Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 16 de 16
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
bioRxiv ; 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38915546

RÉSUMÉ

Purinergic P2X3 receptors form trimeric cation-gated channels, which are activated by extracellular ATP. P2X3 plays a crucial role in chronic cough and affects over 10% of the population. Despite considerable efforts to develop drugs targeting P2X3, the highly conserved structure within the P2X receptor family presents obstacles for achieving selectivity. Camlipixant, a potent and selective P2X3 antagonist, is currently in phase III clinical trials. However, the mechanisms underlying receptor desensitization, ion permeation, principles governing antagonism, and the structure of P2X3 when bound to camlipixant remain elusive. In this study, we established a stable cell line expressing homotrimeric P2X3 and utilized a peptide scaffold to purify the complex and determine its structure using cryo-electron microscopy (cryo-EM). P2X3 binds to camlipixant at a previously unidentified drug-binding site and functions as an allosteric inhibitor. Structure-activity studies combined with modeling and simulations have shed light on the mechanisms underlying the selective targeting and inhibition of P2X3 by camlipixant, distinguishing it from other members of the P2X receptor family.

2.
Int J Parasitol Drugs Drug Resist ; 16: 174-187, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-34252686

RÉSUMÉ

Currently, only a few chemical drug classes are available to control the global burden of nematode infections in humans and animals. Most of these drugs exert their anthelmintic activity by interacting with proteins such as ion channels, and the nematode neuromuscular system remains a promising target for novel intervention strategies. Many commonly-used phenotypic readouts such as motility provide only indirect insight into neuromuscular function and the site(s) of action of chemical compounds. Electrophysiological recordings provide more specific information but are typically technically challenging and lack high throughput for drug discovery. Because drug discovery relies strongly on the evaluation and ranking of drug candidates, including closely related chemical derivatives, precise assays and assay combinations are needed for capturing and distinguishing subtle drug effects. Past studies show that nematode motility and pharyngeal pumping (feeding) are inhibited by most anthelmintic drugs. Here we compare two microfluidic devices ("chips") that record electrophysiological signals from the nematode pharynx (electropharyngeograms; EPGs) ─ the ScreenChip™ and the 8-channel EPG platform ─ to evaluate their respective utility for anthelmintic research. We additionally compared EPG data with whole-worm motility measurements obtained with the wMicroTracker instrument. As references, we used three macrocyclic lactones (ivermectin, moxidectin, and milbemycin oxime), and levamisole, which act on different ion channels. Drug potencies (IC50 and IC95 values) from concentration-response curves, and the time-course of drug effects, were compared across platforms and across drugs. Drug effects on pump timing and EPG waveforms were also investigated. These experiments confirmed drug-class specific effects of the tested anthelmintics and illustrated the relative strengths and limitations of the different assays for anthelmintic research.


Sujet(s)
Anthelminthiques , Nematoda , Animaux , Anthelminthiques/pharmacologie , Caenorhabditis elegans , Humains , Ivermectine , Lévamisole/pharmacologie
3.
Int J Parasitol Drugs Drug Resist ; 14: 237-248, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-33249235

RÉSUMÉ

For more than four decades, the free-living nematode Caenorhabditis elegans has been extensively used in anthelmintic research. Classic genetic screens and heterologous expression in the C. elegans model enormously contributed to the identification and characterization of molecular targets of all major anthelmintic drug classes. Although these findings provided substantial insights into common anthelmintic mechanisms, a breakthrough in the treatment and control of parasitic nematodes is still not in sight. Instead, we are facing increasing evidence that the enormous diversity within the phylum Nematoda cannot be recapitulated by any single free-living or parasitic species and the development of novel broad-spectrum anthelmintics is not be a simple goal. In the present review, we summarize certain milestones and challenges of the C. elegans model with focus on drug target identification, anthelmintic drug discovery and identification of resistance mechanisms. Furthermore, we present new perspectives and strategies on how current progress in C. elegans research will support future anthelmintic research.


