Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
2.
Sci Rep ; 12(1): 43, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34997079

ABSTRACT

The ability to cryopreserve mosquitoes would revolutionize work on these vectors of major human infectious diseases by conserving stocks, new isolates, lab-bred strains, and transgenic lines that currently require continuous life cycle maintenance. Efforts over several decades to develop a method for cryopreservation have, until now, been fruitless: we describe here a method for the cryopreservation of Anopheles stephensi embryos yielding hatch rates of ~ 25%, stable for > 5 years. Hatched larvae developed into fertile, fecund adults and blood-fed females, produced fully viable second generation eggs, that could be infected with Plasmodium falciparum at high intensities. The key components of the cryopreservation method are: embryos at 15-30 min post oviposition, two incubation steps in 100% deuterated methanol at - 7 °C and - 14.5 °C, and rapid cooling. Eggs are recovered by rapid warming with concomitant dilution of cryoprotectant. Eggs of genetically modified A. stephensi and of A. gambiae were also successfully cryopreserved. This enabling methodology will allow long-term conservation of mosquitoes as well as acceleration of genetic studies and facilitation of mass storage of anopheline mosquitoes for release programs.


Subject(s)
Anopheles/embryology , Cryopreservation/methods , Embryo, Nonmammalian/embryology , Animals , Anopheles/parasitology , Cryoprotective Agents , Larva/growth & development , Oviposition , Plasmodium falciparum/parasitology
5.
Eur Urol Focus ; 5(4): 577-584, 2019 Jul.
Article in English | MEDLINE | ID: mdl-29221876

ABSTRACT

CONTEXT: Whether hypofractionated radiation therapy (RT) compared with conventionally fractionated RT provides comparable or possibly improved cancer control without increased toxicity in localized prostate cancer (PC) remains unknown. OBJECTIVE: Realizing from the CHHiP trial that outcomes are highly sensitive to the dose fractionation schedule and number of treatments, we conducted a systematic review and meta-analysis selecting only the randomized noninferiority trials, because the randomized arms closely approximated one another in terms of the dose fractionation schedule, and compared cancer control and toxicity of hypofractionated RT with conventionally fractionated RT for localized PC. EVIDENCE ACQUISITION: Randomized noninferiority trials evaluating hypofractionated (2.4-4Gy daily fractions for 15-30 treatments) versus conventionally fractionated RT (1.8-2Gy daily fractions for 40-45 treatments) in men with localized PC were selected. Studies that were not noninferiority trials, used extreme hypofractionation, or treated metastatic disease were excluded. Three studies were retained for analysis. Data were pooled using a random-effects model to determine hazard ratio (HR) and risk ratio (RR). Heterogeneity was assessed via chi-square test, I2 statistics, and metaregression. The primary outcome was disease-free survival (DFS), defined as death from any cause or biochemical, local, regional, or distant progression. EVIDENCE SYNTHESIS: Of the 5484 men, 3553 (64.8%) had intermediate-risk PC. Hypofractionated RT as compared with conventionally fractionated RT was associated with significantly improved DFS (HR 0.869; 95% confidence interval [CI], 0.757, 0.998; p=0.047), whereas overall survival was not (HR 0.84; 95% CI, 0.66, 1.07; p=0.16). Acute grade 2 or higher gastrointestinal toxicity was significantly increased with hypofractionation (RR 1.42; 95% CI 1.15, 1.77; p=0.002); however, this did not translate into late grade 2 or higher gastrointestinal toxicity. An increase in late grade 2 or higher genitourinary complications was observed (RR 1.18; 95% CI 0.98, 1.43; p=0.08). CONCLUSIONS: Hypofractionated RT as compared with conventionally fractionated RT could improve DFS in men with intermediate-risk PC and, therefore, would be reasonable to consider in men who do not have risk factors for late genitourinary complications. PATIENT SUMMARY: Treatment with a shorter course of radiation, using higher doses per treatment over fewer days, may be the preferred approach in appropriately selected patients with localized prostate cancer.


