Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
PLoS One ; 18(5): e0285323, 2023.
Article in English | MEDLINE | ID: mdl-37141324

ABSTRACT

Monocytes contribute to the pro-inflammatory immune response during the blood stage of a Plasmodium falciparum infection, but their precise role in malaria pathology is not clear. Besides phagocytosis, monocytes are activated by products from P. falciparum infected erythrocytes (IE) and one of the activation pathways is potentially the NLR family pyrin domain containing 3 (NLRP3) inflammasome, a multi-protein complex that leads to the production of interleukin (IL)-1ß. In cerebral malaria cases, monocytes accumulate at IE sequestration sites in the brain microvascular and the locally produced IL-1ß, or other secreted molecules, could contribute to leakage of the blood-brain barrier. To study the activation of monocytes by IE within the brain microvasculature in an in vitro model, we co-cultured IT4var14 IE and the monocyte cell line THP-1 for 24 hours and determined whether generated soluble molecules affect barrier function of human brain microvascular endothelial cells, measured by real time trans-endothelial electrical resistance. The medium produced after co-culture did not affect endothelial barrier function and similarly no effect was measured after inducing oxidative stress by adding xanthine oxidase to the co-culture. While IL-1ß does decrease barrier function, barely any IL-1ß was produced in the co- cultures, indicative of a lack of or incomplete THP-1 activation by IE in this co-culture model.


Subject(s)
Malaria, Falciparum , Plasmodium falciparum , Humans , Monocytes/metabolism , Coculture Techniques , Endothelial Cells/metabolism , Inflammasomes/metabolism , Erythrocytes/metabolism , Cell Line , Brain/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Interleukin-1beta/metabolism
2.
Nat Commun ; 10(1): 3226, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31324806

ABSTRACT

Primaquine (PQ) is an essential antimalarial drug but despite being developed over 70 years ago, its mode of action is unclear. Here, we demonstrate that hydroxylated-PQ metabolites (OH-PQm) are responsible for efficacy against liver and sexual transmission stages of Plasmodium falciparum. The antimalarial activity of PQ against liver stages depends on host CYP2D6 status, whilst OH-PQm display direct, CYP2D6-independent, activity. PQ requires hepatic metabolism to exert activity against gametocyte stages. OH-PQm exert modest antimalarial efficacy against parasite gametocytes; however, potency is enhanced ca.1000 fold in the presence of cytochrome P450 NADPH:oxidoreductase (CPR) from the liver and bone marrow. Enhancement of OH-PQm efficacy is due to the direct reduction of quinoneimine metabolites by CPR with the concomitant and excessive generation of H2O2, leading to parasite killing. This detailed understanding of the mechanism paves the way to rationally re-designed 8-aminoquinolines with improved pharmacological profiles.


Subject(s)
Antimalarials/pharmacology , Plasmodium falciparum/drug effects , Primaquine/metabolism , Primaquine/pharmacology , Aminoquinolines/pharmacology , Bone Marrow/metabolism , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 Enzyme System , Dose-Response Relationship, Drug , Humans , Hydrogen Peroxide/metabolism , Liver/metabolism , Malaria, Falciparum/drug therapy , NADP , Pharmacokinetics
3.
Cell Microbiol ; 21(7): e13028, 2019 07.
Article in English | MEDLINE | ID: mdl-30941868

ABSTRACT

The Plasmodium subtilisin-like serine protease SUB1 is expressed in hepatic and both asexual and sexual blood parasite stages. SUB1 is required for egress of invasive forms of the parasite from both erythrocytes and hepatocytes, but its subcellular localisation, function, and potential substrates in the sexual stages are unknown. Here, we have characterised the expression profile and subcellular localisation of SUB1 in Plasmodium berghei sexual stages. We show that the protease is selectively expressed in mature male gametocytes and localises to secretory organelles known to be involved in gamete egress, called male osmiophilic bodies. We have investigated PbSUB1 function in the sexual stages by generating P. berghei transgenic lines deficient in PbSUB1 expression or enzyme activity in gametocytes. Our results demonstrate that PbSUB1 plays a role in male gamete egress. We also show for the first time that the PbSUB1 substrate PbSERA3 is expressed in gametocytes and processed by PbSUB1 upon gametocyte activation. Taken together, our results strongly suggest that PbSUB1 is not only a promising drug target for asexual stages but could also be an attractive malaria transmission-blocking target.