Sujet(s)
Anthelminthiques , Caenorhabditis elegans , Animaux , Anthelminthiques/usage thérapeutique , Découverte de médicament , Nematoda
4.
ACS Omega ; 5(22): 13034-13041, 2020 Jun 09.
Article de Anglais | MEDLINE | ID: mdl-32548488

RÉSUMÉ

Lactate dehydrogenase A (LDHA) is frequently overexpressed in tumors, thereby sustaining high glycolysis rates, tumor growth, and chemoresistance. High-throughput screening resulted in the identification of phthalimide and dibenzofuran derivatives as novel lactate dehydrogenase inhibitors, selectively inhibiting the activity of the LDHA isoenzyme. Cocrystallization experiments confirmed target engagement in addition to demonstrating binding to a novel allosteric binding site present in all four LDHA subunits of the LDH5 homotetramer.

5.
ChemMedChem ; 11(20): 2261-2271, 2016 10 19.
Article de Anglais | MEDLINE | ID: mdl-27552707

RÉSUMÉ

Despite the long-known fact that the facilitative glucose transporter GLUT1 is one of the key players safeguarding the increase in glucose consumption of many tumor entities even under conditions of normal oxygen supply (known as the Warburg effect), only few endeavors have been undertaken to find a GLUT1-selective small-molecule inhibitor. Because other transporters of the GLUT1 family are involved in crucial processes, these transporters should not be addressed by such an inhibitor. A high-throughput screen against a library of ∼3 million compounds was performed to find a small molecule with this challenging potency and selectivity profile. The N-(1H-pyrazol-4-yl)quinoline-4-carboxamides were identified as an excellent starting point for further compound optimization. After extensive structure-activity relationship explorations, single-digit nanomolar inhibitors with a selectivity factor of >100 against GLUT2, GLUT3, and GLUT4 were obtained. The most promising compound, BAY-876 [N4 -[1-(4-cyanobenzyl)-5-methyl-3-(trifluoromethyl)-1H-pyrazol-4-yl]-7-fluoroquinoline-2,4-dicarboxamide], showed good metabolic stability in vitro and high oral bioavailability in vivo.


Sujet(s)
Transporteur de glucose de type 1/antagonistes et inhibiteurs , Pyrazoles/pharmacologie , Quinoléines/pharmacologie , Administration par voie orale , Biodisponibilité , Transporteur de glucose de type 1/métabolisme , Tests de criblage à haut débit , Humains , Structure moléculaire , Pyrazoles/administration et posologie , Pyrazoles/composition chimique , Quinoléines/administration et posologie , Quinoléines/composition chimique , Relation structure-activité
6.
Proc Natl Acad Sci U S A ; 113(17): 4711-6, 2016 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-27078104

RÉSUMÉ

Cancerous cells have an acutely increased demand for energy, leading to increased levels of human glucose transporter 1 (hGLUT1). This up-regulation suggests hGLUT1 as a target for therapeutic inhibitors addressing a multitude of cancer types. Here, we present three inhibitor-bound, inward-open structures of WT-hGLUT1 crystallized with three different inhibitors: cytochalasin B, a nine-membered bicyclic ring fused to a 14-membered macrocycle, which has been described extensively in the literature of hGLUTs, and two previously undescribed Phe amide-derived inhibitors. Despite very different chemical backbones, all three compounds bind in the central cavity of the inward-open state of hGLUT1, and all binding sites overlap the glucose-binding site. The inhibitory action of the compounds was determined for hGLUT family members, hGLUT1-4, using cell-based assays, and compared with homology models for these hGLUT members. This comparison uncovered a probable basis for the observed differences in inhibition between family members. We pinpoint regions of the hGLUT proteins that can be targeted to achieve isoform selectivity, and show that these same regions are used for inhibitors with very distinct structural backbones. The inhibitor cocomplex structures of hGLUT1 provide an important structural insight for the design of more selective inhibitors for hGLUTs and hGLUT1 in particular.


