACT

Subcellular fractionation techniques have been used to assess the localization of injected 125I-labeled cholera toxin (125I-CT) taken up by rat liver in vivo, and to determine whether internalization of the toxin is required for the generation of the active A1 peptide. The uptake of injected 125I-CT into the liver is maximal at 5 min (about 10% injected dose/g). At this time the radioactivity is for the most part recovered in the microsomal (P) fraction, but later on it progressively associates with the mitochondrial-lysosomal (ML) and supernatant fractions. The radioactivity is enriched 7-fold in plasma membranes at 5-15 min, and 15-60-fold in Golgi-endosome (GE) fractions at 15-60 min. On analytical sucrose gradients the radioactivity associated with the P fraction is progressively displaced from the region of 5'-nucleotidase (a plasma membrane marker) to that of galactosyltransferase (a Golgi marker). On Percoll gradients, however, it is displaced towards acid phosphatase (a lysosomal marker). Density-shift experiments, using Triton WR 1339, suggest that some radioactivity associated with the P fraction (at 30 min) and all the radioactivity present in the ML fraction (at 2 h) is intrinsic to acid-phosphatase-containing structures, presumably lysosomes. Comparable experiments using 3,3'-diaminobenzidine cytochemistry indicate that the radioactivity present in GE fractions is separable from galactosyltransferase, and thus is presumably associated with endosomes. The fate of injected 125I-labeled cholera toxin B subunit differs from that of the whole toxin by a more rapid uptake (and/or clearance) of the ligand into subcellular fractions, and a greater accumulation of ligand in the ML fraction. Analysis of GE fractions by SDS/polyacrylamide gel electrophoresis shows that, up to 10 min after injection of 125I-CT, about 80% of the radioactivity is recovered as A subunit and 20% as B subunit, similarly to control toxin. Later on there is a time-dependent decrease in the amount of A subunit and, at least with the intermediate GE fraction, a concomitant appearance of A1 peptide (about 15% of the total at 60 min). In contrast the radioactivity associated with plasma membranes remains indistinguishable from unused toxin. It is concluded that, upon interaction with hepatocytes, 125I-CT (both subunits A and B) sequentially associates with plasma membranes, endosomes and lysosomes, and that endosomes may represent the major subcellular site at which the A1 peptide is generated.


Subject(s)
Cholera Toxin/metabolism , Liver/metabolism , Subcellular Fractions/metabolism , Animals , Cell Membrane/metabolism , Centrifugation, Density Gradient , Electrophoresis, Polyacrylamide Gel , Golgi Apparatus/metabolism , Liver/enzymology , Male , Peptide Fragments/metabolism , Rats , Rats, Inbred Strains , Subcellular Fractions/enzymology
14.
J Biol Chem ; 266(20): 12858-65, 1991 Jul 15.
Article in English | MEDLINE | ID: mdl-2071575

ABSTRACT

Involvement of acidic cell compartments in processing and action of cholera toxin (CT) in rat liver has been examined using subcellular fractionation. Liver cell fractions prepared various times after CT injection display, after a lag phase, a progressive increase in adenylate cyclase activity, detectable earlier in Golgi-endosomal fractions (20 min) than in plasma membrane fractions (30 min), with a maximum (3-fold basal activity) achieved by 60-90 min. Endosomes containing in vivo internalized CT display a time-dependent increase in their ability to bind anti-A-subunit antibodies and to stimulate exogenous adenylate cyclase, which kinetically parallels the generation of A1 peptide, suggesting a translocation of A-subunit (or A1 peptide) across the endosomal membrane. In vivo chloroquine treatment inhibits endocytosis of CT taken up into the liver, lengthens the lag phase for adenylate cyclase activation by CT, and reduces by 3- to 10-fold the apparent affinity of the toxin for the enzyme. Incubation of endosomes containing internalized toxin at 37 degrees C under isotonic conditions results in a pH-dependent increase in generation of A1 peptide, membrane translocation of A-subunit (or A1 peptide), and degradation of toxin, with a maximum at pH 5. Addition of ATP, by decreasing the internal endosomal pH, stimulates both generation of the A1 peptide and degradation of toxin at pH 6-8. It is concluded that activation of adenylate cyclase by CT in intact liver requires association and subsequent processing of toxin in an acidic cell compartment, presumably endosomal.


