Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
STAR Protoc ; 5(1): 102896, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38363687

ABSTRACT

Artezomibs (ATZs), dual-pharmacophore molecules comprising of artemisinin and a parasite proteasome inhibitor, hijack parasite ubiquitin proteasome system to transform into new proteasome inhibitors following the activation of artemisinin by heme.1 Here, we present a protocol for using a fluorescent activity-based broad-spectrum proteasome inhibitor probe to study intracellular conversion of ATZ molecules into new proteasome inhibitors in malaria parasites. We describe steps for drug treatment and washout, parasite lysis, proteasome labeling, and visualization. For complete details on the use and execution of this protocol, please refer to Zhan et al.1.


Subject(s)
Antimalarials , Artemisinins , Parasites , Animals , Plasmodium falciparum , Proteasome Inhibitors/pharmacology , Proteasome Endopeptidase Complex , Antimalarials/pharmacology , Antimalarials/therapeutic use , Artemisinins/pharmacology , Artemisinins/therapeutic use
2.
Nat Commun ; 14(1): 8302, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38097652

ABSTRACT

The proteasome of the malaria parasite Plasmodium falciparum (Pf20S) is an advantageous drug target because its inhibition kills P. falciparum in multiple stages of its life cycle and synergizes with artemisinins. We recently developed a macrocyclic peptide, TDI-8304, that is highly selective for Pf20S over human proteasomes and is potent in vitro and in vivo against P. falciparum. A mutation in the Pf20S ß6 subunit, A117D, confers resistance to TDI-8304, yet enhances both enzyme inhibition and anti-parasite activity of a tripeptide vinyl sulfone ß2 inhibitor, WLW-vs. Here we present the high-resolution cryo-EM structures of Pf20S with TDI-8304, of human constitutive proteasome with TDI-8304, and of Pf20Sß6A117D with WLW-vs that give insights into the species selectivity of TDI-8304, resistance to it, and the collateral sensitivity associated with resistance, including that TDI-8304 binds ß2 and ß5 in wild type Pf20S as well as WLW-vs binds ß2 and ß5 in Pf20Sß6A117D. We further show that TDI-8304 kills P. falciparum as quickly as chloroquine and artemisinin and is active against P. cynomolgi at the liver stage. This increases interest in using these structures to facilitate the development of Pf20S inhibitors that target multiple proteasome subunits and limit the emergence of resistance.


Subject(s)
Antimalarials , Malaria, Falciparum , Humans , Plasmodium falciparum/genetics , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/chemistry , Proteasome Endopeptidase Complex/metabolism , Drug Collateral Sensitivity , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Antimalarials/pharmacology , Antimalarials/chemistry , Drug Resistance/genetics , Protozoan Proteins/genetics
3.
Cell Chem Biol ; 30(5): 457-469.e11, 2023 05 18.
Article in English | MEDLINE | ID: mdl-37148884

ABSTRACT

Artemisinins (ART) are critical anti-malarials and despite their use in combination therapy, ART-resistant Plasmodium falciparum is spreading globally. To counter ART resistance, we designed artezomibs (ATZs), molecules that link an ART with a proteasome inhibitor (PI) via a non-labile amide bond and hijack parasite's own ubiquitin-proteasome system to create novel anti-malarials in situ. Upon activation of the ART moiety, ATZs covalently attach to and damage multiple parasite proteins, marking them for proteasomal degradation. When damaged proteins enter the proteasome, their attached PIs inhibit protease function, potentiating the parasiticidal action of ART and overcoming ART resistance. Binding of the PI moiety to the proteasome active site is enhanced by distal interactions of the extended attached peptides, providing a mechanism to overcome PI resistance. ATZs have an extra mode of action beyond that of each component, thereby overcoming resistance to both components, while avoiding transient monotherapy seen when individual agents have disparate pharmacokinetic profiles.