Sujet(s)
Cytochalasines/composition chimique , Transporteur de glucose de type 1/antagonistes et inhibiteurs , Transporteur de glucose de type 1/ultrastructure , Glucose/composition chimique , Phénylalanine/analogues et dérivés , Séquence d'acides aminés , Sites de fixation , Simulation numérique , Séquence conservée , Humains , Modèles chimiques , Modèles moléculaires , Phénylalanine/composition chimique , Liaison aux protéines , Conformation des protéines
7.
Bioorg Med Chem Lett ; 26(7): 1732-7, 2016 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-26949183

RÉSUMÉ

The compound class of 1H-pyrazolo[3,4-d]pyrimidines was identified using HTS as very potent inhibitors of facilitated glucose transporter 1 (GLUT1). Extensive structure-activity relationship studies (SAR) of each ring system of the molecular framework was established revealing essential structural motives (i.e., ortho-methoxy substituted benzene, piperazine and pyrimidine). The selectivity against GLUT2 was excellent and initial in vitro and in vivo pharmacokinetic (PK) studies are encouraging.


Sujet(s)
Transporteur de glucose de type 1/antagonistes et inhibiteurs , Pyrimidines/composition chimique , Pyrimidines/pharmacologie , Animaux , Lignée cellulaire , Découverte de médicament , Transporteur de glucose de type 1/métabolisme , Humains , Mâle , Pyrimidines/pharmacocinétique , Rat Wistar , Relation structure-activité
8.
Mol Cancer Ther ; 13(6): 1537-48, 2014 Jun.
Article de Anglais | MEDLINE | ID: mdl-24714131

RÉSUMÉ

Mesothelin is a tumor differentiation antigen frequently overexpressed in tumors such as mesothelioma, ovarian, pancreatic, and lung adenocarcinomas while showing limited expression in nonmalignant tissues. Mesothelin is therefore an attractive target for cancer therapy using antibody-drug conjugates (ADC). This study describes the detailed characterization of anetumab ravtansine, here referred to as BAY 94-9343, a novel ADC consisting of a human anti-mesothelin antibody conjugated to the maytansinoid tubulin inhibitor DM4 via a disulfide-containing linker. Binding properties of the anti-mesothelin antibody were analyzed using surface plasmon resonance, immunohistochemistry, flow cytometry, and fluorescence microscopy. Effects of BAY 94-9343 on cell proliferation were first studied in vitro and subsequently in vivo using subcutaneous, orthotopic, and patient-derived xenograft tumor models. The antibody binds to human mesothelin with high affinity and selectivity, thereby inducing efficient antigen internalization. In vitro, BAY 94-9343 demonstrated potent and selective cytotoxicity of mesothelin-expressing cells with an IC(50) of 0.72 nmol/L, without affecting mesothelin-negative or nonproliferating cells. In vivo, BAY 94-9343 localized specifically to mesothelin-positive tumors and inhibited tumor growth in both subcutaneous and orthotopic xenograft models. In addition, BAY 94-9343 was able to induce a bystander effect on neighboring mesothelin-negative tumor cells. Antitumor efficacy of BAY 94-9343 correlated with the amount of mesothelin expressed and was generally superior to that of standard-of-care regimen resulting in complete tumor eradication in most of the models. BAY 94-9343 is a selective and highly potent ADC, and our data support its development for the treatment of patients with mesothelin-expressing tumors.


Sujet(s)
Anticorps monoclonaux/administration et posologie , Protéines liées au GPI/immunologie , Immunoconjugués/administration et posologie , Maitansine/analogues et dérivés , Thérapie moléculaire ciblée , Tumeurs/traitement médicamenteux , Anticorps monoclonaux/immunologie , Cytotoxicité à médiation cellulaire dépendante des anticorps/immunologie , Effet bystander , Lignée cellulaire tumorale , Protéines liées au GPI/antagonistes et inhibiteurs , Régulation de l'expression des gènes tumoraux/immunologie , Humains , Maitansine/administration et posologie , Mésothéline , Tumeurs/immunologie , Tumeurs/anatomopathologie , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Cancer Med ; 2(5): 611-24, 2013 Oct.
Article de Anglais | MEDLINE | ID: mdl-24403227

