Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Chembiochem ; 24(7): e202200533, 2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-36449557

RESUMO

Inhibiting the formation of a tight junction between two malaria parasite proteins, apical membrane antigen 1 and rhoptry neck protein 2, crucial for red blood cell invasion, prevents progression of the disease. In this work, we have used a unique approach to design a chimeric peptide, prepared by fusion of the best features of two peptide inhibitors, that has displayed parasite growth inhibition ex vivo with nanomolar IC50 , which is 100 times better than any of its parent peptides. Furthermore, to gain structural insights, we computationally modelled the hybrid peptide on its receptor.


Assuntos
Plasmodium falciparum , Proteínas de Protozoários , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Antígenos de Protozoários/química , Antígenos de Protozoários/metabolismo , Proteínas de Membrana/química , Peptídeos/química , Eritrócitos/metabolismo
2.
Mol Microbiol ; 115(5): 1025-1038, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33538363

RESUMO

Sirtuins (PfSIR2A and PfSIR2B) are implicated to play pivotal roles in the silencing of sub-telomeric genes and the maintenance of telomere length in P. falciparum 3D7 strain. Here, we identify the key factors that regulate the cellular abundance and activity of these two histone deacetylases. Our results demonstrate that PfSIR2A and PfSIR2B are transcriptionally downregulated at the mid-ring stage in response to febrile temperature. We found that the molecular chaperone PfHsp90 acts as a repressor of PfSIR2A & B transcription. By virtue of its presence in the PfSIR2A & B promoter proximal regions PfHsp90 helps recruiting H3K9me3, conferring heterochromatic state, and thereby leading to the downregulation of PfSIR2A & B transcription. Such transcriptional downregulation can be reversed by the addition of 17-(allylamino)-17-demethoxygeldanamycin or Radicicol, two potent inhibitors of PfHsp90. The reduced occupancy of PfSir2 at sub-telomeric var promoters leads to the de-repression of var genes. Thus, here we uncover how exposure to febrile temperature, a hallmark of malaria, enables the parasites to manipulate the expression of the two prominent epigenetic modifiers PfSir2A and PfSir2B.


Assuntos
Epigênese Genética , Proteínas de Choque Térmico HSP90/metabolismo , Malária Falciparum/parasitologia , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Sirtuínas/genética , Regulação para Baixo , Proteínas de Choque Térmico HSP90/genética , Humanos , Plasmodium falciparum/metabolismo , Regiões Promotoras Genéticas , Proteínas de Protozoários/metabolismo , Sirtuínas/metabolismo , Temperatura
3.
Antimicrob Agents Chemother ; 65(9): e0045721, 2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34097485

RESUMO

The DNA recombinase Rad51 from the human malaria parasite Plasmodium falciparum has emerged as a potential drug target due to its central role in the homologous recombination (HR)-mediated double-strand break (DSB) repair pathway. Inhibition of the ATPase and strand exchange activity of P. falciparum Rad51 (PfRad51) by a small-molecule inhibitor, B02 [3-(phenylmethyl)-2-[(1E)-2-(3-pyridinyl)ethenyl]-4(3H)-quinazolinone], renders the parasite more sensitive to genotoxic agents. Here, we investigated whether the inhibition of the molecular chaperone PfHsp90 potentiates the antimalarial action of B02. We found that the PfHsp90 inhibitor 17-AAG [17-(allylamino)-17-demethoxygeldanamycin] exhibits strong synergism with B02 in both drug-sensitive (strain 3D7) and multidrug-resistant (strain Dd2) P. falciparum parasites. 17-AAG causes a greater than 200-fold decrease in the half-maximal inhibitory concentration (IC50) of B02 in 3D7 parasites. Our results provide mechanistic insights into such profound synergism between 17-AAG and B02. We report that PfHsp90 physically interacts with PfRad51 and promotes the UV irradiation-induced DNA repair activity of PfRad51 by controlling its stability. We find that 17-AAG reduces PfRad51 protein levels by accelerating proteasomal degradation. Consequently, PfHsp90 inhibition renders the parasites more susceptible to the potent DNA-damaging agent methyl methanesulfonate (MMS) in a dose-dependent manner. Thus, our study provides a rationale for targeting PfHsp90 along with the recombinase PfRad51 for controlling malaria propagation.