Subject(s)
Malaria/genetics , Plasmodium berghei/genetics , Serine Endopeptidases/genetics , Subtilisins/genetics , Animals , Erythrocytes/parasitology , Germ Cells/parasitology , Hepatocytes/parasitology , Malaria/parasitology , Male , Organelles/parasitology , Plasmodium berghei/pathogenicity , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity
4.
Mol Microbiol ; 104(2): 306-318, 2017 04.
Article in English | MEDLINE | ID: mdl-28118506

ABSTRACT

The goal to prevent Plasmodium falciparum transmission from humans to mosquitoes requires the identification of targetable metabolic processes in the mature (stage V) gametocytes, the sexual stages circulating in the bloodstream. This task is complicated by the apparently low metabolism of these cells, which renders them refractory to most antimalarial inhibitors and constrains the development of specific and sensitive cell-based assays. Here, we identify and functionally characterize the regulatory regions of the P. falciparum gene PF3D7_1234700, encoding a CPW-WPC protein and named here Upregulated in Late Gametocytes (ULG8), which we have leveraged to express reporter genes in mature male and female gametocytes. Using transgenic parasites containing a pfULG8-luciferase cassette, we investigated the susceptibility of stage V gametocytes to compounds specifically affecting redox metabolism. Our results reveal a high sensitivity of mature gametocytes to the glutathione reductase inhibitor and redox cycler drug methylene blue (MB). Using isobologram analysis, we find that a concomitant inhibition of the parasite enzyme glucose-6-phosphate dehydrogenase-6-phosphogluconolactonase, a key component of NADPH synthesis, potently synergizes MB activity. These data suggest that redox metabolism and detoxification activity play an unsuspected yet vital role in stage V gametocytes, rendering these cells exquisitely sensitive to decreases in NADPH concentration.


Subject(s)
Plasmodium falciparum/drug effects , Antimalarials/pharmacology , Carboxylic Ester Hydrolases/metabolism , Carboxylic Ester Hydrolases/physiology , Gene Expression Regulation , Genes, Reporter , Glucosephosphate Dehydrogenase/metabolism , Glucosephosphate Dehydrogenase/physiology , Luciferases , Multienzyme Complexes/metabolism , Multienzyme Complexes/physiology , Oxidation-Reduction/drug effects , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Plasmodium falciparum/physiology
5.
PLoS Pathog ; 12(7): e1005763, 2016 07.
Article in English | MEDLINE | ID: mdl-27467575

ABSTRACT

A major cause of the paucity of new starting points for drug discovery is the lack of interaction between academia and industry. Much of the global resource in biology is present in universities, whereas the focus of medicinal chemistry is still largely within industry. Open source drug discovery, with sharing of information, is clearly a first step towards overcoming this gap. But the interface could especially be bridged through a scale-up of open sharing of physical compounds, which would accelerate the finding of new starting points for drug discovery. The Medicines for Malaria Venture Malaria Box is a collection of over 400 compounds representing families of structures identified in phenotypic screens of pharmaceutical and academic libraries against the Plasmodium falciparum malaria parasite. The set has now been distributed to almost 200 research groups globally in the last two years, with the only stipulation that information from the screens is deposited in the public domain. This paper reports for the first time on 236 screens that have been carried out against the Malaria Box and compares these results with 55 assays that were previously published, in a format that allows a meta-analysis of the combined dataset. The combined biochemical and cellular assays presented here suggest mechanisms of action for 135 (34%) of the compounds active in killing multiple life-cycle stages of the malaria parasite, including asexual blood, liver, gametocyte, gametes and insect ookinete stages. In addition, many compounds demonstrated activity against other pathogens, showing hits in assays with 16 protozoa, 7 helminths, 9 bacterial and mycobacterial species, the dengue fever mosquito vector, and the NCI60 human cancer cell line panel of 60 human tumor cell lines. Toxicological, pharmacokinetic and metabolic properties were collected on all the compounds, assisting in the selection of the most promising candidates for murine proof-of-concept experiments and medicinal chemistry programs. The data for all of these assays are presented and analyzed to show how outstanding leads for many indications can be selected. These results reveal the immense potential for translating the dispersed expertise in biological assays involving human pathogens into drug discovery starting points, by providing open access to new families of molecules, and emphasize how a small additional investment made to help acquire and distribute compounds, and sharing the data, can catalyze drug discovery for dozens of different indications. Another lesson is that when multiple screens from different groups are run on the same library, results can be integrated quickly to select the most valuable starting points for subsequent medicinal chemistry efforts.