Subject(s)
Antimalarials , Artemisinins , Parasites , Plasmodium , Animals , Antimalarials/chemistry , Proteasome Endopeptidase Complex/metabolism , Parasites/metabolism , Pharmacophore , Ubiquitin , Plasmodium/metabolism , Artemisinins/pharmacology , Drug Resistance
4.
Cell Chem Biol ; 30(5): 470-485.e6, 2023 05 18.
Article in English | MEDLINE | ID: mdl-36963402

ABSTRACT

The Plasmodium falciparum proteasome constitutes a promising antimalarial target, with multiple chemotypes potently and selectively inhibiting parasite proliferation and synergizing with the first-line artemisinin drugs, including against artemisinin-resistant parasites. We compared resistance profiles of vinyl sulfone, epoxyketone, macrocyclic peptide, and asparagine ethylenediamine inhibitors and report that the vinyl sulfones were potent even against mutant parasites resistant to other proteasome inhibitors and did not readily select for resistance, particularly WLL that displays covalent and irreversible binding to the catalytic ß2 and ß5 proteasome subunits. We also observed instances of collateral hypersensitivity, whereby resistance to one inhibitor could sensitize parasites to distinct chemotypes. Proteasome selectivity was confirmed using CRISPR/Cas9-edited mutant and conditional knockdown parasites. Molecular modeling of proteasome mutations suggested spatial contraction of the ß5 P1 binding pocket, compromising compound binding. Dual targeting of P. falciparum proteasome subunits using covalent inhibitors provides a potential strategy for restoring artemisinin activity and combating the spread of drug-resistant malaria.


Subject(s)
Antimalarials , Artemisinins , Malaria, Falciparum , Plasmodium , Humans , Antimalarials/pharmacology , Antimalarials/chemistry , Proteasome Endopeptidase Complex/metabolism , Plasmodium/metabolism , Artemisinins/chemistry , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/chemistry
5.
J Med Chem ; 66(2): 1484-1508, 2023 01 26.
Article in English | MEDLINE | ID: mdl-36630286

ABSTRACT

With increasing reports of resistance to artemisinins and artemisinin-combination therapies, targeting the Plasmodium proteasome is a promising strategy for antimalarial development. We recently reported a highly selective Plasmodium falciparum proteasome inhibitor with anti-malarial activity in the humanized mouse model. To balance the permeability of the series of macrocycles with other drug-like properties, we conducted further structure-activity relationship studies on a biphenyl ether-tethered macrocyclic scaffold. Extensive SAR studies around the P1, P3, and P5 groups and peptide backbone identified compound TDI-8414. TDI-8414 showed nanomolar antiparasitic activity, no toxicity to HepG2 cells, high selectivity against the Plasmodium proteasome over the human constitutive proteasome and immunoproteasome, improved solubility and PAMPA permeability, and enhanced metabolic stability in microsomes and plasma of both humans and mice.


Subject(s)
Antimalarials , Plasmodium , Humans , Animals , Mice , Antimalarials/pharmacology , Antimalarials/chemistry , Proteasome Endopeptidase Complex/metabolism , Structure-Activity Relationship , Plasmodium falciparum/metabolism , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/chemistry
6.
Antimicrob Agents Chemother ; 66(10): e0081722, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36094216

ABSTRACT

The proteasome is a promising target for antimalarial chemotherapy. We assessed ex vivo susceptibilities of fresh Plasmodium falciparum isolates from eastern Uganda to seven proteasome inhibitors: two asparagine ethylenediamines, two macrocyclic peptides, and three peptide boronates; five had median IC50 values <100 nM. TDI8304, a macrocylic peptide lead compound with drug-like properties, had a median IC50 of 16 nM. Sequencing genes encoding the ß2 and ß5 catalytic proteasome subunits, the predicted targets of the inhibitors, and five additional proteasome subunits, identified two mutations in ß2 (I204T, S214F), three mutations in ß5 (V2I, A142S, D150E), and three mutations in other subunits. The ß2 S214F mutation was associated with decreased susceptibility to two peptide boronates, with IC50s of 181 nM and 2635 nM against mutant versus 62 nM and 477 nM against wild type parasites for MMV1579506 and MMV1794229, respectively, although significance could not be formally assessed due to the small number of mutant parasites with available data. The other ß2 and ß5 mutations and mutations in other subunits were not associated with susceptibility to tested compounds. Against culture-adapted Ugandan isolates, two asparagine ethylenediamines and the peptide proteasome inhibitors WLW-vinyl sulfone and WLL-vinyl sulfone (which were not studied ex vivo) demonstrated low nM activity, without decreased activity against ß2 S214F mutant parasites. Overall, proteasome inhibitors had potent activity against P. falciparum isolates circulating in Uganda, and genetic variation in proteasome targets was uncommon.