Assuntos
Malária Falciparum , Malária , Parasitos , Animais , Dano ao DNA , Humanos , Plasmodium falciparum/genética
4.
J Biol Chem ; 294(20): 8171-8183, 2019 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-30936202

RESUMO

Malaria parasites repair DNA double-strand breaks (DSBs) primarily through homologous recombination (HR). Here, because the unrepaired DSBs lead to the death of the unicellular parasite Plasmodium falciparum, we investigated its recombinase, PfRad51, as a potential drug target. Undertaking an in silico screening approach, we identified a compound, B02, that docks to the predicted tertiary structure of PfRad51 with high affinity. B02 inhibited a drug-sensitive P. falciparum strain (3D7) and multidrug-resistant parasite (Dd2) in culture, with IC50 values of 8 and 3 µm, respectively. We found that B02 is more potent against these P. falciparum strains than against mammalian cell lines. Our findings also revealed that the antimalarial activity of B02 synergizes with those of two first-line malaria drugs, artemisinin (ART) and chloroquine (CQ), lowering the IC50 values of ART and CQ by 15- and 8-fold, respectively. Our results also provide mechanistic insights into the anti-parasitic activity of B02, indicating that it blocks the ATPase and strand-exchange activities of PfRad51 and abrogates the formation of PfRad51 foci on damaged DNA at chromosomal sites, probably by blocking homomeric interactions of PfRad51 proteins. The B02-mediated PfRad51 disruption led to the accumulation of unrepaired parasitic DNA and rendered parasites more sensitive to DNA-damaging agents, including ART. Our findings provide a rationale for targeting the Plasmodium DSB repair pathway in combination with ART. We propose that identification of a specific inhibitor of HR in Plasmodium may enable investigations of HR's role in Plasmodium biology, including generation of antigenic diversity.


Assuntos
Antimaláricos , Artemisininas , Cloroquina , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Inibidores Enzimáticos , Lactonas , Plasmodium falciparum/enzimologia , Proteínas de Protozoários/antagonistas & inibidores , Rad51 Recombinase/antagonistas & inibidores , Antimaláricos/química , Antimaláricos/farmacologia , Artemisininas/agonistas , Artemisininas/química , Artemisininas/farmacologia , Cloroquina/agonistas , Cloroquina/química , Cloroquina/farmacologia , Simulação por Computador , Sinergismo Farmacológico , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Lactonas/agonistas , Lactonas/química , Lactonas/farmacologia , Simulação de Acoplamento Molecular , Plasmodium falciparum/genética , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Rad51 Recombinase/química , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo
5.
J Vector Borne Dis ; 57(4): 325-330, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-34856712

RESUMO

BACKGROUND AND OBJECTIVES: The mitochondrial electron transport chain (mtETC) of Plasmodium falciparum is an important drug target. Identification and functional validation of putative mitochondrial proteins of the mtETC is critical for drug development. Many of the regulatory subunits and assembly factors of cytochrome c oxidase readily identifiable in humans and yeast are missing in P. falciparum. Here, we describe our efforts to identify and validate the function of putative Pfsurf1, a key assembly factor of complex IV of the mtETC. METHODS: Multiple sequence alignment of SURF 1/Shy 1 was carried out in Clustal X 2.1. Phylogenetic tree was constructed using "Draw tree" option in Clustal X, and was analyzed using interactive Tree of Life software. To identify the conserved sequences, domain search was done using Jalview version 2.8.2 (BLOSUM 62 scoring). The haploid Saccharomyces cerevisiae strain (BY4741) containing the null allele shy1 (Orf: YGR112w) (shy1::Kan) was complemented with putative Pfsurf1 to study its ability to rescue the growth defect. RESULTS: Similarity searches of PfSURF1-like protein in the Pfam shows statistically significant E = 4.7e-10 match to SURF1 family. Sequence alignment of PfSURF1 with other SURF1-like proteins reveals the conservation of transmembrane domains, α-helices and ß-pleated sheets. Phylogenetic analysis clusters putative PfSURF1 with apicomplexan SURF1-like proteins. Yeast complementation studies show that Pfsurf1 can partially rescue the yeast shy1 mutant, YGR112w. INTERPRETATION & CONCLUSION: Bioinformatics and complementation studies in yeast show that P. falciparum's SURF1 is the functional ortholog of human SURF1 and yeast Shy1.