Subject(s)
Antimalarials/therapeutic use , Datasets as Topic , Drug Discovery/methods , Malaria/drug therapy , Neglected Diseases/drug therapy , Drug Evaluation, Preclinical , Humans , Small Molecule Libraries
6.
J Antimicrob Chemother ; 71(5): 1148-58, 2016 May.
Article in English | MEDLINE | ID: mdl-26888912

ABSTRACT

OBJECTIVES: As most available antimalarial drugs are ineffective against the Plasmodium falciparum transmission stages, new drugs against the parasite's gametocytes are urgently needed to combat malaria globally. The unique biology of gametocytes requires assays that need to be specific, to faithfully monitor anti-gametocyte activity, and to be easy to perform, cheap and scalable to high-throughput screening (HTS). METHODS: We developed an HTS cell-based assay with P. falciparum gametocytes specifically expressing a potent luciferase. To confirm HTS hit activity for several parasite genotypes, the luciferase assay and the gametocyte lactate dehydrogenase (LDH) assay, usable on any parasite isolate, were compared by screening antimalarial drugs and determining IC50 values of anti-gametocyte hits from the 'Malaria Box' against early- and late-stage gametocytes. RESULTS: Comparison of the two assays, conducted on the early and on late gametocyte stages, revealed an excellent correlation (R(2) > 0.9) for the IC50 values obtained by the respective readouts. Differences in susceptibility to drugs and compounds between the two parasite developmental stages were consistently measured in both assays. CONCLUSIONS: This work indicates that the luciferase and gametocyte LDH assays are interchangeable and that their specific advantages can be exploited to design an HTS pipeline leading to new transmission-blocking compounds. Results from these assays consistently defined a gametocyte chemical susceptibility profile, relevant to the planning of future drug discovery strategies.


Subject(s)
Antimalarials/pharmacology , Drug Evaluation, Preclinical/methods , Plasmodium falciparum/drug effects , Cytological Techniques/methods , Genes, Reporter , High-Throughput Screening Assays/methods , Humans , Inhibitory Concentration 50 , L-Lactate Dehydrogenase/analysis , Luciferases/analysis , Plasmodium falciparum/enzymology , Staining and Labeling
7.
Anal Chem ; 86(17): 8814-21, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25102353

ABSTRACT

New reliable and cost-effective antimalarial drug screening assays are urgently needed to identify drugs acting on different stages of the parasite Plasmodium falciparum, and particularly those responsible for human-to-mosquito transmission, that is, the P. falciparum gametocytes. Low Z' factors, narrow dynamic ranges, and/or extended assay times are commonly reported in current gametocyte assays measuring gametocyte-expressed fluorescent or luciferase reporters, endogenous ATP levels, activity of gametocyte enzymes, or redox-dependent dye fluorescence. We hereby report on a dual-luciferase gametocyte assay with immature and mature P. falciparum gametocyte stages expressing red and green-emitting luciferases from Pyrophorus plagiophthalamus under the control of the parasite sexual stage-specific pfs16 gene promoter. The assay was validated with reference antimalarial drugs and allowed to quantitatively and simultaneously measure stage-specific drug effects on parasites at different developmental stages. The optimized assay, requiring only 48 h incubation with drugs and using a cost-effective luminogenic substrate, significantly reduces assay cost and time in comparison to state-of-the-art analogous assays. The assay had a Z' factor of 0.71 ± 0.03, and it is suitable for implementation in 96- and 384-well microplate formats. Moreover, the use of a nonlysing D-luciferin substrate significantly improved the reliability of the assay and allowed one to perform, for the first time, P. falciparum bioluminescence imaging at single-cell level.