RÉSUMÉ

The activation of the transcription factor hypoxia-inducible factor-1 (HIF-1) plays an essential role in tumor development, tumor progression, and resistance to chemo- and radiotherapy. In order to identify compounds targeting the HIF pathway, a small molecule library was screened using a luciferase-driven HIF-1 reporter cell line under hypoxia. The high-throughput screening led to the identification of a class of aminoalkyl-substituted compounds that inhibited hypoxia-induced HIF-1 target gene expression in human lung cancer cell lines at low nanomolar concentrations. Lead structure BAY 87-2243 was found to inhibit HIF-1α and HIF-2α protein accumulation under hypoxic conditions in non-small cell lung cancer (NSCLC) cell line H460 but had no effect on HIF-1α protein levels induced by the hypoxia mimetics desferrioxamine or cobalt chloride. BAY 87-2243 had no effect on HIF target gene expression levels in RCC4 cells lacking Von Hippel-Lindau (VHL) activity nor did the compound affect the activity of HIF prolyl hydroxylase-2. Antitumor activity of BAY 87-2243, suppression of HIF-1α protein levels, and reduction of HIF-1 target gene expression in vivo were demonstrated in a H460 xenograft model. BAY 87-2243 did not inhibit cell proliferation under standard conditions. However under glucose depletion, a condition favoring mitochondrial ATP generation as energy source, BAY 87-2243 inhibited cell proliferation in the nanomolar range. Further experiments revealed that BAY 87-2243 inhibits mitochondrial complex I activity but has no effect on complex III activity. Interference with mitochondrial function to reduce hypoxia-induced HIF-1 activity in tumors might be an interesting therapeutic approach to overcome chemo- and radiotherapy-resistance of hypoxic tumors.


Sujet(s)
Complexe I de la chaîne respiratoire/antagonistes et inhibiteurs , Tumeurs du poumon/métabolisme , Oxadiazoles/pharmacologie , Pyrazoles/pharmacologie , Animaux , Antigènes néoplasiques/biosynthèse , Antigènes néoplasiques/génétique , Carbonic anhydrase IX , Carbonic anhydrases/biosynthèse , Carbonic anhydrases/génétique , Hypoxie cellulaire/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Découverte de médicament/méthodes , Complexe I de la chaîne respiratoire/métabolisme , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Gènes tumoraux , Gènes rapporteurs , Humains , Facteur-1 induit par l'hypoxie/biosynthèse , Facteur-1 induit par l'hypoxie/génétique , Hypoxia-inducible factor-proline dioxygenases/génétique , Hypoxia-inducible factor-proline dioxygenases/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Souris , Souris nude , Données de séquences moléculaires , Thérapie moléculaire ciblée/méthodes , Oxadiazoles/administration et posologie , Oxadiazoles/sang , Oxadiazoles/usage thérapeutique , Pyrazoles/administration et posologie , Pyrazoles/sang , Pyrazoles/usage thérapeutique , Petit ARN interférent/génétique , Bibliothèques de petites molécules , Charge tumorale/effets des médicaments et des substances chimiques , Cellules cancéreuses en culture , Protéine Von Hippel-Lindau supresseur de tumeur/physiologie , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes
10.
Cancer Biol Ther ; 7(2): 237-42, 2008 Feb.
Article de Anglais | MEDLINE | ID: mdl-18059188

RÉSUMÉ

Treatment of oral squamous cell carcinoma (OSCC) is currently based on surgery and radiotherapy. Prolongation of the survival time of patients with progressing tumors is infrequently achieved. To improve the therapeutic options, targeted therapies are a favorable alternative. Therefore, we analyzed the effect of a chimeric toxin (CT) named SE consisting of the epidermal growth factor and the plant protein toxin saporin from Saponaria officinalis. A second construct (SA2E) additionally contains a peptidic adapter designed to enhance efficacy of the CT in vivo and to reduce side effects. The IC(50) values for an OSCC cell line (BHY) were 0.27 nM and 0.73 nM for SE and SA2E, respectively, while fibroblasts remained unaffected. To investigate primary tumor cells, we developed a technique to analyze freshly prepared OSCC cells of 28 patients in a stem cell assay directly after surgery. Cells were treated for 1 h with the CTs, subsequently seeded into soft agar and colony growth determined after 1-2 weeks In spite of the short time of CT incubation, the amount of colonies was reduced to about 78% by 10 nM and to 69% by 100 nM of either toxin. A combined application of 10 nM SA2E with a saponin from Gypsophila paniculata reduced the amount of surviving cells to 68%. The results demonstrate the impact of the CTs on OSCC cells and depict that the stem cell assay is suitable to determine the potential of anti-tumor drugs before studies in vivo will be initiated.