Assuntos
Plasmodium falciparum , Saccharomyces cerevisiae , Complexo IV da Cadeia de Transporte de Elétrons/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Filogenia , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo
6.
Eukaryot Cell ; 14(1): 64-77, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25380755

RESUMO

The inhibition of Hsp90 in cancerous cells has been correlated with the reduction in double-strand break (DSB repair) activity. However, the precise effect of Hsp90 on the DSB repair pathway in normal cells has remained enigmatic. Our results show that the Hsp82 chaperone, the ortholog of mammalian Hsp90, is indispensable for homologous-recombination (HR)-mediated DNA repair in the budding yeast Saccharomyces cerevisiae. A considerable reduction in cell viability is observed in an Hsp82-inactivated mutant upon methyl methanesulfonate (MMS) treatment as well as upon UV treatment. The loss of Hsp82 function results in a dramatic decrease in gene-targeting efficiency and a marked decrease in the endogenous levels of the key recombination proteins Rad51 and Rad52 without any notable change in the levels of RAD51 or RAD52 transcripts. Our results establish Rad51 as a client of Hsp82, since they interact physically in vivo, and also show that when Hsp82 is inhibited by 17-AAG, Rad51 undergoes proteasomal degradation. By analyzing a number of point mutants with mutations in different domains of Hsp82, we observe a strong association between the sensitivity of an ATPase mutant of Hsp82 to DNA damage and the decreases in the amounts of Rad51 and Rad52 proteins. The most significant observations include the dramatic abrogation of HR activity and the marked decrease in Rad51 focus formation in the charged linker deletion mutant of Hsp82 upon MMS treatment. The charged linker region of Hsp82 is evolutionarily conserved in all eukaryotes, but until now, no biological significance has been assigned to it. Our findings elucidate the importance of this region in DNA repair for the first time.


Assuntos
Reparo do DNA , Proteínas de Choque Térmico HSP90/metabolismo , Rad51 Recombinase/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Motivos de Aminoácidos , Domínio Catalítico , Proteínas de Choque Térmico HSP90/química , Proteínas de Choque Térmico HSP90/genética , Ligação Proteica , Proteólise , Proteína Rad52 de Recombinação e Reparo de DNA/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética
7.
Mol Microbiol ; 94(2): 353-66, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25145341

RESUMO

Malaria parasites survive through repairing a plethora of DNA double-stranded breaks (DSBs) experienced during their asexual growth. In Plasmodium Rad51 mediated homologous recombination (HR) mechanism and homology-independent alternative end-joining mechanism have been identified. Here we address whether loss of HR activity can be compensated by other DSB repair mechanisms. Creating a transgenic Plasmodium line defective in HR function, we demonstrate that HR is the most important DSB repair pathway in malarial parasite. Using mouse malaria model we have characterized the dominant negative effect of PfRad51(K143R) mutant on Plasmodium DSB repair and host-parasite interaction. Our work illustrates that Plasmodium berghei harbouring the mutant protein (PfRad51(K143R)) failed to repair DSBs as evidenced by hypersensitivity to DNA-damaging agent. Mice infected with mutant parasites lived significantly longer with markedly reduced parasite burden. To better understand the effect of mutant PfRad51(K143R) on HR, we used yeast as a surrogate model and established that the presence of PfRad51(K143R) completely inhibited DNA repair, gene conversion and gene targeting. Biochemical experiment confirmed that very low level of mutant protein was sufficient for complete disruption of wild-type PfRad51 activity. Hence our work provides evidence that HR pathway of Plasmodium could be efficiently targeted to curb malaria.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Recombinação Homóloga , Proteínas Mutantes/metabolismo , Plasmodium berghei/enzimologia , Rad51 Recombinase/metabolismo , Sequência de Aminoácidos , Animais , Modelos Animais de Doenças , Interações Hospedeiro-Parasita , Malária/parasitologia , Camundongos , Dados de Sequência Molecular , Proteínas Mutantes/genética , Carga Parasitária , Plasmodium berghei/genética , Rad51 Recombinase/genética , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Análise de Sobrevida
8.
Antimicrob Agents Chemother ; 58(8): 4341-52, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24841259