Subject(s)
Luminescent Measurements , Microscopy, Video , Parasitology/methods , Plasmodium falciparum/isolation & purification , Antimalarials/pharmacology , Cell Line , Fluorescent Antibody Technique , Humans , Luciferases/genetics , Luciferases/metabolism , Plasmids/genetics , Plasmids/metabolism , Plasmodium falciparum/drug effects , Plasmodium falciparum/metabolism , Promoter Regions, Genetic , Protozoan Proteins/genetics , Single-Cell Analysis
8.
J Proteome Res ; 11(11): 5323-37, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23025827

ABSTRACT

The asexual blood stages of Plasmodium falciparum cause the most lethal form of human malaria. During growth within an infected red blood cell, parasite multiplication and formation of invasive merozoites is called schizogony. Here, we present a detailed analysis of the phosphoproteome of P. falciparum schizonts revealing 2541 unique phosphorylation sites, including 871 novel sites. Prominent roles for cAMP-dependent protein kinase A- and phosphatidylinositol-signaling were identified following analysis by functional enrichment, phosphoprotein interaction network clustering and phospho-motif identification tools. We observed that most key enzymes in the inositol pathway are phosphorylated, which strongly suggests additional levels of regulation and crosstalk with other protein kinases that coregulate different biological processes. A distinct pattern of phosphorylation of proteins involved in merozoite egress and red blood cell invasion was noted. The analyses also revealed that cAMP-PKA signaling is implicated in a wide variety of processes including motility. We verified this finding experimentally using an in vitro kinase assay and identified three novel PKA substrates associated with the glideosome motor complex: myosin A, GAP45 and CDPK1. Therefore, in addition to an established role for CDPK1 in the motor complex, this study reveals the coinvolvement of PKA, further implicating cAMP as an important regulator of host cell invasion.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Phosphatidylinositols/metabolism , Phosphoproteins/metabolism , Plasmodium falciparum/metabolism , Proteome , Protozoan Proteins/metabolism , Signal Transduction , Animals , Chromatography, Liquid , Electrophoresis, Polyacrylamide Gel , Tandem Mass Spectrometry
9.
Mol Cell Proteomics ; 9(7): 1437-48, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20332084

ABSTRACT

Despite over a century of study of malaria parasites, parts of the Plasmodium falciparum life cycle remain virtually unknown. One of these is the early gametocyte stage, a round shaped cell morphologically similar to an asexual trophozoite in which major cellular transformations ensure subsequent development of the elongated gametocyte. We developed a protocol to obtain for the first time highly purified preparations of early gametocytes using a transgenic line expressing a green fluorescent protein from the onset of gametocytogenesis. We determined the cellular proteome (1427 proteins) of this parasite stage by high accuracy tandem mass spectrometry and newly determined the proteomes of asexual trophozoites and mature gametocytes, identifying altogether 1090 previously undetected parasite proteins. Quantitative label-free comparative proteomics analysis determined enriched protein clusters for the three parasite developmental stages. Gene set enrichment analysis on the 251 proteins enriched in the early gametocyte proteome revealed that proteins putatively exported and involved in erythrocyte remodeling are the most overrepresented protein set in these stages. One-tenth of the early gametocyte-enriched proteome is constituted of putatively exported proteins, here named PfGEXPs (P. falciparum gametocyte-exported proteins). N-terminal processing and N-acetylation at a conserved leucine residue within the Plasmodium export element pentamotif were detected by mass spectrometry for three such proteins in the early but not in the mature gametocyte sample, further supporting a specific role in protein export in early gametocytogenesis. Previous reports and results of our experiments confirm that the three proteins are indeed exported in the erythrocyte cytoplasm. This work indicates that protein export profoundly marks early sexual differentiation in P. falciparum, probably contributing to host cell remodeling in this phase of the life cycle, and that gametocyte-enriched molecules are recruited to modulate this process in gametocytogenesis.