Subject(s)
Prostatic Neoplasms/radiotherapy , Radiation Dose Hypofractionation , Equivalence Trials as Topic , Humans , Male , Prostatic Neoplasms/pathology , Randomized Controlled Trials as Topic
7.
Sci Rep ; 8(1): 12716, 2018 08 24.
Article in English | MEDLINE | ID: mdl-30143707

ABSTRACT

P-glycoprotein (P-gp), an ATP-dependent efflux pump, is associated with the development of multidrug resistance in cancer cells. Antibody-mediated blockade of human P-gp activity has been shown to overcome drug resistance by re-sensitizing resistant cancer cells to anticancer drugs. Despite the potential clinical application of this finding, the epitopes of the three human P-gp-specific monoclonal antibodies MRK-16, UIC2 and 4E3, which bind to the extracellular loops (ECLs) have not yet been mapped. By generating human-mouse P-gp chimeras, we mapped the epitopes of these antibodies to ECLs 1 and 4. We then identified key amino acids in these regions by replacing mouse residues with homologous human P-gp residues to recover binding of antibodies to the mouse P-gp. We found that changing a total of ten residues, five each in ECL1 and ECL4, was sufficient to recover binding of both MRK-16 and 4E3 antibodies, suggesting a common epitope. However, recovery of the conformation-sensitive UIC2 epitope required replacement of thirteen residues in ECL1 and the same five residues replaced in the ECL4 for MRK-16 and 4E3 binding. These results demonstrate that discontinuous epitopes for MRK-16, UIC2 and 4E3 are located in the same regions of ECL1 and 4 of the multidrug transporter.


Subject(s)
Antibodies, Monoclonal/chemistry , Epitope Mapping , Epitopes/chemistry , ATP Binding Cassette Transporter, Subfamily B/chemistry , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Epitopes/genetics , Epitopes/metabolism , HeLa Cells , Humans , Mice , Protein Structure, Secondary
8.
Nat Rev Cancer ; 18(7): 452-464, 2018 07.
Article in English | MEDLINE | ID: mdl-29643473

ABSTRACT

Most patients who die of cancer have disseminated disease that has become resistant to multiple therapeutic modalities. Ample evidence suggests that the expression of ATP-binding cassette (ABC) transporters, especially the multidrug resistance protein 1 (MDR1, also known as P-glycoprotein or P-gp), which is encoded by ABC subfamily B member 1 (ABCB1), can confer resistance to cytotoxic and targeted chemotherapy. However, the development of MDR1 as a therapeutic target has been unsuccessful. At the time of its discovery, appropriate tools for the characterization and clinical development of MDR1 as a therapeutic target were lacking. Thirty years after the initial cloning and characterization of MDR1 and the implication of two additional ABC transporters, the multidrug resistance-associated protein 1 (MRP1; encoded by ABCC1)), and ABCG2, in multidrug resistance, interest in investigating these transporters as therapeutic targets has waned. However, with the emergence of new data and advanced techniques, we propose to re-evaluate whether these transporters play a clinical role in multidrug resistance. With this Opinion article, we present recent evidence indicating that it is time to revisit the investigation into the role of ABC transporters in efficient drug delivery in various cancer types and at the blood-brain barrier.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Blood-Brain Barrier , Humans , Molecular Targeted Therapy , Multidrug Resistance-Associated Proteins/genetics
9.
J Biol Chem ; 292(2): 446-461, 2017 Jan 13.
Article in English | MEDLINE | ID: mdl-27864369

ABSTRACT

P-glycoprotein (P-gp) is a polyspecific ATP-dependent transporter linked to multidrug resistance in cancer; it plays important roles in determining the pharmacokinetics of many drugs. Understanding the structural basis of P-gp, substrate polyspecificity has been hampered by its intrinsic flexibility, which is facilitated by a 75-residue linker that connects the two halves of P-gp. Here we constructed a mutant murine P-gp with a shortened linker to facilitate structural determination. Despite dramatic reduction in rhodamine 123 and calcein-AM transport, the linker-shortened mutant P-gp possesses basal ATPase activity and binds ATP only in its N-terminal nucleotide-binding domain. Nine independently determined structures of wild type, the linker mutant, and a methylated P-gp at up to 3.3 Å resolution display significant movements of individual transmembrane domain helices, which correlated with the opening and closing motion of the two halves of P-gp. The open-and-close motion alters the surface topology of P-gp within the drug-binding pocket, providing a mechanistic explanation for the polyspecificity of P-gp in substrate interactions.