RESUMO

Radicicol, an antifungal antibiotic, was previously identified as a compound having antimalarial activity. However, its mechanism of action in Plasmodium falciparum was not elucidated. While characterizing its antimalarial function, we observed that radicicol manifested two distinct developmental defects in cultured P. falciparum in a concentration-dependent manner. At a low concentration of radicicol, a significant percentage of drug-treated parasites were arrested at the schizont stage, while at a higher concentration, the parasites were unable to multiply from schizont to ring. Also, the newly formed rings and trophozoites were extremely delayed in development, eventually leading to cell death. We intended to characterize the potential molecular target of radicicol at its sublethal doses. Our results demonstrated that radicicol specifically impaired mitochondrial replication. This decrement was associated with a severalfold increment of the topoisomerase VIB transcript as well as protein in treated cells over that of untreated parasites. Topoisomerase VIB was found to be localized in the organelle fraction. Our docking study revealed that radicicol fits into the Bergerat fold of Pf topoisomerase VIB present in its ATPase domain. Altogether, these data allow us to conclude that P. falciparum topoisomerase VIB might be one of the targets of radicicol causing inhibition of mitochondrial replication. Hence, radicicol can be suitably employed to explore the mitochondrial physiology of malaria parasites.


Assuntos
Antimaláricos/farmacologia , Macrolídeos/farmacologia , Renovação Mitocondrial/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Esquizontes/efeitos dos fármacos , Proteínas Arqueais/antagonistas & inibidores , Proteínas Arqueais/química , Proteínas Arqueais/genética , Proteínas Arqueais/metabolismo , DNA Topoisomerases Tipo II/química , DNA Topoisomerases Tipo II/genética , DNA Topoisomerases Tipo II/metabolismo , Relação Dose-Resposta a Droga , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Expressão Gênica , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Mitocôndrias/genética , Simulação de Acoplamento Molecular , Plasmodium falciparum/enzimologia , Plasmodium falciparum/genética , Plasmodium falciparum/crescimento & desenvolvimento , Conformação Proteica , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Esquizontes/enzimologia , Esquizontes/crescimento & desenvolvimento , Trofozoítos/efeitos dos fármacos , Trofozoítos/enzimologia , Trofozoítos/crescimento & desenvolvimento
9.
Front Mol Biosci ; 10: 1223682, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37593128

RESUMO

The human malaria parasite Plasmodium falciparum maintains the chronicity of infections through antigenic variation, a well-coordinated immune evasion mechanism. The most prominent molecular determinant of antigenic variation in this parasite includes the members of the var multigene family. Homologous recombination (HR)-mediated genomic rearrangements have been implicated to play a major role in var gene diversification. However, the key molecular factors involved in the generation of diversity at var loci are less known. Here, we tested the hypothesis that PfRad51 could carry out recombination between var genes that are not homologous but homeologous in nature. We employed the whole-genome sequencing (WGS) approach to investigate recombination events among var sequences over 100 generations and compared the rate of sequence rearrangement at the var loci in both PfRad51-proficient and -deficient parasite lines. This brief report provides evidence that the loss of the key recombinase function renders the parasite with inefficient HR and results in fewer recombination events among the var sequences, thereby impacting the diversification of the var gene repertoire.