Subject(s)
Plasmodium falciparum/cytology , Plasmodium falciparum/metabolism , Plasmodium falciparum/physiology , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , Female , Humans , Life Cycle Stages/physiology , Malaria, Falciparum , Male , Molecular Sequence Data , Plasmodium falciparum/pathogenicity , Proteome/analysis , Tandem Mass Spectrometry/methods
10.
Int J Parasitol ; 40(6): 663-73, 2010 May.
Article in English | MEDLINE | ID: mdl-19968995

ABSTRACT

Gametocytes of the protozoan Plasmodium falciparum ensure malaria parasite transmission from humans to the insect vectors. In their development, they produce the abundant specific protein Pfg27, the function and in vivo molecular interactions of which are unknown. Here we reveal a previously unreported localisation of Pfg27 in the gametocyte nucleus by immunoelectron microscopy and studies with HaloTag and Green Fluorescent Protein fusions, and identify a network of interactions established by the protein during gametocyte development. We report the ability of endogenous Pfg27 to form oligomeric complexes that are affected by phosphorylation of the protein, possibly through the identified phosphorylation sites, Ser32 and Thr208. We show that Pfg27 binds RNA molecules through specific residues and that the protein interacts with parasite RNA-binding proteins such as EF1alpha and PfH45. We propose a structural model for Pfg27 oligomerisation, based on the sequence and structural conservation here recognised between Pfg27 and sterile alpha motif. This study provides a molecular basis for Pfg27 to establish an interaction network with RNA and RNA-binding proteins and to govern its dynamic oligomerisation in developing gametocytes.


Subject(s)
Plasmodium falciparum/growth & development , Protein Multimerization , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , Cell Nucleus/chemistry , Microscopy, Fluorescence , Microscopy, Immunoelectron , Models, Molecular , Molecular Sequence Data , Phosphorylation , Protein Binding , Protein Interaction Mapping , Protein Processing, Post-Translational , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , RNA, Protozoan/metabolism
11.
Mol Microbiol ; 73(2): 180-93, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19570101

ABSTRACT

In the human malaria parasite Plasmodium falciparum, gametocyte maturation is a process remarkably longer than in other malaria species, accompanied by expression of 2-300 sexual stage-specific proteins. Disruption of several of their encoding genes so far showed that only the abundant protein Pfg27, produced at the onset of sexual differentiation, is essential for gametocyte production. In contrast with what has been previously described, here we show that P. falciparum pfg27 disruptant lines are able to undergo all stages of gametocyte maturation, and are able to mature into gametes. A fraction of Pfg27-defective gametocytes show, however, distinct abnormalities in intra- and extra-cellular membranous compartments, such as accumulation of parasitophorous vacuole-derived vesicles in the erythrocyte cytoplasm, large intracellular vacuoles and discontinuities in their trilaminar cell membrane. This work revises current knowledge on the role of Pfg27, indicating that the protein is not required for parasite entry into sexual differentiation, and suggesting that it is instead involved in maintaining cell integrity in the uniquely long gametocytogenesis of P. falciparum.