Subject(s)
Mutation, Missense , Rhodamine 123/chemistry , ATP Binding Cassette Transporter, Subfamily B/chemistry , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Amino Acid Substitution , Binding Sites , Biological Transport, Active/physiology , Crystallography, X-Ray , HeLa Cells , Humans , Rhodamine 123/metabolism , Substrate Specificity/physiology
10.
Biochemistry ; 55(7): 1010-23, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26820614

ABSTRACT

The efflux transporter P-glycoprotein (P-gp) plays a vital role in the transport of molecules across cell membranes and has been shown to interact with a panoply of functionally and structurally unrelated compounds. How human P-gp interacts with this large number of drugs has not been well understood, although structural flexibility has been implicated. To gain insight into this transporter's broad substrate specificity and to assess its ability to accommodate a variety of molecular and structural changes, we generated human-mouse P-gp chimeras by the exchange of homologous transmembrane and nucleotide-binding domains. High-level expression of these chimeras by BacMam- and baculovirus-mediated transduction in mammalian (HeLa) and insect cells, respectively, was achieved. There were no detectable differences between wild-type and chimeric P-gp in terms of cell surface expression, ability to efflux the P-gp substrates rhodamine 123, calcein-AM, and JC-1, or to be inhibited by the substrate cyclosporine A and the inhibitors tariquidar and elacridar. Additionally, expression of chimeric P-gp was able to confer a paclitaxel-resistant phenotype to HeLa cells characteristic of P-gp-mediated drug resistance. P-gp ATPase assays and photo-cross-linking with [(125)I]iodoarylazidoprazosin confirmed that transport and biochemical properties of P-gp chimeras were similar to those of wild-type P-gp, although differences in drug binding were detected when human and mouse transmembrane domains were combined. Overall, chimeras with one or two mouse P-gp domains were deemed functionally equivalent to human wild-type P-gp, demonstrating the ability of human P-gp to tolerate major structural changes.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Fluorescent Dyes/metabolism , Models, Molecular , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/chemistry , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Antineoplastic Agents/pharmacology , Biological Transport/drug effects , Cell Line , Cell Line, Transformed , Drug Resistance, Neoplasm , HeLa Cells , Humans , Kinetics , Lepidoptera , Membrane Transport Modulators/pharmacology , Mice , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
11.
PLoS One ; 10(8): e0136396, 2015.
Article in English | MEDLINE | ID: mdl-26309032

ABSTRACT

The efflux transporter P-glycoprotein (P-gp) is an important mediator of various pharmacokinetic parameters, being expressed at numerous physiological barriers and also in multidrug-resistant cancer cells. Molecular cloning of homologous cDNAs is an important tool for the characterization of functional differences in P-gp between species. However, plasmids containing mouse mdr1a cDNA display significant genetic instability during cloning in bacteria, indicating that mdr1a cDNA may be somehow toxic to bacteria, allowing only clones containing mutations that abrogate this toxicity to survive transformation. We demonstrate here the presence of a cryptic promoter in mouse mdr1a cDNA that causes mouse P-gp expression in bacteria. This expression may account for the observed toxicity of mdr1a DNA to bacteria. Sigma 70 binding site analysis and GFP reporter plasmids were used to identify sequences in the first 321 bps of mdr1a cDNA capable of initiating bacterial protein expression. An mdr1a M107L cDNA containing a single residue mutation at the proposed translational start site was shown to allow sub-cloning of mdr1a in E. coli while retaining transport properties similar to wild-type P-gp. This mutant mdr1a cDNA may prove useful for efficient cloning of mdr1a in E. coli.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , DNA, Complementary/genetics , Escherichia coli/growth & development , Promoter Regions, Genetic/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Binding Sites , Blotting, Western , DNA-Directed RNA Polymerases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Flow Cytometry , HEK293 Cells , Humans , Mice , Microscopy, Confocal , Plasmids/genetics , Protein Conformation , Sigma Factor/metabolism , Spectrometry, Fluorescence
12.
J Biol Chem ; 289(31): 21473-89, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24930045