Sujet(s)
Carcinome épidermoïde/traitement médicamenteux , Facteur de croissance épidermique/pharmacologie , Immunotoxines/pharmacologie , Tumeurs de la bouche/traitement médicamenteux , Saponines/pharmacologie , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Test clonogénique , Relation dose-effet des médicaments , Association médicamenteuse , Synergie des médicaments , Facteur de croissance épidermique/isolement et purification , Escherichia coli/génétique , Humains , Immunotoxines/composition chimique , Concentration inhibitrice 50 , Saponines/isolement et purification , Facteurs temps
11.
J Control Release ; 117(3): 342-50, 2007 Feb 26.
Article de Anglais | MEDLINE | ID: mdl-17207883

RÉSUMÉ

Two of the main problems associated with administration of receptor-targeted toxins in tumor therapy are severe systemic side effects and low transfer of the toxins into the cytosol after binding to the tumor cell surface. To improve chimeric toxins in this respect we have developed a molecular adapter that links the toxic moiety and ligand. The adapter is designed to improve cytosolic uptake, retain the toxin inside the cytosol and detoxify the drug after cell death. The plant toxin saporin linked either directly or via the adapter to epidermal growth factor (EGF) served to evaluate efficacy to inhibit tumor growth and reduce side effects in vivo. The lethal dose for BALB/c mice was three times less for the adapter-containing toxin (SA2E) than for the adapter-free construct (SE). Furthermore, SE only reduced the average weight of induced tumors by 33% whereas SA2E-treated mice exhibited 71% reduction with an almost complete suppression in 60% of the cases. Additionally, severe side effects like hyperalgesia, alopecia and death were drastically reduced in SA2E-treated animals. Tumors without target receptor were only slightly affected by SA2E and the reduction in side effects less pronounced indicating specific depletion from the blood by target receptor expressing cells.


Sujet(s)
Antinéoplasiques/administration et posologie , Antinéoplasiques/usage thérapeutique , Systèmes de délivrance de médicaments , Tumeurs/traitement médicamenteux , Alanine transaminase/métabolisme , Animaux , Antinéoplasiques/effets indésirables , Aspartate aminotransferases/métabolisme , Technique de Western , Poids/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Test ELISA , Récepteurs ErbB/biosynthèse , Escherichia coli/génétique , Immunotoxines/administration et posologie , Immunotoxines/effets indésirables , Immunotoxines/usage thérapeutique , Souris , Souris de lignée BALB C , Plasmides/génétique , Transfection
12.
Exp Biol Med (Maywood) ; 231(4): 412-20, 2006 Apr.
Article de Anglais | MEDLINE | ID: mdl-16565437

RÉSUMÉ

Saponins are a group of plant glycosides consisting of a steroid or triterpenoid aglycone to which one or more sugar chains are attached. They exhibit cell membrane-permeabilizing properties and, thus, have been investigated for their therapeutic potential. Recently, at a non-permeabilizing concentration saponinum album from Gypsophila paniculata L. has been described to enhance the cytotoxicity of a chimeric toxin in a cell culture model. To elucidate whether this enhancing effect is also mediated by other saponins, we analyzed the ability of seven different saponins to enhance the cytotoxicity of a targeted chimeric toxin. The chimeric toxin is composed of saporin, a plant ribosome-inactivating toxin, a cleavable adapter, and human epidermal growth factor (EGF). Cytotoxicity on EGF receptor (EGFR)-bearing cells was analyzed both alone and after combined application of saponin and chimeric toxin. Only two of the tested saponins, quillajasaponin and saponinum album, enhanced cytotoxicity by more than 1,000-fold, whereas the enhancement factors of the other saponins were only approximately 10-fold. In contrast to saponinum album, quillajasaponin enhanced the cytotoxicity both on control cells lacking EGFR and on target cells, indicating that, in this case, the enhancement is not target cell receptor specific. This is also the case for some of the saponins with low enhancement factors. Saponinum album resulted in a more than 13,600-fold receptor-specific enhancement, decreasing the 50% inhibitory concentration (IC(50)) from 2.4 nM to 0.18 pM, which renders it the best option to promote saporin-3-based drug uptake while retaining specificity for the EGFR.