Subject(s)
Antigens, Protozoan/metabolism , Germ Cells/growth & development , Plasmodium falciparum/genetics , Protozoan Proteins/metabolism , Aedes/parasitology , Animals , DNA, Protozoan/genetics , Gene Expression Regulation, Developmental , Genes, Protozoan , Germ Cells/ultrastructure , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Plasmodium falciparum/ultrastructure , Protozoan Proteins/genetics , Sequence Analysis, DNA , Transfection
12.
Cell Microbiol ; 11(8): 1272-88, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19438517

ABSTRACT

Malaria parasites invade erythrocytes of their host both for asexual multiplication and for differentiation to male and female gametocytes - the precursor cells of Plasmodium gametes. For further development the parasite is dependent on efficient release of the asexual daughter cells and of the gametes from the host erythrocyte. How malarial parasites exit their host cells remains largely unknown. We here report the characterization of a Plasmodium berghei protein that is involved in egress of both male and female gametes from the host erythrocyte. Protein MDV-1/PEG3, like its Plasmodium falciparum orthologue, is present in gametocytes of both sexes, but more abundant in the female, where it is associated with dense granular organelles, the osmiophilic bodies. Deltamdv-1/peg3 parasites in which MDV-1/PEG3 production was abolished by gene disruption had a strongly reduced capacity to form zygotes resulting from a reduced capability of both the male and female gametes to disrupt the surrounding parasitophorous vacuole and to egress from the host erythrocyte. These data demonstrate that emergence from the host cell of male and female gametes relies on a common, MDV-1/PEG3-dependent mechanism that is distinct from mechanisms used by asexual parasites.


Subject(s)
Erythrocytes/metabolism , Germ Cells/physiology , Plasmodium berghei/physiology , Protozoan Proteins/metabolism , Animals , Anopheles , Female , Fertilization , Genes, Protozoan , Host-Pathogen Interactions , Malaria/metabolism , Malaria/parasitology , Male , Mice , Microscopy, Electron, Transmission , Plasmodium berghei/ultrastructure , Protozoan Proteins/chemistry , Sequence Analysis, Protein , Sex Factors
13.
Mol Microbiol ; 67(2): 278-90, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18086189

ABSTRACT

Osmiophilic bodies are membrane-bound vesicles, found predominantly in Plasmodium female gametocytes, that become progressively more abundant as the gametocyte reaches full maturity. These vesicles lie beneath the subpellicular membrane of the gametocyte, and the release of their contents into the parasitophorous vacuole has been postulated to aid in the escape of gametocytes from the erythrocyte after ingestion by the mosquito. Currently, the only protein known to be associated with osmiophilic bodies in Plasmodium falciparum is Pfg377, a gametocyte-specific protein expressed at the onset of osmiophilic body development. Here we show by targeted gene disruption that Pfg377 plays a fundamental role in the formation of these organelles, and that female gametocytes lacking the full complement of osmiophilic bodies are significantly less efficient both in vitro and in vivo in their emergence from the erythrocytes upon induction of gametogenesis, a process whose timing is critical for fertilization with the short-lived male gamete. This reduced efficiency of emergence explains the significant defect in oocyst formation in mosquitoes fed blood meals containing Pfg377-negative gametocytes, resulting in an almost complete blockade of infection.


Subject(s)
Culicidae/parasitology , Germ Cells/physiology , Plasmodium falciparum/cytology , Plasmodium falciparum/pathogenicity , Protozoan Proteins/metabolism , Animals , Electroporation , Erythrocytes/parasitology , Female , Fluorescent Antibody Technique, Indirect , Gametogenesis , Germ Cells/ultrastructure , Humans , Male , Microscopy, Electron, Transmission , Organelles/physiology , Plasmodium falciparum/genetics , Polymerase Chain Reaction , Protozoan Proteins/genetics
14.
Int J Oncol ; 27(3): 799-806, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16077931

ABSTRACT

The development of chemoresistance is a major obstacle for successful anticancer therapy. Understanding the molecular mechanisms leading to chemoresistance is a rational step to improve the therapeutic efficacy of cytotoxic drugs. Since anthracyclines play an important role in cancer chemotherapy, we have generated a human ovarian tumor cell line resistant to sabarubicin (MEN 10755), the newest anthracycline molecule in clinical development. Expression of the transporter protein MRP that affected sabarubicin uptake, and a reduced DNA topoisomerase II content in A2780/saba cells was observed. Since the poisoning of DNA topoisomerase II results in DNA damage, which is a critical signal for NF-kappaB activation, we explored if this transcription factor has a role in the chemoresistance to anthracyclines. We showed a reduced NF-kappaB activation in the resistant cell line. Moreover, qualitative changes in NF-kappaB dimer formation between the two cell lines were observed. In agreement with the hypothesis of a role of NF-kappaB in mediating drug resistance, we showed that the pharmacological inhibition of NF-kappaB activation attenuated drug resistance in A2780/saba cells whereas it had no effect in A2780 cells. Altogether, these findings show that anthracycline resistance in A2780 cell lines is due to the coexpression of several molecular mechanisms.