ABSTRACT

Multidrug resistance (MDR) is a major obstacle to the successful chemotherapy of cancer. MDR is often the result of overexpression of ATP-binding cassette transporters following chemotherapy. A common ATP-binding cassette transporter that is overexpressed in MDR cancer cells is P-glycoprotein, which actively effluxes drugs against a concentration gradient, producing an MDR phenotype. Collateral sensitivity (CS), a phenomenon of drug hypersensitivity, is defined as the ability of certain compounds to selectively target MDR cells, but not the drug-sensitive parent cells from which they were derived. The drug tiopronin has been previously shown to elicit CS. However, unlike other CS agents, the mechanism of action was not dependent on the expression of P-glycoprotein in MDR cells. We have determined that the CS activity of tiopronin is mediated by the generation of reactive oxygen species (ROS) and that CS can be reversed by a variety of ROS-scavenging compounds. Specifically, selective toxicity of tiopronin toward MDR cells is achieved by inhibition of glutathione peroxidase (GPx), and the mode of inhibition of GPx1 by tiopronin is shown in this report. Why MDR cells are particularly sensitive to ROS is discussed, as is the difficulty in exploiting this hypersensitivity to tiopronin in the clinic.


Subject(s)
Enzyme Inhibitors/pharmacology , Glutathione Peroxidase/antagonists & inhibitors , Tiopronin/pharmacology , Amino Acid Sequence , Base Sequence , Cell Line, Tumor , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Glutathione Peroxidase/chemistry , Humans , Molecular Sequence Data , Oligodeoxyribonucleotides , Reactive Oxygen Species/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , Thiomalates/pharmacology
13.
Mol Pharm ; 11(8): 2692-702, 2014 Aug 04.
Article in English | MEDLINE | ID: mdl-24800945

ABSTRACT

Acquired drug resistance in cancer continues to be a challenge in cancer therapy, in part due to overexpression of the drug efflux transporter P-glycoprotein (P-gp, MDR1, ABCB1). NSC73306 is a thiosemicarbazone compound that displays greater toxicity against cells expressing functional P-gp than against other cells. Here, we investigate the cellular uptake of NSC73306, and examine its interaction with P-gp and copper transporter 1 (CTR1, SLC31A1). Overexpression of P-gp sensitizes LLC-PK1 cells to NSC73306. Cisplatin (IC50 = 77 µM), cyclosporin A (IC50 = 500 µM), and verapamil (IC50 = 700 µM) inhibited cellular accumulation of [(3)H]NSC73306. Cellular hypertoxicity of NSC73306 to P-gp-expressing cells was inhibited by cisplatin in a dose-dependent manner. Cells transiently expressing the cisplatin uptake transporter CTR1 (SLC31A1) showed increased [(3)H]NSC73306 accumulation. In contrast, CTR1 knockdown decreased [(3)H]NSC73306 accumulation. The presence of NSC73306 reduced CTR1 levels, similar to the negative feedback of CTR1 levels by copper or cisplatin. Surprisingly, although cisplatin is a substrate of CTR1, we found that CTR1 protein was overexpressed in high-level cisplatin-resistant KB-CP20 and BEL7404-CP20 cell lines. We confirmed that the CTR1 protein was functional, as uptake of NSC73306 was increased in KB-CP20 cells compared to their drug-sensitive parental cells, and downregulation of CTR1 in KB-CP20 cells reduced [(3)H]NSC73306 accumulation. These results suggest that NSC73306 is a transport substrate of CTR1.