Sujet(s)
Récepteurs ErbB/métabolisme , Immunotoxines/pharmacologie , N-Glycosyl hydrolases/pharmacologie , Protéines végétales/pharmacologie , Saponines/pharmacologie , Animaux , Survie cellulaire/effets des médicaments et des substances chimiques , Facteur de croissance épidermique/pharmacologie , Récepteurs ErbB/génétique , Humains , Ligands , Souris , Cellules NIH 3T3 , Préparations pharmaceutiques/métabolisme , Protéines inactivant les ribosomes de type 1 , Saponines/composition chimique , Saporines , Relation structure-activité , Transfection
13.
Biochem Biophys Res Commun ; 337(2): 602-9, 2005 Nov 18.
Article de Anglais | MEDLINE | ID: mdl-16199006

RÉSUMÉ

Direct targeting to the cytoplasm and nucleus using protein transduction domains (PTD) has been described to be efficient but non-cell-type-specific, and only has clinical relevance when the molecule is active exclusively in the diseased cell. The use of PTDs is an attractive mechanism to improve drug delivery. In this work, we designed recombinant proteins that contain epidermal growth factor as ligand to render uptake target cell-specific. We evaluated the potential of several PTDs to induce the cytosolic uptake of the catalytic domain of diphtheria toxin by measuring cytotoxicity. Although PTD-dependent membrane transfer is very low, the proteins exhibited concentration-dependent cytotoxic activity. Higher binding at 4 degrees C compared to 37 degrees C suggests that uptake by the PTDs MTS and TLM occurs via an endocytic pathway. Non-specific binding is predominantly a function of the PTD and greatly increases by substitution of a non-polar glycine with a negatively charged glutamate in the PTD HA2.


Sujet(s)
Structure tertiaire des protéines/physiologie , Protéines de fusion recombinantes/métabolisme , Transduction génétique , Séquence d'acides aminés , Cellules cultivées , Systèmes de délivrance de médicaments , Acide glutamique/composition chimique , Acide glutamique/métabolisme , Humains , Données de séquences moléculaires , Liaison aux protéines , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/génétique , Température
14.
J Control Release ; 106(1-2): 123-37, 2005 Aug 18.
Article de Anglais | MEDLINE | ID: mdl-15935506

RÉSUMÉ

Immunotoxins have to be administered in high doses due to low cytosolic uptake with the consequence of severe side effects. Recently we found that the cytotoxic activity from Agrostemma githago seeds can be attributed to a synergistic toxicity of a triterpenoid saponin and a ribosome-inactivating protein. Here we investigated whether saponins are able to enhance the efficacy of a receptor-specific chimeric toxin consisting of saporin-3, epidermal growth factor and a molecular adapter previously shown to reduce side effects on non-target cells. Pre-applied saponin enhances the target cell-specific cytotoxic effect, dependent on the cell line, between 3560- and 385,000-fold with an IC50 up to 0.67 pM. Non-target cells are not affected at the same concentration. At the optimal concentrations of the chimeric toxin and saponin application of either one of the components shows no cytotoxicity at all proving a synergistic effect. In the presence of saponin ligand-free saporin-3 does not exhibit any cytotoxic effect up to 0.1 nM providing further evidence for an increased specificity. This synergistic effect is in the same order of magnitude as in a mouse model. Our investigations clearly demonstrate that a combined administration of saponin and chimeric toxins opens up a promising perspective for tumor therapy.