Subject(s)
Anthracyclines/pharmacology , Disaccharides/pharmacology , Doxorubicin/analogs & derivatives , Drug Resistance, Neoplasm , NF-kappa B/metabolism , Anthracyclines/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , DNA Topoisomerases, Type I/metabolism , Disaccharides/pharmacokinetics , Dose-Response Relationship, Drug , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Enzyme Activation/drug effects , Etoposide/pharmacology , Female , Gene Expression/drug effects , Humans , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , NF-kappa B/antagonists & inhibitors , Nitriles/pharmacology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Sulfones/pharmacology , Time Factors , Vinblastine/pharmacology
15.
J Cell Biol ; 167(3): 417-23, 2004 Nov 08.
Article in English | MEDLINE | ID: mdl-15520231

ABSTRACT

In skeletal muscle differentiation, the retinoblastoma protein (pRb) is absolutely necessary to establish definitive mitotic arrest. It is widely assumed that pRb is equally essential to sustain the postmitotic state, but this contention has never been tested. Here, we show that terminal proliferation arrest is maintained in skeletal muscle cells by a pRb-independent mechanism. Acute Rb excision from conditional knockout myotubes caused reexpression of E2F transcriptional activity, cyclin-E and -A kinase activities, PCNA, DNA ligase I, RPA, and MCM2, but did not induce DNA synthesis, showing that pRb is not indispensable to preserve the postmitotic state of these cells. Muscle-specific gene expression was significantly down-regulated, showing that pRb is constantly required for optimal implementation of the muscle differentiation program. Rb-deleted myotubes were efficiently reactivated by forced expression of cyclin D1 and Cdk4, indicating a functionally significant target other than pRb for these molecules. Finally, Rb removal induced no DNA synthesis even in pocket-protein null cells. Thus, the postmitotic state of myotubes is maintained by at least two mechanisms, one of which is pocket-protein independent.


Subject(s)
Cell Differentiation , Mitosis , Muscle Cells/cytology , Muscle, Skeletal/cytology , Retinoblastoma Protein/physiology , Animals , Cell Cycle , Cells, Cultured , Cyclin D1/genetics , Cyclin D1/physiology , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/physiology , Down-Regulation , Gene Expression , Mice , Mice, Knockout , Muscle Cells/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology
16.
Anticancer Drugs ; 15(2): 151-6, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15075671

ABSTRACT

We describe the identification of MEN15658, a molecule characterized by a promising cytotoxic effect against human tumor cell lines, including platinum- and anthracycline-resistant ovarian carcinoma. MEN15658 induces p53 accumulation, and activation of gadd-45, p21, c-fos and bcl-2 family genes in human ovarian carcinoma A2780 cell line. The compound causes a block in S phase of the cell cycle, inducing apoptotic cell death, thus suggesting an involvement of DNA damage in the MEN15658 effect on tumor cells. The anti-tumoral activity observed against the human ovarian carcinoma A2780 cell line xenotransplanted in nude mice makes this compound a new promising anti-tumoral drug.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/drug effects , Hydrazones/pharmacology , Phenanthrolines/pharmacology , Phenanthrolines/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Division/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/drug effects , Cyclins/genetics , Cyclins/metabolism , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/physiology , Drug Screening Assays, Antitumor/methods , Female , Forecasting , Gene Expression Profiling/methods , Genes, p53/drug effects , Genes, p53/genetics , Genes, p53/physiology , Humans , Hydrazones/chemistry , Hydrazones/therapeutic use , Intracellular Signaling Peptides and Proteins , Lung Neoplasms/pathology , Mice , Mice, Nude , Ovarian Neoplasms/pathology , Phenanthrolines/chemistry , Proteins/drug effects , Proteins/genetics , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , S Phase/drug effects , Time Factors , Transplantation, Heterologous/methods , Tumor Cells, Cultured , GADD45 Proteins
17.
J Leukoc Biol ; 73(1): 201-7, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12525579