Subject(s)
Adenylyl Cyclases/metabolism , Cholera Toxin/pharmacology , Liver/enzymology , Organelles/metabolism , Adenosine Triphosphate/metabolism , Animals , Biological Transport , Cell Fractionation , Cholera Toxin/metabolism , Enzyme Activation , Hydrogen-Ion Concentration , Intracellular Membranes/metabolism , Kinetics , Male , Models, Biological , Rats , Rats, Inbred Strains
15.
C R Seances Soc Biol Fil ; 175(4): 490-5, 1981.
Article in French | MEDLINE | ID: mdl-6457672

ABSTRACT

The O(H) foetal red cells are not always constantly agglutinated by the eel test serum, but the Sambucus ebulus L. fruit lectin provides a regular agglutination. Different experiments with treated red cells suggest that the lectin acts upon the membrane's inner sites. Besides the H sites, it is possible that other receptors might be bonded with the lectin.


Subject(s)
Erythrocytes/immunology , Hemagglutination Tests , Infant, Newborn , Lectins/pharmacology , Animals , Erythrocytes/drug effects , Fetus , Humans , Sheep
16.
Proc Natl Acad Sci U S A ; 86(8): 2642-6, 1989 Apr.
Article in English | MEDLINE | ID: mdl-2649887

ABSTRACT

Xenopus laevis oocytes possess a glucose transport system that is activated 3- to 5-fold by insulin-like growth factor I (Ka = 3 nM) and insulin (Ka = 200-250 nM), properties suggesting activation mediated by an insulin-like growth factor I receptor. This activation increases the Vmax of hexose uptake and has little or no effect on the Km for deoxyglucose (Km = 1-2 mM). Activation by hormone requires about 60 min and is inhibited by cytochalasin B but not by cycloheximide. The dependence of hexose uptake rate on hexose concentration exhibits cooperativity with Hill coefficients of 1.8 and 1.4 for the basal and hormone-activated states, respectively. Microinjection of a monoclonal antibody directed against the tyrosine kinase domain of the human insulin receptor blocks activation of hexose uptake by insulin-like growth factor I and insulin but has no effect on basal uptake. Taken together the results implicate the tyrosine-specific protein kinase activity of a cell-surface insulin-like growth factor I receptor in the activation of glucose transport in the Xenopus oocyte.


Subject(s)
Glucose/metabolism , Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Monosaccharide Transport Proteins/metabolism , Protein-Tyrosine Kinases/physiology , Somatomedins/pharmacology , Xenopus laevis/physiology , Animals , Biological Transport , Dose-Response Relationship, Drug , Hexoses/metabolism , Kinetics , Oocytes
17.
Eur J Biochem ; 193(2): 501-12, 1990 Oct 24.
Article in English | MEDLINE | ID: mdl-2146119