Sujet(s)
Survie cellulaire/effets des médicaments et des substances chimiques , Immunotoxines/pharmacologie , Protéines végétales/pharmacologie , Saponines/pharmacologie , Animaux , Caryophyllaceae/composition chimique , Lignée cellulaire tumorale , Association médicamenteuse , Synergie des médicaments , Humains , Souris , Cellules NIH 3T3
15.
Int J Cancer ; 103(2): 277-82, 2003 Jan 10.
Article de Anglais | MEDLINE | ID: mdl-12455044

RÉSUMÉ

One of the problems associated with the administration of immunotoxins is hypersensitivity reaction such as vascular leak syndrome. This may be prevented by decreasing the plasma half-life. To improve immunotoxins with respect to reduced side effects, we have previously described the development of a cleavable adapter. This adapter links the toxic moiety and ligand that are usually directly coupled. In our study, the cytotoxicity of saporin linked either directly or via the adapter to epidermal growth factor (EGF) was evaluated in vitro. The immunotoxins exhibited similar cytotoxic activity towards A-431 and HER14 cells (IC(50) < 10 nM). The supernatant from 6 hr cultures of HER14 cells incubated in the presence of the adapter-containing immunotoxin exhibited a significantly reduced cytotoxicity as compared to the directly coupled immunotoxin. Western blotting revealed that the adapter was cleaved, thus supporting our proposal that cleavable adapters may reduce nonspecific effects. A similar reduced half-life was detected in platelet-poor plasma. In contrast MCF-7 cells remain unaffected by the immunotoxins. This was shown to be due to the absence of detectable EGF-receptor in comparison to A-431 and HER14 cells as determined by Western blotting. Furthermore, we could show that the adapter does not exert an effect on the N-glycosidase activity of saporin. These results suggest that the use of cleavable adapters may be a useful tool in immunotoxins for reducing the killing of surrounding noncancerous cells due to nonspecific binding.


Sujet(s)
Immunotoxines/toxicité , N-Glycosyl hydrolases/toxicité , Protéines végétales/toxicité , Cellules cancéreuses en culture/effets des médicaments et des substances chimiques , Adénine/métabolisme , Division cellulaire/effets des médicaments et des substances chimiques , Amorces ADN/composition chimique , Facteur de croissance épidermique/toxicité , Récepteurs ErbB/métabolisme , Glycosidases/métabolisme , Période , Antigènes de surface du virus de l'hépatite B/génétique , Antigènes de surface du virus de l'hépatite B/métabolisme , Humains , Techniques in vitro , Réaction de polymérisation en chaîne , Protéines recombinantes/métabolisme , Protéines recombinantes/toxicité , Protéines inactivant les ribosomes de type 1 , Saporines , Cellules cancéreuses en culture/métabolisme
16.
Anal Biochem ; 302(1): 114-22, 2002 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-11846384

RÉSUMÉ

Adenine quantitation is required for a variety of applications. To date, the prevalent method for quantifying free adenine, in a variety of applications, is the detection of fluorescent-derivatized adenine by HPLC. For the present study, we developed a high-throughput, nonradioactive, enzyme-based colorimetric adenine quantitation assay that is performed in one multireaction incubation step. The assay does not require adenine derivatization and is designed for microplates. The key step is the conversion of adenine to adenosine monophosphate by adenine phosphoribosyl transferase. Subsequent reactions finally produce three inorganic phosphate ions per adenine molecule. Phosphate is quantitated by the color-generating phosphorylysis of a particular purine derivate. Ribosome-inactivating proteins that release adenine from polynucleotides are often used to investigate intracellular protein trafficking and are important for the design of immunotoxins. We therefore used ricin, dianthin, saporin, and a variety of saporin fusion proteins to show that this method is suitable for quantifying adenine release using different substrates. The measured rate of adenine release and substrate specificity are comparable to those determined by HPLC and radioactive detection techniques.


Sujet(s)
Adénine/analyse , Immunotoxines , N-Glycosyl hydrolases , Protéines végétales/analyse , Ricine/analyse , Dosage biologique , Colorimétrie/méthodes , Cinétique , Protéines inactivant les ribosomes de type 1 , Ribosomes , Saporines , Sensibilité et spécificité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...