ABSTRACT

Several chemokines have been shown to regulate cellular apoptosis following discrete stimuli. It was previously demonstrated that the CC chemokine CCL1 (I-309) rescues thymic lymphoma cells from apoptosis by unknown mechanisms. The aim of our study was to characterize the role of the CC chemokine receptor 8 (CCR8), the only described receptor for CCL1, in the rescue of murine thymic lymphoma cells and murine thymocytes from dexamethasone (dex)-induced apoptosis. We show here that the CCR8-restricted agonist Kaposi sarcoma-associated herpesvirus-encoded chemokine viral macrophage-inflammatory protein-1 (vMIP-1) rescues thymic lymphoma cells from dex-induced apoptosis, similar to CCL1, and that such rescue is extracellular-regulated kinase-dependent. Although it has been hypothesized that the rescuing effect of CCL1 from apoptosis could be CCR8-mediated, here, we formally demonstrate the role of such receptor as its selective antagonist encoded by the MC148 gene of molluscum contagiosum virus MC148/vMCC-I inhibits v-MIP-1- and CCL1-induced rescue activity. In addition, CCR8 ligands inhibit dex-induced apoptosis of murine thymocytes with potential implications for thymic selection.


Subject(s)
Apoptosis/drug effects , Dexamethasone/pharmacology , Mitogen-Activated Protein Kinases/physiology , Receptors, Chemokine/physiology , Viral Proteins , Animals , Binding Sites , Calcium Signaling , Chemokine CCL1 , Chemokines, CC/antagonists & inhibitors , Chemokines, CC/metabolism , Chemotaxis, Leukocyte , Female , Humans , Lymphoma, T-Cell/pathology , Macrophage Inflammatory Proteins/pharmacology , Mice , Receptors, CCR8 , Receptors, Chemokine/agonists , Signal Transduction , Thymus Gland/cytology
18.
Int J Cancer ; 102(5): 476-82, 2002 Dec 10.
Article in English | MEDLINE | ID: mdl-12432549

ABSTRACT

The new disaccharide anthracycline MEN 10755 induces activation of both NF-kappaB and p53 transcription factors in A2780 cells. Nevertheless, pharmacologic inhibition of NF-kappaB activation does not modify the sensitivity of A2780 cells to MEN 10755 treatment. To better characterize the role of NF-kappaB in MEN 10755-induced cytotoxicity, we analyzed the expression of a number of genes that are known to be regulated by NF-kappaB. None of these genes is modified by MEN 10755 treatment. On the contrary, our results suggest that the p53 DNA damage-responsive pathway is fully activated in A2780 cells, several genes controlled by p53 being up- or downregulated according to the described action of p53 on their promoters. Thus, in the A2780 cell line, the role of p53 in transducing the DNA-damage signal appears to be relevant, whereas NF-kappaB, although activated, appears to be nonfunctional. Other human carcinoma cell lines besides A2780 activate NF-kappaB DNA binding in response to MEN 10755 treatment, but again, this binding does not always lead to target gene activation. These results suggest that other factors, tumor type-specific and different from mere activation, could influence NF-kappaB transcriptional activity. Therefore, care should be taken when considering the pharmacologic inhibition of NF-kappaB as a means to improve anticancer therapy efficacy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma/metabolism , Disaccharides/pharmacology , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , NF-kappa B/metabolism , Gene Expression Regulation, Neoplastic , Humans , NF-kappa B/antagonists & inhibitors , Transcriptional Activation , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...