11.
mSphere ; 7(5): e0032922, 2022 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-36121150

RESUMO

Sir2 protein of Plasmodium falciparum has been implicated to play crucial roles in the silencing of subtelomeric var genes and rRNA. It is also involved in telomere length maintenance. Epigenetic regulation of PfSIR2 transcription occurs through a direct participation of the molecular chaperon PfHsp90, wherein PfHsp90 acts as a transcriptional repressor. However, whether the chaperonic activity of PfHsp90 is essential for the maturation and stability of PfSir2A protein has not yet been explored. Here, we show that PfSir2A protein is a direct client of PfHsp90. We demonstrate that PfHsp90 physically interacts with PfSir2A, and the inhibition of PfHsp90 activity via chemical inhibitors, such as 17-AAG or Radicicol, results in the depletion of PfSir2A protein, and consequently its histone deacetylase activity. Thus, derepression of var genes and ribosomal silencing were observed under PfHsp90 inactivation. This finding that PfHsp90 provides stability to PfSir2A protein, in addition to the previous finding that PfHsp90 downregulates PfSIR2A transcription and subsequently cellular abundance, uncovers the multifaceted roles of PfHsp90 in regulating PfSir2 abundance and activity. Given the importance of PfSir2 protein in Plasmodium biology, it is reasonable to propose that the PfHsp90-PfSir2 axis can be exploited as a novel druggable target. IMPORTANCE Malaria continues to severely impact the global public health not only due to the mortality and morbidity associated with it, but also because of the huge burden on the world economy it imparts. Despite the intensive vaccine-research and drug-development programs, there is not a single effective vaccine suitable for all age groups, and there is no drug on the market against which resistance is not developed. Thus, there is an urgent need to develop novel intervention strategies by identifying the crucial targets from Plasmodium biology. Here, we uncover that the molecular chaperone PfHsp90 regulates the abundance and activity of the histone-deacetylase PfSir2, a prominent regulator of Plasmodium epigenome. Given that PfSir2 controls both virulence and multiplicity of the parasite, and that PfHsp90 is an essential chaperone involved in diverse cellular processes, our findings argue that the PfHsp90-PfSir2 axis could be targeted to curb malaria.


Assuntos
Proteínas de Choque Térmico HSP90 , Histona Desacetilases , Plasmodium falciparum , Humanos , Epigênese Genética , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Histonas/genética , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Plasmodium falciparum/enzimologia
12.
mSphere ; 6(6): e0071821, 2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34730376

RESUMO

The homologous recombination (HR) pathway has been implicated as the predominant mechanism for the repair of chromosomal DNA double-strand breaks (DSBs) of the malarial parasite. Although the extrachromosomal mitochondrial genome of this parasite experiences a greater number of DSBs due to its close proximity to the electron transport chain, nothing is known about the proteins involved in the repair of the mitochondrial genome. We investigated the involvement of nucleus-encoded HR proteins in the repair of the mitochondrial genome, as this genome does not code for any DNA repair proteins. Here, we provide evidence that the nucleus-encoded "recombinosome" of the parasite is also involved in mitochondrial genome repair. First, two crucial HR proteins, namely, Plasmodium falciparum Rad51 (PfRad51) and P. falciparum Bloom helicase (PfBlm) are located in the mitochondria. They are recruited to the mitochondrial genome at the schizont stage, a stage that is prone to DSBs due to exposure to various endogenous and physiologic DNA-damaging agents. Second, the recruitment of these two proteins to the damaged mitochondrial genome coincides with the DNA repair kinetics. Moreover, both the proteins exit the mitochondrial DNA (mtDNA) once the genome is repaired. Most importantly, the specific chemical inhibitors of PfRad51 and PfBlm block the repair of UV-induced DSBs of the mitochondrial genome. Additionally, overexpression of these two proteins resulted in a kinetically faster repair. Given the essentiality of the mitochondrial genome, blocking its repair by inhibiting the HR pathway could offer a novel strategy for curbing malaria. IMPORTANCE The impact of malaria on global public health and the world economy continues to surge despite decades of vaccine research and drug development efforts. An alarming rise in resistance toward all the commercially available antimalarial drugs and the lack of an effective malaria vaccine brings us to the urge to identify novel intervention strategies for curbing malaria. Here, we uncover the molecular mechanism behind the repair of the most deleterious form of DNA lesions on the parasitic mitochondrial genome. Given that the single-copy mitochondrion is an indispensable organelle of the malaria parasite, we propose that targeting the mitochondrial DNA repair pathways should be exploited as a potential malaria control strategy. The establishment of the parasitic homologous recombination machinery as the predominant repair mechanism of the mitochondrial DNA double-strand breaks underscores the importance of this pathway as a novel druggable target.