Subject(s)
Cation Transport Proteins/metabolism , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Indoles/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , COS Cells , Chlorocebus aethiops , Cisplatin/administration & dosage , Cisplatin/chemistry , Copper/chemistry , Copper Transporter 1 , Cyclosporine/chemistry , HEK293 Cells , Humans , Inhibitory Concentration 50 , LLC-PK1 Cells , RNA, Small Interfering/metabolism , Reproducibility of Results , Swine , Thiosemicarbazones/chemistry , Verapamil/administration & dosage
14.
Drug Resist Updat ; 15(1-2): 98-105, 2012.
Article in English | MEDLINE | ID: mdl-22483810

ABSTRACT

While chemotherapy remains the most effective treatment for disseminated tumors, acquired or intrinsic drug resistance accounts for approximately 90% of treatment failure. Multidrug resistance (MDR), the simultaneous resistance to drugs that differ both structurally and mechanistically, often results from drug efflux pumps in the cell membrane that reduce intracellular drug levels to less than therapeutic concentrations. Expression of the MDR transporter P-glycoprotein (P-gp, MDR1, ABCB1) has been shown to correlate with overall poor chemotherapy response and prognosis. This review will focus on collateral sensitivity (CS), the ability of compounds to kill MDR cells selectively over the parental cells from which they were derived. Insights into CS may offer an alternative strategy for the clinical resolution of MDR, as highly selective and potent CS agents may lead to drugs that are effective at MDR cell killing and tumor resensitization. Four main mechanistic hypotheses for CS will be reviewed, followed by a discussion on quantitative and experimental evaluation of CS.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Neoplasms/drug therapy , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Biological Transport/drug effects , Calcium Channel Blockers/therapeutic use , Cell Membrane/drug effects , Cell Membrane/metabolism , Energy Metabolism/drug effects , Humans , Neoplasms/metabolism , Neoplasms/pathology , Prognosis , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Reactive Oxygen Species/metabolism , Verapamil/therapeutic use
15.
J Med Chem ; 54(16): 5878-89, 2011 Aug 25.
Article in English | MEDLINE | ID: mdl-21721528

ABSTRACT

Cancer multidrug resistance (MDR) mediated by ATP-binding cassette (ABC) transporters presents a significant unresolved clinical challenge. One strategy to resolve MDR is to develop compounds that selectively kill cells overexpressing the efflux transporter P-glycoprotein (MDR1, P-gp, ABCB1). We have previously reported structure-activity studies based around the lead compound NSC73306 (1, 1-isatin-4-(4'-methoxyphenyl)-3-thiosemicarbazone, 4.3-fold selective). Here we sought to extend this work on MDR1-selective analogues by establishing whether 1 showed "robust" activity against a range of cell lines expressing P-gp. We further aimed to synthesize and test analogues with varied substitution at the N4-position, and substitution around the N4-phenyl ring of isatin-ß-thiosemicarbazones (IBTs), to identify compounds with increased MDR1-selectivity. Compound 1 demonstrated MDR1-selectivity against all P-gp-expressing cell lines examined. This selectivity was reversed by inhibitors of P-gp ATPase activity. Structural variation at the 4'-phenyl position of 1 yielded compounds of greater MDR1-selectivity. Two of these analogues, 1-isatin-4-(4'-nitrophenyl)-3-thiosemicarbazone (22, 8.3-fold selective) and 1-isatin-4-(4'-tert-butyl phenyl)-3-thiosemicarbazone (32, 14.8-fold selective), were selected for further testing and were found to retain the activity profile of 1. These compounds are the most active IBTs identified to date.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Drug Resistance, Multiple/drug effects , Isatin/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , CHO Cells , Cell Line, Tumor , Cell Survival/drug effects , Cricetinae , Cricetulus , Crystallography, X-Ray , HEK293 Cells , Humans , Indoles/chemical synthesis , Indoles/chemistry , Indoles/pharmacology , Inhibitory Concentration 50 , Isatin/chemical synthesis , Isatin/pharmacology , Mice , Models, Chemical , Molecular Structure , NIH 3T3 Cells , Structure-Activity Relationship
16.
J Med Chem ; 54(14): 4987-97, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21657271