ABSTRACT

The degradation of insulin in isolated liver endosomes and the relationships of this process with ATP-dependent endosomal acidification have been studied. Incubation of endosomal fractions containing 125I-insulin in isotonic KCl at 30 degrees C resulted in a rapid loss of insulin integrity as judged from trichloroacetic acid precipitability, Sephadex G-50 chromatography, immunoreactivity and receptor binding ability, with a maximum at pH 5-6 (t1/2: 10, 10, 6 and 6 min, respectively). On a log/log plot, the amount of acid-soluble products generated was linearly related to the amount of insulin associated with endosomes (slope, 0.80). Upon incubation, virtually all acid-soluble products diffused out of endosomes as judged from their solubility in aqueous poly(ethyleneglycol). In permeabilized endosomes, intact insulin was also released in part extraluminally, but only when degradation was inhibited did this release increase with lowering pH. ATP shifted the pH for maximal insulin degradation to about 7.5-8.5 and caused endosomal acidification as judged from the uptake of acridine orange and the fluorescence of internalized fluorescein-labeled dextran and galactosylated bovine serum albumin (delta pH about 0.8-0.9). GTP, ITP and UTP exerted comparable effects but with lower potencies. The ability of ATP to alter the pH dependence of insulin degradation was maximal in the presence of Cl-, other anions being less effective (Br- greater than gluconate = SO4(2-) greater than NO3- = sucrose = mannitol) and/or inhibitory (NO3-). Na+, K+ and Li+ supported more effectively ATP-dependent insulin degradation than did choline. Divalent cations were required for the ATP effect (Mg2+ = Mn2+ greater than Co2+ greater than Ni2+ = Zn2 greater than Ca2+). Little or no effects of ATP occurred in the presence of proton ionophores such as monensin and carbonyl cyanide chlorophenylhydrazone, and inhibitors of the proton ATPase such as N-ethylmaleimide. The abilities of nucleotides, ions and inhibitors to support or inhibit ATP-dependent insulin degradation were well correlated with their abilities to affect ATP-dependent acidification. The acidotropic agents chloroquine and quinacrine caused a leftward shift in the pH dependence of insulin degradation and a decrease in maximal degradation; in the presence of ATP, chloroquine almost completely inhibited degradation at pH 5-9. It is concluded that ATP-dependent acidification, in part by enhancing the dissociation of the insulin-receptor complex, is required for optimum degradation of insulin within liver endosomes.


Subject(s)
Adenosine Triphosphate/physiology , Insulin/metabolism , Liver/metabolism , Organelles/metabolism , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphate/antagonists & inhibitors , Animals , Cations , Chloroquine/pharmacology , Guanosine Triphosphate/pharmacology , Hydrogen-Ion Concentration , Inosine Triphosphate/pharmacology , Liver/drug effects , Male , Organelles/drug effects , Quinacrine/pharmacology , Rats , Rats, Inbred Strains , Subcellular Fractions , Uridine Triphosphate/pharmacology
18.
J Biol Chem ; 266(15): 9382-91, 1991 May 25.
Article in English | MEDLINE | ID: mdl-1851744

ABSTRACT

Competitive hormone binding studies with membrane and partially purified receptors from Xenopus laevis oocytes revealed that the oocyte possesses high affinity (KD = 1-3 nM) binding sites for both insulin growth factors 1 and 2 (IGF-1 and IGF-2), but not for insulin. Consistent with these findings, IGF-1 activates hexose uptake by Xenopus oocytes with a KA (3 nM) identical with its KD, while IGF-2 and insulin activate hexose uptake with KA values of 50 nM and 200-250 nM, respectively, suggesting activation mediated through an IGF-1 receptor. Both IGF-1 and insulin activate receptor beta-subunit autophosphorylation and, thereby, protein substrate (reduced and carboxyamidomethylated lysozyme, i.e. RCAM-lysozyme) phosphorylation with KA values comparable to their respective KD values for ligand binding and KA values for activation of hexose uptake. The autophosphorylated beta-subunit(s) of the receptor were resolved into two discrete components, beta 1 and beta 2 (108 kDa and 94 kDa, respectively), which were phosphorylated exclusively on tyrosine and which exhibited similar extents of IGF-1-activated autophosphorylation. When added prior to autophosphorylation, RCAM-lysozyme blocks IGF-1-activated autophosphorylation and, thereby, IGF-1-activated protein substrate (RCAM-lysozyme) phosphorylation. Based on these findings, we conclude that IGF-1-stimulated autophosphorylation of its receptor is a prerequisite for catalysis of protein substrate phosphorylation by the receptor's tyrosine-specific protein kinase. The IGF-1 receptor kinase is implicated in signal transmission from the receptor, since anti-tyrosine kinase domain antibody blocks IGF-1-stimulated kinase activity in vitro and, when microinjected into intact oocytes, prevents IGF-1-stimulated hexose uptake.