Subject(s)
Antimalarials , Plasmodium falciparum , Proteasome Inhibitors , Humans , Antimalarials/pharmacology , Antimalarials/chemistry , Asparagine , Drug Resistance/genetics , Ethylenediamines/pharmacology , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Peptides/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Inhibitors/chemistry , Proteasome Inhibitors/pharmacology , Uganda
7.
J Med Chem ; 65(13): 9350-9375, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35727231

ABSTRACT

With over 200 million cases and close to half a million deaths each year, malaria is a threat to global health, particularly in developing countries. Plasmodium falciparum, the parasite that causes the most severe form of the disease, has developed resistance to all antimalarial drugs. Resistance to the first-line antimalarial artemisinin and to artemisinin combination therapies is widespread in Southeast Asia and is emerging in sub-Saharan Africa. The P. falciparum proteasome is an attractive antimalarial target because its inhibition kills the parasite at multiple stages of its life cycle and restores artemisinin sensitivity in parasites that have become resistant through mutation in Kelch K13. Here, we detail our efforts to develop noncovalent, macrocyclic peptide malaria proteasome inhibitors, guided by structural analysis and pharmacokinetic properties, leading to a potent, species-selective, metabolically stable inhibitor.


Subject(s)
Antimalarials , Artemisinins , Malaria, Falciparum , Antimalarials/pharmacology , Antimalarials/therapeutic use , Artemisinins/pharmacology , Drug Resistance , Humans , Malaria, Falciparum/drug therapy , Peptides/therapeutic use , Plasmodium falciparum , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Protozoan Proteins/genetics
8.
iScience ; 24(2): 102082, 2021 Feb 19.
Article in English | MEDLINE | ID: mdl-33644714

ABSTRACT

Malaria remains a major cause of morbidity and mortality in the developing world. Recent work has implicated chromosome end stability and the repair of DNA breaks through telomere healing as potent drivers of variant antigen diversification, thus associating basic mechanisms for maintaining genome integrity with aspects of host-parasite interactions. Here we applied long-read sequencing technology to precisely examine the dynamics of telomere addition and chromosome end stabilization in response to double-strand breaks within subtelomeric regions. We observed that the process of telomere healing induces the initial synthesis of telomere repeats well in excess of the minimal number required for end stability. However, once stabilized, these newly created telomeres appear to function normally, eventually returning to a length nearing that of intact chromosome ends. These results parallel recent observations in humans, suggesting an evolutionarily conserved mechanism for chromosome end repair.

9.
Angew Chem Int Ed Engl ; 60(17): 9279-9283, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33433953

ABSTRACT

Plasmodium falciparum proteasome (Pf20S) inhibitors are active against Plasmodium at multiple stages-erythrocytic, gametocyte, liver, and gamete activation stages-indicating that selective Pf20S inhibitors possess the potential to be therapeutic, prophylactic, and transmission-blocking antimalarials. Starting from a reported compound, we developed a noncovalent, macrocyclic peptide inhibitor of the malarial proteasome with high species selectivity and improved pharmacokinetic properties. The compound demonstrates specific, time-dependent inhibition of the ß5 subunit of the Pf20S, kills artemisinin-sensitive and artemisinin-resistant P. falciparum isolates in vitro and reduces parasitemia in humanized, P. falciparum-infected mice.


Subject(s)
Antimalarials/pharmacology , Drug Development , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Animals , Antimalarials/chemical synthesis , Antimalarials/chemistry , Malaria, Falciparum/metabolism , Mice , Models, Molecular , Molecular Conformation , Parasitic Sensitivity Tests , Plasmodium falciparum/enzymology , Proteasome Inhibitors/chemical synthesis , Proteasome Inhibitors/chemistry
10.
Mol Microbiol ; 115(4): 503-507, 2021 04.
Article in English | MEDLINE | ID: mdl-33103309

ABSTRACT

Malaria caused by the protozoan parasite Plasmodium falciparum continues to impose significant morbidity and mortality, despite substantial investment into drug and vaccine development and deployment. Underlying the resilience of this parasite is its remarkable ability to undergo genome modifications, thus, providing parasite populations with extensive genetic variability that accelerates selection of drug resistance and limits the efficacy of most vaccines. This genome plasticity is rooted in the mechanisms of DNA repair that parasites employ to maintain genome integrity, a process skewed toward homologous recombination through the evolutionary loss of classical nonhomologous end joining. Repair of DNA double-strand breaks have been shown to enable "shuffling" of antigen-encoding gene sequences to vastly increase antigen diversity and to enable copy number expansion of genes that contribute to drug resistance. The latter phenomenon has been proposed to be a major contributor to the rise of resistance to several classes of antimalarial drugs. In this issue of Molecular Microbiology, McDaniels and colleagues add yet another mechanism that malaria parasites use to reduce drug susceptibility by demonstrating that P. falciparum can maintain expanded arrays of drug resistance cassettes as stably replicating, circular, extrachromosomal DNAs, thus, expanding genome plasticity beyond the parasite's 14 nuclear chromosomes.


Subject(s)
Drug Resistance/genetics , Genome, Protozoan , Malaria, Falciparum/prevention & control , Malaria, Falciparum/parasitology , Plasmodium falciparum/drug effects , Plasmodium falciparum/physiology , Animals , Antigenic Variation , Antimalarials/pharmacology , DNA Repair , Humans , Vaccines
11.
Trends Parasitol ; 36(6): 504-511, 2020 06.
Article in English | MEDLINE | ID: mdl-32407681

ABSTRACT

Experimental research into malaria biology and pathogenesis has historically focused on two model systems, in vitro culture of the human parasite Plasmodium falciparum and in vivo infections of laboratory animals using rodent parasites. While there is clear value in having a manipulatable animal model for studying malaria, there have occasionally been controversies around how representative the rodent model is of the human disease, and therefore significant emphasis has been placed on the similarities between the two biological systems. By focusing on basic nuclear functions, we wish to highlight that identifying key differences in the parasites and their interactions with their mammalian hosts can be equally informative and provide remarkable insights into the biology and evolution of these important infectious organisms.


Subject(s)
Host-Parasite Interactions/physiology , Malaria/parasitology , Plasmodium/physiology , Rodentia/parasitology , Animals , Disease Models, Animal , Humans
12.
Am J Trop Med Hyg ; 102(6): 1319-1322, 2020 06.
Article in English | MEDLINE | ID: mdl-32228792

ABSTRACT

The six previously reported civilian cases of mucosal leishmaniasis (ML) diagnosed in the United States have all represented imported New World ML. We describe two new patients with ML diagnosed in New York City-a Syrian immigrant with a nasal mass (Leishmania tropica), the first report of Old World ML in the United States, and an American ecologist who worked in Bolivia and had been treated for cutaneous infection 23 years before developing lesions (L. (Viannia) braziliensis) initially of the uvula, soft palate, and posterior pharynx and subsequently the larynx.


Subject(s)
Leishmaniasis, Mucocutaneous/epidemiology , Leishmaniasis, Mucocutaneous/pathology , Aged , Amphotericin B/therapeutic use , Antiprotozoal Agents/therapeutic use , Humans , Leishmania braziliensis/isolation & purification , Leishmaniasis, Mucocutaneous/drug therapy , Male , Middle Aged , New York City/epidemiology , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/therapeutic use
13.
Article in English | MEDLINE | ID: mdl-32179524

ABSTRACT

Malaria parasites invade and replicate within red blood cells (RBCs), extensively modifying their structure and gaining access to the extracellular environment by placing the plasmodial surface anion channel (PSAC) into the RBC membrane. Expression of members of the cytoadherence linked antigen gene 3 (clag3) family is required for PSAC activity, a process that is regulated epigenetically. PSAC is a well-established route of uptake for large, hydrophilic antimalarial compounds, and parasites can acquire resistance by silencing clag3 gene expression, thereby reducing drug uptake. We found that exposure to sub-IC50 concentrations of the histone methyltransferase inhibitor chaetocin caused substantial changes in both clag3 gene expression and RBC permeability, and reversed acquired resistance to the antimalarial compound blasticidin S that is transported through PSACs. Chaetocin treatment also altered progression of parasites through their replicative cycle, presumably by changing their ability to modify chromatin appropriately to enable DNA replication. These results indicate that targeting histone modifiers could represent a novel tool for reversing epigenetically acquired drug resistance in P. falciparum.


Subject(s)
Malaria, Falciparum , Parasites , Pharmaceutical Preparations , Animals , Drug Resistance/genetics , Erythrocytes/metabolism , Histone Methyltransferases , Histones/genetics , Parasites/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
14.
mBio ; 11(2)2020 03 17.
Article in English | MEDLINE | ID: mdl-32184256

ABSTRACT

The protozoan parasites that cause malaria infect a wide variety of vertebrate hosts, including birds, reptiles, and mammals, and the evolutionary pressures inherent to the host-parasite relationship have profoundly shaped the genomes of both host and parasite. Here, we report that these selective pressures have resulted in unexpected alterations to one of the most basic aspects of eukaryotic biology, the maintenance of genome integrity through DNA repair. Malaria parasites that infect humans continuously generate genetic diversity within their antigen-encoding gene families through frequent ectopic recombination between gene family members, a process that is a crucial feature of the persistence of malaria globally. The continuous generation of antigen diversity ensures that different parasite isolates are antigenically distinct, thus preventing extensive cross-reactive immunity and enabling parasites to maintain stable transmission within human populations. However, the molecular basis of the recombination between gene family members is not well understood. Through computational analyses of the antigen-encoding, multicopy gene families of different Plasmodium species, we report the unexpected observation that malaria parasites that infect rodents do not display the same degree of antigen diversity as observed in Plasmodium falciparum and appear to undergo significantly less ectopic recombination. Using comparative genomics, we also identify key molecular components of the diversification process, thus shedding new light on how malaria parasites balance the maintenance of genome integrity with the requirement for continuous genetic diversification.IMPORTANCE Malaria remains one of the most prevalent and deadly infectious diseases of the developing world, causing approximately 228 million clinical cases and nearly half a million deaths annually. The disease is caused by protozoan parasites of the genus Plasmodium, and of the five species capable of infecting humans, infections with P. falciparum are the most severe. In addition to the parasites that infect people, there are hundreds of additional species that infect birds, reptiles, and other mammals, each exquisitely evolved to meet the specific challenges inherent to survival within their respective hosts. By comparing the unique strategies that each species has evolved, key insights into host-parasite interactions can be gained, including discoveries regarding the pathogenesis of human disease. Here, we describe the surprising observation that closely related parasites with different hosts have evolved remarkably different methods for repairing their genomes. This observation has important implications for the ability of parasites to maintain chronic infections and for the development of host immunity.


Subject(s)
Antigenic Variation/genetics , Evolution, Molecular , Genome, Protozoan , Host Specificity/genetics , Host-Parasite Interactions/genetics , Plasmodium/genetics , Animals , DNA Repair , Genomics , Humans , Malaria/parasitology , Phylogeny , Plasmodium/classification , Rodentia/parasitology
15.
J Med Chem ; 62(13): 6137-6145, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31177777

ABSTRACT

The Plasmodium proteasome (Pf20S) emerged as a target for antimalarials. Pf20S inhibitors are active at multiple stages of the parasite life cycle and synergize with artemisinins, suggesting that Pf20S inhibitors have potential to be prophylactic, therapeutic, and transmission blocking as well as are useful for combination therapy. We recently reported asparagine ethylenediamines (AsnEDAs) as immunoproteasome inhibitors and modified AsnEDAs as selective Pf20S inhibitors. Here, we report further a structure-activity relationship study of AsnEDAs for selective inhibition of Pf20S over human proteasomes. Additionally, we show new mutation that conferred resistance to AsnEDAs and collateral sensitivity to an inhibitor of the Pf20S ß2 subunit, the same as previously identified resistant mutation. This resistance could be overcome through the use of the structure-guided inhibitor design. Collateral sensitivity to inhibitors among respective proteasome subunits underscores the potential value of treating malaria with combinations of inhibitors of different proteasome subunits to minimize the emergence of drug resistance.


Subject(s)
Antimalarials/pharmacology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Antimalarials/chemistry , Antimalarials/metabolism , Asparagine/chemistry , Asparagine/metabolism , Drug Resistance/drug effects , Drug Resistance/genetics , Ethylenediamines/chemistry , Ethylenediamines/metabolism , Humans , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Mutation , Plasmodium falciparum/genetics , Plasmodium falciparum/physiology , Proteasome Endopeptidase Complex/genetics
16.
PLoS Biol ; 17(5): e3000271, 2019 05.
Article in English | MEDLINE | ID: mdl-31083650

ABSTRACT

Malaria parasites possess the remarkable ability to maintain chronic infections that fail to elicit a protective immune response, characteristics that have stymied vaccine development and cause people living in endemic regions to remain at risk of malaria despite previous exposure to the disease. These traits stem from the tremendous antigenic diversity displayed by parasites circulating in the field. For Plasmodium falciparum, the most virulent of the human malaria parasites, this diversity is exemplified by the variant gene family called var, which encodes the major surface antigen displayed on infected red blood cells (RBCs). This gene family exhibits virtually limitless diversity when var gene repertoires from different parasite isolates are compared. Previous studies indicated that this remarkable genome plasticity results from extensive ectopic recombination between var genes during mitotic replication; however, the molecular mechanisms that direct this process to antigen-encoding loci while the rest of the genome remains relatively stable were not determined. Using targeted DNA double-strand breaks (DSBs) and long-read whole-genome sequencing, we show that a single break within an antigen-encoding region of the genome can result in a cascade of recombination events leading to the generation of multiple chimeric var genes, a process that can greatly accelerate the generation of diversity within this family. We also found that recombinations did not occur randomly, but rather high-probability, specific recombination products were observed repeatedly. These results provide a molecular basis for previously described structured rearrangements that drive diversification of this highly polymorphic gene family.


Subject(s)
Antigenic Variation/genetics , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Mitosis/genetics , Parasites/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Recombination, Genetic , Animals , Base Sequence , Chromosomes/genetics , DNA Breaks, Double-Stranded , Humans , Telomere/genetics
17.
Proc Natl Acad Sci U S A ; 115(29): E6863-E6870, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29967165

ABSTRACT

We describe noncovalent, reversible asparagine ethylenediamine (AsnEDA) inhibitors of the Plasmodium falciparum proteasome (Pf20S) ß5 subunit that spare all active subunits of human constitutive and immuno-proteasomes. The compounds are active against erythrocytic, sexual, and liver-stage parasites, against parasites resistant to current antimalarials, and against P. falciparum strains from patients in Africa. The ß5 inhibitors synergize with a ß2 inhibitor in vitro and in mice and with artemisinin. P. falciparum selected for resistance to an AsnEDA ß5 inhibitor surprisingly harbored a point mutation in the noncatalytic ß6 subunit. The ß6 mutant was resistant to the species-selective Pf20S ß5 inhibitor but remained sensitive to the species-nonselective ß5 inhibitors bortezomib and carfilzomib. Moreover, resistance to the Pf20S ß5 inhibitor was accompanied by increased sensitivity to a Pf20S ß2 inhibitor. Finally, the ß5 inhibitor-resistant mutant had a fitness cost that was exacerbated by irradiation. Thus, used in combination, multistage-active inhibitors of the Pf20S ß5 and ß2 subunits afford synergistic antimalarial activity with a potential to delay the emergence of resistance to artemisinins and each other.


Subject(s)
Antimalarials/chemistry , Plasmodium falciparum/enzymology , Proteasome Endopeptidase Complex/chemistry , Proteasome Inhibitors/chemistry , Protozoan Proteins/antagonists & inhibitors , Artemisinins/chemistry , Bortezomib/chemistry , Drug Resistance, Microbial , Humans , Lactones/chemistry , Oligopeptides/chemistry , Protozoan Proteins/chemistry
18.
mBio ; 8(4)2017 08 08.
Article in English | MEDLINE | ID: mdl-28790200

ABSTRACT

The human malaria parasite Plasmodium falciparum replicates within circulating red blood cells, where it is subjected to conditions that frequently cause DNA damage. The repair of DNA double-stranded breaks (DSBs) is thought to rely almost exclusively on homologous recombination (HR), due to a lack of efficient nonhomologous end joining. However, given that the parasite is haploid during this stage of its life cycle, the mechanisms involved in maintaining genome stability are poorly understood. Of particular interest are the subtelomeric regions of the chromosomes, which contain the majority of the multicopy variant antigen-encoding genes responsible for virulence and disease severity. Here, we show that parasites utilize a competitive balance between de novo telomere addition, also called "telomere healing," and HR to stabilize chromosome ends. Products of both repair pathways were observed in response to DSBs that occurred spontaneously during routine in vitro culture or resulted from experimentally induced DSBs, demonstrating that both pathways are active in repairing DSBs within subtelomeric regions and that the pathway utilized was determined by the DNA sequences immediately surrounding the break. In combination, these two repair pathways enable parasites to efficiently maintain chromosome stability while also contributing to the generation of genetic diversity.IMPORTANCE Malaria is a major global health threat, causing approximately 430,000 deaths annually. This mosquito-transmitted disease is caused by Plasmodium parasites, with infection with the species Plasmodium falciparum being the most lethal. Mechanisms underlying DNA repair and maintenance of genome integrity in P. falciparum are not well understood and represent a gap in our understanding of how parasites survive the hostile environment of their vertebrate and insect hosts. Our work examines DNA repair in real time by using single-molecule real-time (SMRT) sequencing focused on the subtelomeric regions of the genome that harbor the multicopy gene families important for virulence and the maintenance of infection. We show that parasites utilize two competing molecular mechanisms to repair double-strand breaks, homologous recombination and de novo telomere addition, with the pathway used being determined by the surrounding DNA sequence. In combination, these two pathways balance the need to maintain genome stability with the selective advantage of generating antigenic diversity.


Subject(s)
Chromosomes/genetics , Genome, Protozoan/genetics , Genomic Instability , Plasmodium falciparum/genetics , Telomere/genetics , Animals , Antigenic Variation , DNA Breaks, Double-Stranded , DNA Damage , Gene Conversion , Genetic Variation , Homologous Recombination , Humans , Plasmodium falciparum/pathogenicity , Sequence Analysis, DNA/methods
19.
J Clin Microbiol ; 55(10): 2903-2912, 2017 10.
Article in English | MEDLINE | ID: mdl-28747374

ABSTRACT

Babesia microti, a zoonotic intraerythrocytic parasite, is the primary etiological agent of human babesiosis in the United States. Human infections range from subclinical illness to severe disease resulting in death, with symptoms being related to host immune status. Despite advances in our understanding and management of B. microti, the incidence of infection in the United States has increased. Therefore, research focused on eradicating disease and optimizing clinical management is essential. Here we review this remarkable organism, with emphasis on the clinical, diagnostic, and therapeutic aspects of human disease.


Subject(s)
Antiparasitic Agents/therapeutic use , Babesia microti/immunology , Babesiosis/diagnosis , Babesiosis/drug therapy , Tick-Borne Diseases/epidemiology , Tick-Borne Diseases/transmission , Animals , Babesiosis/epidemiology , Babesiosis/transmission , Hemolysis/physiology , Humans , Ixodes/parasitology , Mice , Tick-Borne Diseases/diagnosis , Tick-Borne Diseases/parasitology , United States/epidemiology
20.
Clin Infect Dis ; 65(7): 1222-1225, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28541469

ABSTRACT

Babesiosis treatment failures with standard therapy have been reported, but the molecular mechanisms are not well understood. We describe the emergence of atovaquone and azithromycin resistance associated with mutations in the binding regions of the target proteins of both drugs during treatment of an immunosuppressed patient with relapsing babesiosis.


Subject(s)
Antiprotozoal Agents/therapeutic use , Atovaquone/therapeutic use , Azithromycin/therapeutic use , Babesiosis/drug therapy , Drug Resistance/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/parasitology , Rituximab/therapeutic use , Aged, 80 and over , Amino Acid Sequence , Babesia microti/drug effects , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...