ABSTRACT

A major challenge in the treatment of cancer is multidrug resistance (MDR) that develops during chemotherapy. Here we demonstrate that tiopronin (1), a thiol-substituted N-propanoylglycine derivative, was selectively toxic to a series of cell lines expressing the drug efflux pump P-glycoprotein (P-gp, ABCB1) and MRP1 (ABCC1). Treatment of MDR cells with 1 led to instability of the ABCB1 mRNA and consequently a reduction in P-gp protein, despite functional assays demonstrating that tiopronin does not interact with P-gp. Long-term exposure of P-gp-expressing cells to 1 sensitized them to doxorubicin and paclitaxel, both P-gp substrates. Treatment of MRP1-overexpressing cells with tiopronin led to a significant reduction in MRP1 protein. Synthesis and screening of analogues of tiopronin demonstrated that the thiol functional group was essential for collateral sensitivity while substitution of the amino acid backbone altered but did not destroy specificity, pointing to future development of targeted analogues.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Tiopronin/pharmacology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , HEK293 Cells , Humans , Multidrug Resistance-Associated Proteins/metabolism , Orphan Drug Production , Paclitaxel/pharmacology , RNA Stability , RNA, Messenger/metabolism , Structure-Activity Relationship , Tiopronin/chemical synthesis , Tiopronin/chemistry
17.
Org Lett ; 13(13): 3518-21, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21644548

ABSTRACT

Extracts of the sponge genus Candidaspongia showed selective cytotoxicity toward melanoma cells in the NCI 60-cell-line screen. Continued investigation of the Candidaspongia sp. extracts led to the isolation of three new tedanolide analogues, precandidaspongiolides A (1) and B (2) and candidaspongiolide B (4), as well as candidaspongiolide A (3) and tedanolide (5). Semisynthetic derivatives were also generated to develop SAR. Candidaspongiolides A/B were the most potent and showed low nanomolar activity against several melanoma cell lines.


Subject(s)
Cell Proliferation/drug effects , Macrolides/chemistry , Macrolides/chemical synthesis , Melanoma/pathology , Cell Line, Tumor , Humans , Macrolides/pharmacology , Structure-Activity Relationship
18.
Vet Immunol Immunopathol ; 122(1-2): 182-7, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18068810

ABSTRACT

Human patients with atopic dermatitis (AD) commonly exhibit IgE reactivity to cutaneous self-antigens. The presence of serum IgE autoantibodies appears to correlate with disease severity, and it is suspected to reflect or contribute to tissue damage. The objective of this study was to determine whether IgE autoantibodies specific for cutaneous antigens could be detected in the serum of dogs with AD. Serum was collected from 19 dogs with untreated moderate to severe AD and four specific-pathogen free (SPF) dogs. Indirect immunofluorescence was performed using normal canine skin collected at four different locations (concave ear, nose, medial thigh and lateral thorax), while Western immunoblotting was done using normal canine ear pinna epidermal and dermal extracts and reducing conditions. In both methods, IgE was detected using a monoclonal antibody specific for heat stable epitopes of canine IgE. At 1:10 dilution, specific IgE autoantibodies against cutaneous autoantigens were not detected, with either method, in AD and SPF canine sera. Either IgE autoreactivity is not associated with moderate to severe AD in dogs, or the methods employed herein were not sensitive enough to permit IgE autoantibody detection.


Subject(s)
Autoantibodies/blood , Dermatitis, Atopic/veterinary , Dog Diseases/immunology , Immunoglobulin E/blood , Skin/immunology , Animals , Dogs , Female , Fluorescent Antibody Technique, Indirect , Immunoblotting , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...