Subject(s)
Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor I/metabolism , Oocytes/metabolism , Receptors, Cell Surface/metabolism , Animals , Binding, Competitive , Chromatography, High Pressure Liquid , Deoxyglucose/metabolism , Electrophoresis, Polyacrylamide Gel , Hexoses/metabolism , Kinetics , Peptide Mapping , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Receptors, Somatomedin , Trypsin , Xenopus laevis
19.
Biochem J ; 253(3): 735-43, 1988 Aug 01.
Article in English | MEDLINE | ID: mdl-2845931

ABSTRACT

The major steps in cholera-toxin action, i.e. binding, internalization, generation of A1 peptide and activation of adenylate cyclase, were examined in isolated hepatocytes. The binding of toxin involves a single class of high-affinity sites (KD congruent to 0.1 nM; Bmax. congruent to 10(7) sites/cell). At 37 degrees C, cell-associated toxin is progressively internalized, as judged by the loss of its accessibility to antibodies against whole toxin, A and B subunits (about 50, 75 and 30% of initially bound toxin after 40 min respectively). Two distinct pathways are involved in this process: endocytosis of the whole toxin, and selective penetration of the A subunit into the plasma membrane. Exposure of hepatocytes to an acidic medium (pH 5) results in a rapid and marked disappearance of the A subunit from the cell surface. Generation of A1 peptide and activation of adenylate cyclase by the toxin occur after a lag phase (10 min at 37 degrees C), and increase with time in a parallel manner up to 2-3% A1 peptide generated; they are unaffected by exposure of cells to an acidic medium. Chloroquine and monensin, which elevate the pH in acidic organelles, inhibit by 2-4-fold both the generation of A1 peptide and the activation of adenylate cyclase. Unexpectedly, these drugs also inhibit the internalization of the toxin. These results suggest that an acidic pH facilitates the penetration of A subunit into the plasma membrane and presumably the endosomal membrane as well, and that endocytosis of cholera toxin is required for generation of A1 peptide and activation of adenylate cyclase.


Subject(s)
Cholera Toxin/pharmacology , Liver/metabolism , Adenylyl Cyclases/metabolism , Animals , Binding Sites , Cell Membrane/drug effects , Cell Membrane/metabolism , Chloroquine/pharmacology , Cholera Toxin/metabolism , Cyclic AMP/metabolism , Enzyme Activation/drug effects , Hydrogen-Ion Concentration , Immune Sera , In Vitro Techniques , Liver/drug effects , Male , Monensin/pharmacology , Peptides/metabolism , Rats , Rats, Inbred Strains
20.
C R Seances Soc Biol Fil ; 170(4): 814-9, 1976.
Article in French | MEDLINE | ID: mdl-137048

ABSTRACT

The antigenic properties of a lamb mucin and of a glycoprotein isolated from it are investigated. The proteins are studied by immunoelectrophoresis of the mucin and of the glycoprotein against either a sheep serum antiserum or an exclusively glycoprotein antiserum. No serum protein could be shown in the mucin. In addition, the glycoprotein gives a precipitin line only with the specific antiserum; its antigenic power seems to be concealed in the original mucin. The carbohydrates are studied through the investigation of the blood group activities by the hemagglutination inhibition technique. The mucin and the isolated glycoprotein show the same types of activities: they both prevent the agglutination of A1 and 0 red cells. The presence of two blood group activities in that glycoportein and its non-reactivity against A2 red cells corroborate the hypothesis that carbohydrate chains must be branched for the elicitation of these activities in a single molecule.


Subject(s)
Antigens , Blood Group Antigens , Mucoproteins/immunology , Amino Acids/analysis , Animals , Hemagglutination Inhibition Tests , Immunoelectrophoresis , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL