Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
JAMA Oncol ; 8(5): 698-705, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35238873

ABSTRACT

Importance: The drug HD201 is a biosimilar candidate for breast cancer treatment as the reference trastuzumab. Objective: To compare the efficacy of HD201 with referent trastuzumab. Design, Setting, and Participants: This randomized clinical trial (TROIKA) included 502 women with ERBB2-positive early breast cancer treated with either HD201 or referent trastuzumab. It was conducted across 70 centers in 12 countries, including Western and Eastern Europe and Asian countries. Randomization was stratified by tumor hormone receptor status, clinical stage, and geographic region of recruitment. This analysis was conducted on February 12, 2021, after the completion of the adjuvant phase at a median of 31 months (IQR, 28-33 months) of follow-up. Interventions: Patients with ERBB2-positive early breast cancer were randomly assigned to receive HD201 or referent trastuzumab in the neoadjuvant setting for 8 cycles, concurrently with 4 cycles of docetaxel, which was followed by 4 cycles of epirubicin and cyclophosphamide. Patients then underwent surgery, which was followed by treatment with 10 cycles of adjuvant HD201 or referent trastuzumab. Main Outcome and Measures: The primary end point was the total pathological complete response (tpCR) assessed after neoadjuvant treatment. Equivalence was concluded if the 95% CI of the absolute difference in tpCR between arms in the per-protocol set was within the margin of more or less than 15%. Other objectives included the breast pathological complete response, overall response, event-free and overall survival, safety, pharmacokinetics, and immunogenicity. Results: A total of 502 female patients (mean [range] age, 53 [26-82] years) were randomized to receive either HD201 or referent trastuzumab, and 474 (94.2%) were eligible for inclusion in the per-protocol set. The baseline characteristics were well balanced between the 2 arms; 195 tumors (38.8%) were hormone receptor-negative , and 213 patients (42.4%) had clinical stage III disease. The tpCR rates were 45% and 48.7% for HD201 and referent trastuzumab, respectively. The difference between the 2 groups was not significant at -3.8% (95% CI, -12.8% to 5.4%) and fell within the predefined equivalence margins. The ratio of the tpCR rates between the 2 arms was 0.92 (95% CI, 0.76 to 1.12). A total of 433 patients (86.1%) presented with 2232 treatment-emergent adverse events of special interest for trastuzumab during the entire treatment period, with 220 (88.0%) and 213 (84.5%) patients in the HD201 and referent trastuzumab groups, respectively. Conclusions and Relevance: The results of this randomized clinical trial found that HD201 demonstrated equivalence to referent trastuzumab in terms of efficacy for the end point of tpCR, with a similar safety profile. Trial Registration: ClinicalTrials.gov Identifier: NCT03013504.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Neoadjuvant Therapy , Adult , Aged , Aged, 80 and over , Female , Humans , Middle Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Receptor, ErbB-2 , Trastuzumab/adverse effects , Trastuzumab/therapeutic use
2.
Sci Transl Med ; 13(579)2021 02 03.
Article in English | MEDLINE | ID: mdl-33536278

ABSTRACT

Dengue virus (DENV) is a mosquito-borne flavivirus that poses a threat to public health, yet no antiviral drug is available. We performed a high-throughput phenotypic screen using the Novartis compound library and identified candidate chemical inhibitors of DENV. This chemical series was optimized to improve properties such as anti-DENV potency and solubility. The lead compound, NITD-688, showed strong potency against all four serotypes of DENV and demonstrated excellent oral efficacy in infected AG129 mice. There was a 1.44-log reduction in viremia when mice were treated orally at 30 milligrams per kilogram twice daily for 3 days starting at the time of infection. NITD-688 treatment also resulted in a 1.16-log reduction in viremia when mice were treated 48 hours after infection. Selection of resistance mutations and binding studies with recombinant proteins indicated that the nonstructural protein 4B is the target of NITD-688. Pharmacokinetic studies in rats and dogs showed a long elimination half-life and good oral bioavailability. Extensive in vitro safety profiling along with exploratory rat and dog toxicology studies showed that NITD-688 was well tolerated after 7-day repeat dosing, demonstrating that NITD-688 may be a promising preclinical candidate for the treatment of dengue.


Subject(s)
Dengue Virus , Dengue , Animals , Antiviral Agents/therapeutic use , Dengue/drug therapy , Dogs , Mice , Models, Animal , Rats , Serogroup
3.
Article in English | MEDLINE | ID: mdl-29530849

ABSTRACT

Artemisinin (ART) resistance has spread through Southeast Asia, posing a serious threat to the control and elimination of malaria. ART resistance has been associated with mutations in the Plasmodium falciparum kelch-13 (Pfk13) propeller domain. Phenotypically, ART resistance is defined as delayed parasite clearance in patients due to the reduced susceptibility of early ring-stage parasites to the active metabolite of ART dihydroartemisinin (DHA). Early rings can enter a state of quiescence upon DHA exposure and resume growth in its absence. These quiescent rings are referred to as dormant rings or DHA-pretreated rings (here called dormant rings). The imidazolopiperazines (IPZ) are a novel class of antimalarial drugs that have demonstrated efficacy in early clinical trials. Here, we characterized the stage of action of the IPZ GNF179 and evaluated its activity against rings and dormant rings in wild-type and ART-resistant parasites. Unlike DHA, GNF179 does not induce dormancy. We show that GNF179 is more rapidly cidal against schizonts than against ring and trophozoite stages. However, with 12 h of exposure, the compound effectively kills rings and dormant rings of both susceptible and ART-resistant parasites within 72 h. We further demonstrate that in combination with ART, GNF179 effectively prevents recrudescence of dormant rings, including those bearing pfk13 propeller mutations.


Subject(s)
Antimalarials/pharmacology , Artemisinins/pharmacology , Imidazoles/pharmacology , Piperazines/pharmacology , Plasmodium falciparum/drug effects , Parasitic Sensitivity Tests , Plasmodium falciparum/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Schizonts/drug effects , Schizonts/metabolism , Trophozoites/drug effects , Trophozoites/metabolism
4.
ACS Infect Dis ; 4(4): 635-645, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29341586

ABSTRACT

Cryptosporidiosis is a diarrheal disease predominantly caused by Cryptosporidium parvum ( Cp) and Cryptosporidium hominis ( Ch), apicomplexan parasites which infect the intestinal epithelial cells of their human hosts. The only approved drug for cryptosporidiosis is nitazoxanide, which shows limited efficacy in immunocompromised children, the most vulnerable patient population. Thus, new therapeutics and in vitro infection models are urgently needed to address the current unmet medical need. Toward this aim, we have developed novel cytopathic effect (CPE)-based Cp and Ch assays in human colonic tumor (HCT-8) cells and compared them to traditional imaging formats. Further model validation was achieved through screening a collection of FDA-approved drugs and confirming many previously known anti- Cryptosporidium hits as well as identifying a few novel candidates. Collectively, our data reveals this model to be a simple, functional, and homogeneous gain of signal format amenable to high throughput screening, opening new avenues for the discovery of novel anticryptosporidials.


Subject(s)
Antiprotozoal Agents/isolation & purification , Cryptosporidium parvum/drug effects , Cryptosporidium parvum/growth & development , Drug Evaluation, Preclinical/methods , Epithelial Cells/parasitology , Antiprotozoal Agents/pharmacology , Cell Line , Humans
5.
Elife ; 62017 12 07.
Article in English | MEDLINE | ID: mdl-29215331

ABSTRACT

Plasmodium liver hypnozoites, which cause disease relapse, are widely considered to be the last barrier towards malaria eradication. The biology of this quiescent form of the parasite is poorly understood which hinders drug discovery. We report a comparative transcriptomic dataset of replicating liver schizonts and dormant hypnozoites of the relapsing parasite Plasmodium cynomolgi. Hypnozoites express only 34% of Plasmodium physiological pathways, while 91% are expressed in replicating schizonts. Few known malaria drug targets are expressed in quiescent parasites, but pathways involved in microbial dormancy, maintenance of genome integrity and ATP homeostasis were robustly expressed. Several transcripts encoding heavy metal transporters were expressed in hypnozoites and the copper chelator neocuproine was cidal to all liver stage parasites. This transcriptomic dataset is a valuable resource for the discovery of vaccines and effective treatments to combat vivax malaria.


Subject(s)
Gene Expression Profiling , Liver/parasitology , Macaca mulatta/parasitology , Plasmodium cynomolgi/growth & development , Plasmodium cynomolgi/genetics , Schizonts/growth & development , Schizonts/genetics , Animals , Female , Male
6.
SLAS Discov ; 22(9): 1106-1119, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28731783

ABSTRACT

The intramembrane protease signal peptide peptidase-like 2a (SPPL2a) is a potential drug target for the treatment of autoimmune diseases due to an essential role in B cells and dendritic cells. To screen a library of 1.4 million compounds for inhibitors of SPPL2a, we developed an imaging assay detecting nuclear translocation of the proteolytically released cytosolic substrate fragment. The state-of-the-art hit calling approach based on nuclear translocation resulted in numerous false-positive hits, mainly interrupting intracellular protein trafficking. To filter the false positives, we extracted 340 image-based readouts and developed a novel multiparametric analysis method that successfully triaged the primary hit list. The identified scaffolds were validated by demonstrating activity on endogenous SPPL2a and substrate CD74/p8 in B cells. The multiparametric analysis discovered diverse cellular phenotypes and provided profiles for the whole library. The principle of the presented imaging assay, the screening strategy, and multiparametric analysis are potentially applicable in future screening campaigns.

7.
Nature ; 546(7658): 376-380, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28562588

ABSTRACT

Diarrhoeal disease is responsible for 8.6% of global child mortality. Recent epidemiological studies found the protozoan parasite Cryptosporidium to be a leading cause of paediatric diarrhoea, with particularly grave impact on infants and immunocompromised individuals. There is neither a vaccine nor an effective treatment. Here we establish a drug discovery process built on scalable phenotypic assays and mouse models that take advantage of transgenic parasites. Screening a library of compounds with anti-parasitic activity, we identify pyrazolopyridines as inhibitors of Cryptosporidium parvum and Cryptosporidium hominis. Oral treatment with the pyrazolopyridine KDU731 results in a potent reduction in intestinal infection of immunocompromised mice. Treatment also leads to rapid resolution of diarrhoea and dehydration in neonatal calves, a clinical model of cryptosporidiosis that closely resembles human infection. Our results suggest that the Cryptosporidium lipid kinase PI(4)K (phosphatidylinositol-4-OH kinase) is a target for pyrazolopyridines and that KDU731 warrants further preclinical evaluation as a drug candidate for the treatment of cryptosporidiosis.


Subject(s)
1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , Cryptosporidiosis/drug therapy , Cryptosporidiosis/parasitology , Cryptosporidium/drug effects , Cryptosporidium/enzymology , Pyrazoles/pharmacology , Pyridines/pharmacology , Animals , Animals, Newborn , Cattle , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immunocompromised Host , Interferon-gamma/deficiency , Interferon-gamma/genetics , Male , Mice , Mice, Knockout , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Pyridines/chemistry , Pyridines/pharmacokinetics , Rats , Rats, Wistar
8.
Cell Rep ; 19(3): 451-460, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28423309

ABSTRACT

Flavivirus infections by Zika and dengue virus impose a significant global healthcare threat with no US Food and Drug Administration (FDA)-approved vaccination or specific antiviral treatment available. Here, we present the discovery of an anti-flaviviral natural product named cavinafungin. Cavinafungin is a potent and selectively active compound against Zika and all four dengue virus serotypes. Unbiased, genome-wide genomic profiling in human cells using a novel CRISPR/Cas9 protocol identified the endoplasmic-reticulum-localized signal peptidase as the efficacy target of cavinafungin. Orthogonal profiling in S. cerevisiae followed by the selection of resistant mutants pinpointed the catalytic subunit of the signal peptidase SEC11 as the evolutionary conserved target. Biochemical analysis confirmed a rapid block of signal sequence cleavage of both host and viral proteins by cavinafungin. This study provides an effective compound against the eukaryotic signal peptidase and independent confirmation of the recently identified critical role of the signal peptidase in the replicative cycle of flaviviruses.


Subject(s)
Biological Products/pharmacology , Dengue Virus/physiology , Lipopeptides/pharmacology , Virus Replication/drug effects , Zika Virus/physiology , Biological Products/chemistry , CRISPR-Cas Systems/genetics , Dengue Virus/drug effects , Gene Knockdown Techniques , Genome, Human , Genomics , HCT116 Cells , Humans , Lipopeptides/chemistry , Membrane Proteins , Protein Subunits/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Serine Endopeptidases , Viral Proteins/metabolism , Zika Virus/drug effects
9.
Bioorg Med Chem Lett ; 27(6): 1385-1389, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28216045

ABSTRACT

A series of 2-oxopiperazine derivatives were designed from the pyrrolopiperazinone cell-based screening hit 4 as a dengue virus inhibitor. Systematic investigation of the structure-activity relationship (SAR) around the piperazinone ring led to the identification of compound (S)-29, which exhibited potent anti-dengue activity in the cell-based assay across all four dengue serotypes with EC50<0.1µM. Cross-resistant analysis confirmed that the virus NS4B protein remained the target of the new oxopiperazine analogs obtained via scaffold morphing from the HTS hit 4.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Piperazines/pharmacology , Cell Line , Drug Discovery , High-Throughput Screening Assays , Humans , Structure-Activity Relationship
10.
Nature ; 541(7638): 541-545, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28068668

ABSTRACT

Cell fate perturbations underlie many human diseases, including breast cancer. Unfortunately, the mechanisms by which breast cell fate are regulated are largely unknown. The mammary gland epithelium consists of differentiated luminal epithelial and basal myoepithelial cells, as well as undifferentiated stem cells and more restricted progenitors. Breast cancer originates from this epithelium, but the molecular mechanisms that underlie breast epithelial hierarchy remain ill-defined. Here, we use a high-content confocal image-based short hairpin RNA screen to identify tumour suppressors that regulate breast cell fate in primary human breast epithelial cells. We show that ablation of the large tumour suppressor kinases (LATS) 1 and 2 (refs 5, 6), which are part of the Hippo pathway, promotes the luminal phenotype and increases the number of bipotent and luminal progenitors, the proposed cells-of-origin of most human breast cancers. Mechanistically, we have identified a direct interaction between Hippo and oestrogen receptor-α (ERα) signalling. In the presence of LATS, ERα was targeted for ubiquitination and Ddb1-cullin4-associated-factor 1 (DCAF1)-dependent proteasomal degradation. Absence of LATS stabilized ERα and the Hippo effectors YAP and TAZ (hereafter YAP/TAZ), which together control breast cell fate through intrinsic and paracrine mechanisms. Our findings reveal a non-canonical (that is, YAP/TAZ-independent) effect of LATS in the regulation of human breast cell fate.


Subject(s)
Breast/cytology , Breast/enzymology , Cell Differentiation , Cell Lineage , Estrogen Receptor alpha/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/agonists , Adaptor Proteins, Signal Transducing/metabolism , Breast/pathology , Carrier Proteins/metabolism , Cells, Cultured , Estrogen Receptor alpha/agonists , Female , Genes, Tumor Suppressor , Humans , Phosphoproteins/agonists , Phosphoproteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/deficiency , Proteolysis , Signal Transduction , Transcription Factors , Tumor Suppressor Proteins/deficiency , Ubiquitin/metabolism , Ubiquitin-Protein Ligases , YAP-Signaling Proteins
11.
Nat Microbiol ; 1: 16166, 2016 Sep 19.
Article in English | MEDLINE | ID: mdl-27642791

ABSTRACT

A molecular understanding of drug resistance mechanisms enables surveillance of the effectiveness of new antimicrobial therapies during development and deployment in the field. We used conventional drug resistance selection as well as a regime of limiting dilution at early stages of drug treatment to probe two antimalarial imidazolopiperazines, KAF156 and GNF179. The latter approach permits the isolation of low-fitness mutants that might otherwise be out-competed during selection. Whole-genome sequencing of 24 independently derived resistant Plasmodium falciparum clones revealed four parasites with mutations in the known cyclic amine resistance locus (pfcarl) and a further 20 with mutations in two previously unreported P. falciparum drug resistance genes, an acetyl-CoA transporter (pfact) and a UDP-galactose transporter (pfugt). Mutations were validated both in vitro by CRISPR editing in P. falciparum and in vivo by evolution of resistant Plasmodium berghei mutants. Both PfACT and PfUGT were localized to the endoplasmic reticulum by fluorescence microscopy. As mutations in pfact and pfugt conveyed resistance against additional unrelated chemical scaffolds, these genes are probably involved in broad mechanisms of antimalarial drug resistance.

12.
PLoS One ; 8(1): e51671, 2013.
Article in English | MEDLINE | ID: mdl-23300950

ABSTRACT

Emerging evidence suggests that some cancers contain a population of stem-like TICs (tumor-initiating cells) and eliminating TICs may offer a new strategy to develop successful anti-cancer therapies. As molecular mechanisms underlying the maintenance of the TIC pool are poorly understood, the development of TIC-specific therapeutics remains a major challenge. We first identified and characterized TICs and non-TICs isolated from a mouse breast cancer model. TICs displayed increased tumorigenic potential, self-renewal, heterogeneous differentiation, and bipotency. Gene expression analysis and immunostaining of TICs and non-TICs revealed that FGFR2 was preferentially expressed in TICs. Loss of FGFR2 impaired self-renewal of TICs, thus resulting in marked decreases in the TIC population and tumorigenic potential. Restoration of FGFR2 rescued the defects in TIC pool maintenance, bipotency, and breast tumor growth driven by FGFR2 knockdown. In addition, pharmacological inhibition of FGFR2 kinase activity led to a decrease in the TIC population which resulted in suppression of breast tumor growth. Moreover, human breast TICs isolated from patient tumor samples were found enriched in a FGFR2+ population that was sufficient to initiate tumor growth. Our data suggest that FGFR2 is essential in sustaining the breast TIC pool through promotion of self-renewal and maintenance of bipotent TICs, and raise the possibility of FGFR2 inhibition as a strategy for anti-cancer therapy by eradicating breast TICs.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/metabolism , Neoplastic Stem Cells/immunology , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Animals , CD24 Antigen/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Female , Flow Cytometry , Gene Expression Profiling , Humans , Integrin beta1/metabolism , Mice , Mice, Inbred NOD , Microscopy, Fluorescence , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Signal Transduction
13.
Chem Biol ; 19(8): 955-62, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22884261

ABSTRACT

Hippo signaling represents a tumor suppressor pathway that regulates organ size and tumorigenesis through phosphorylation and inhibition of the transcription coactivator YAP. Here, we show that serum deprivation dramatically induces YAP Ser127 phosphorylation and cytoplasmic retention, independent of cell-cell contact. Through chemical isolation and activity profiling, we identified serum-derived sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA) as small molecule activators of YAP. S1P induces YAP nuclear localization through S1P(2) receptor, Rho GTPase activation, and F-actin polymerization, independent of the core Hippo pathway kinases. Bioinformatics studies also showed that S1P stimulation induces YAP target gene expression in mouse liver and human embryonic stem cells. These results revealed potent small molecule regulators of YAP and suggest that S1P and LPA might modulate cell proliferation and tumorigenesis through YAP activation.


Subject(s)
Lysophospholipids/pharmacology , Nuclear Proteins/metabolism , Sphingosine/analogs & derivatives , Transcription Factors/metabolism , Actins/metabolism , Animals , Cell Cycle Proteins , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Embryonic Stem Cells/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Lysophospholipids/blood , Lysophospholipids/chemistry , Lysophospholipids/isolation & purification , Mice , Nuclear Proteins/chemistry , Phosphorylation/drug effects , Receptors, Lysosphingolipid/metabolism , Signal Transduction/drug effects , Sphingosine/blood , Sphingosine/isolation & purification , Sphingosine/pharmacology , Transcription Factors/chemistry , rho GTP-Binding Proteins/metabolism
15.
Cell Host Microbe ; 11(3): 306-18, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22423970

ABSTRACT

Pathogens commonly utilize endocytic pathways to gain cellular access. The endosomal pattern recognition receptors TLR7 and TLR9 detect pathogen-encoded nucleic acids to initiate MyD88-dependent proinflammatory responses to microbial infection. Using genome-wide RNAi screening and integrative systems-based analysis, we identify 190 cofactors required for TLR7- and TLR9-directed signaling responses. A set of cofactors were crossprofiled for their activities downstream of several immunoreceptors and then functionally mapped based on the known architecture of NF-κB signaling pathways. Protein complexes and pathways involved in ubiquitin-protein ligase activities, sphingolipid metabolism, chromatin modifications, and ancient stress responses were found to modulate innate recognition of endosomal nucleic acids. Additionally, hepatocyte growth factor-regulated tyrosine kinase substrate (HRS) was characterized as necessary for ubiquitin-dependent TLR9 targeting to the endolysosome. Proteins and pathways identified here should prove useful in delineating strategies to manipulate innate responses for treatment of autoimmune disorders and microbial infection.


Subject(s)
Immunity, Innate/genetics , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/metabolism , Animals , Chick Embryo , Computer Simulation , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomal Sorting Complexes Required for Transport/physiology , Endosomes/metabolism , Gene Expression Profiling , Gene Knockdown Techniques , Gene Regulatory Networks , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Models, Biological , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Phosphoproteins/metabolism , Phosphoproteins/physiology , Protein Transport , RNA Interference , Signal Transduction , Support Vector Machine
16.
Science ; 334(6061): 1372-7, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22096101

ABSTRACT

Most malaria drug development focuses on parasite stages detected in red blood cells, even though, to achieve eradication, next-generation drugs active against both erythrocytic and exo-erythrocytic forms would be preferable. We applied a multifactorial approach to a set of >4000 commercially available compounds with previously demonstrated blood-stage activity (median inhibitory concentration < 1 micromolar) and identified chemical scaffolds with potent activity against both forms. From this screen, we identified an imidazolopiperazine scaffold series that was highly enriched among compounds active against Plasmodium liver stages. The orally bioavailable lead imidazolopiperazine confers complete causal prophylactic protection (15 milligrams/kilogram) in rodent models of malaria and shows potent in vivo blood-stage therapeutic activity. The open-source chemical tools resulting from our effort provide starting points for future drug discovery programs, as well as opportunities for researchers to investigate the biology of exo-erythrocytic forms.


Subject(s)
Antimalarials/pharmacology , Drug Discovery , Imidazoles/pharmacology , Liver/parasitology , Malaria/drug therapy , Piperazines/pharmacology , Plasmodium/drug effects , Animals , Antimalarials/chemistry , Antimalarials/pharmacokinetics , Antimalarials/therapeutic use , Cell Line, Tumor , Drug Evaluation, Preclinical , Drug Resistance , Erythrocytes/parasitology , Humans , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Imidazoles/therapeutic use , Malaria/parasitology , Malaria/prevention & control , Mice , Mice, Inbred BALB C , Molecular Structure , Piperazines/chemistry , Piperazines/pharmacokinetics , Piperazines/therapeutic use , Plasmodium/cytology , Plasmodium/growth & development , Plasmodium/physiology , Plasmodium berghei/cytology , Plasmodium berghei/drug effects , Plasmodium berghei/growth & development , Plasmodium berghei/physiology , Plasmodium falciparum/cytology , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development , Plasmodium falciparum/physiology , Plasmodium yoelii/cytology , Plasmodium yoelii/drug effects , Plasmodium yoelii/growth & development , Plasmodium yoelii/physiology , Polymorphism, Single Nucleotide , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Random Allocation , Small Molecule Libraries , Sporozoites/drug effects , Sporozoites/growth & development
17.
ACS Chem Biol ; 6(11): 1214-22, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-21866942

ABSTRACT

Decoquinate has single-digit nanomolar activity against in vitro blood stage Plasmodium falciparum parasites, the causative agent of human malaria. In vitro evolution of decoquinate-resistant parasites and subsequent comparative genomic analysis to the drug-sensitive parental strain revealed resistance was conferred by two nonsynonymous single nucleotide polymorphisms in the gene encoding cytochrome b. The resultant amino acid mutations, A122T and Y126C, reside within helix C in the ubiquinol-binding pocket of cytochrome b, an essential subunit of the cytochrome bc(1) complex. As with other cytochrome bc(1) inhibitors, such as atovaquone, decoquinate has low nanomolar activity against in vitro liver stage P. yoelii and provides partial prophylaxis protection when administered to infected mice at 50 mg kg(-1). In addition, transgenic parasites expressing yeast dihydroorotate dehydrogenase are >200-fold less sensitive to decoquinate, which provides additional evidence that this drug inhibits the parasite's mitochondrial electron transport chain. Importantly, decoquinate exhibits limited cross-resistance to a panel of atovaquone-resistant parasites evolved to harbor various mutations in cytochrome b. The basis for this difference was revealed by molecular docking studies, in which both of these inhibitors were shown to have distinctly different modes of binding within the ubiquinol-binding site of cytochrome b.


Subject(s)
Antimalarials/pharmacology , Cytochromes b/antagonists & inhibitors , Decoquinate/pharmacology , Plasmodium falciparum/drug effects , Animals , Antimalarials/administration & dosage , Antimalarials/chemistry , Crystallography, X-Ray , Cytochromes b/genetics , Cytochromes b/metabolism , Decoquinate/administration & dosage , Decoquinate/chemistry , Drug Discovery , Drug Resistance, Fungal/drug effects , Drug Resistance, Fungal/genetics , Female , Humans , Mice , Mice, Inbred ICR , Models, Molecular , Molecular Structure , Parasitic Sensitivity Tests , Plasmodium falciparum/genetics , Structure-Activity Relationship
18.
Proc Natl Acad Sci U S A ; 107(7): 2944-9, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133616

ABSTRACT

Many proteins or other biological macromolecules are localized to more than one subcellular structure. The fraction of a protein in different cellular compartments is often measured by colocalization with organelle-specific fluorescent markers, requiring availability of fluorescent probes for each compartment and acquisition of images for each in conjunction with the macromolecule of interest. Alternatively, tailored algorithms allow finding particular regions in images and quantifying the amount of fluorescence they contain. Unfortunately, this approach requires extensive hand-tuning of algorithms and is often cell type-dependent. Here we describe a machine-learning approach for estimating the amount of fluorescent signal in different subcellular compartments without hand tuning, requiring only the acquisition of separate training images of markers for each compartment. In testing on images of cells stained with mixtures of probes for different organelles, we achieved a 93% correlation between estimated and expected amounts of probes in each compartment. We also demonstrated that the method can be used to quantify drug-dependent protein translocations. The method enables automated and unbiased determination of the distributions of protein across cellular compartments, and will significantly improve imaging-based high-throughput assays and facilitate proteome-scale localization efforts.


Subject(s)
Algorithms , Artificial Intelligence , Fluorescent Dyes/metabolism , Image Processing, Computer-Assisted/methods , Intracellular Space/metabolism , Protein Transport/physiology , Proteomics/methods
19.
Cell ; 135(1): 49-60, 2008 Oct 03.
Article in English | MEDLINE | ID: mdl-18854154

ABSTRACT

Human Immunodeficiency Viruses (HIV-1 and HIV-2) rely upon host-encoded proteins to facilitate their replication. Here, we combined genome-wide siRNA analyses with interrogation of human interactome databases to assemble a host-pathogen biochemical network containing 213 confirmed host cellular factors and 11 HIV-1-encoded proteins. Protein complexes that regulate ubiquitin conjugation, proteolysis, DNA-damage response, and RNA splicing were identified as important modulators of early-stage HIV-1 infection. Additionally, over 40 new factors were shown to specifically influence the initiation and/or kinetics of HIV-1 DNA synthesis, including cytoskeletal regulatory proteins, modulators of posttranslational modification, and nucleic acid-binding proteins. Finally, 15 proteins with diverse functional roles, including nuclear transport, prostaglandin synthesis, ubiquitination, and transcription, were found to influence nuclear import or viral DNA integration. Taken together, the multiscale approach described here has uncovered multiprotein virus-host interactions that likely act in concert to facilitate the early steps of HIV-1 infection.


Subject(s)
HIV Infections/virology , HIV-1/physiology , Host-Pathogen Interactions , Proteins/metabolism , Virus Replication , Cell Line , Humans , RNA Interference , Two-Hybrid System Techniques
20.
J Biol Chem ; 283(37): 25576-25588, 2008 Sep 12.
Article in English | MEDLINE | ID: mdl-18641393

ABSTRACT

The thyroid hormone receptor alpha1 (TRalpha) exhibits a dual role as an activator or repressor of its target genes in response to thyroid hormone (T(3)). Previously, we have shown that TRalpha, formerly thought to reside solely in the nucleus bound to DNA, actually shuttles rapidly between the nucleus and cytoplasm. An important aspect of the shuttling activity of TRalpha is its ability to exit the nucleus through the nuclear pore complex. TRalpha export is not sensitive to treatment with the CRM1-specific inhibitor leptomycin B (LMB) in heterokaryon assays, suggesting a role for an export receptor other than CRM1. Here, we have used a combined approach of in vivo fluorescence recovery after photobleaching experiments, in vitro permeabilized cell nuclear export assays, and glutathione S-transferase pull-down assays to investigate the export pathway used by TRalpha. We show that, in addition to shuttling in heterokaryons, TRalpha shuttles rapidly in an unfused monokaryon system as well. Furthermore, our data show that TRalpha directly interacts with calreticulin, and point to the intriguing possibility that TRalpha follows a cooperative export pathway in which both calreticulin and CRM1 play a role in facilitating efficient translocation of TRalpha from the nucleus to cytoplasm.


Subject(s)
Calreticulin/metabolism , Karyopherins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Thyroid Hormone Receptors alpha/metabolism , Active Transport, Cell Nucleus , Animals , Blotting, Western , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , Fatty Acids, Unsaturated/pharmacology , Glutathione Transferase/metabolism , HeLa Cells , Humans , Mice , Models, Biological , Exportin 1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...