Assuntos
Antimaláricos/farmacologia , Genoma Mitocondrial/efeitos dos fármacos , Plasmodium falciparum/enzimologia , Proteínas de Protozoários/antagonistas & inibidores , Rad51 Recombinase/antagonistas & inibidores , RecQ Helicases/metabolismo , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Recombinação Homóloga , Humanos , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo
13.
Biochem Biophys Rep ; 26: 100950, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33665380

RESUMO

Apical membrane antigen 1 (AMA1) is a surface protein of Plasmodium sp. that plays a crucial role in forming moving junction (MJ) during the invasion of human red blood cells. The obligatory presence of AMA1 in the parasite lifecycle designates this protein as a potential vaccine candidate and an essential target for the development of novel peptide or protein therapeutics. However, due to multiple cysteine residues in the protein sequence, attaining the native fold with correct disulfide linkages during the refolding process after expression in bacteria has remained challenging for years. Although several approaches to obtain the refolded protein from bacterial expression have been reported previously, achieving high yield during refolding and proper functional validation of the expressed protein was lacking. We report here an improved method of refolding to obtain higher quantity of refolded protein. We have also validated the refolded protein's functional activity by evaluating the expressed AMA1 protein binding with a known inhibitory peptide, rhoptry neck protein 2 (RON2), using surface plasmon resonance (SPR) and isothermal titration calorimetry (ITC).

14.
mSphere ; 5(6)2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33239368

RESUMO

Artemisinin (ART)-based combination therapies are recommended as first- and second-line treatments for Plasmodium falciparum malaria. Here, we investigated the impact of the RecQ inhibitor ML216 on the repair of ART-mediated damage in the genome of P. falciparumPfBLM and PfWRN were identified as members of the RecQ helicase family in P. falciparum However, the role of these RecQ helicases in DNA double-strand break (DSB) repair in this parasite has not been explored. Here, we provide several lines of evidence to establish the involvement of PfBlm in DSB repair in P. falciparum First, we demonstrate that PfBlm interacts with two well-characterized DSB repair proteins of this parasite, namely, PfRad51 and PfalMre11. Second, we found that PfBLM expression was upregulated in response to DNA-damaging agents. Third, through yeast complementation studies, we demonstrated that PfBLM could complement the DNA damage sensitivity of a Δsgs1 mutant of Saccharomyces cerevisiae, in contrast to the helicase-dead mutant PfblmK83R Finally, we observe that the overexpression of PfBLM induces resistance to DNA-damaging agents and offers a survival advantage to the parasites. Most importantly, we found that the RecQ inhibitor ML216 inhibits the repair of DSBs and thereby renders parasites more sensitive to ART. Such synergism between ART and ML216 actions was observed for both drug-sensitive and multidrug-resistant strains of P. falciparum Taken together, these findings establish the implications of PfBlm in the Plasmodium DSB repair pathway and provide insights into the antiparasitic activity of the ART-ML216 combination.IMPORTANCE Malaria continues to be a serious threat to humankind not only because of the morbidity and mortality associated with the disease but also due to the huge economic burden that it imparts. Resistance to all available drugs and the unavailability of an effective vaccine cry for an urgent discovery of newer drug targets. Here, we uncovered a role of the PfBlm helicase in Plasmodium DNA double-strand break repair and established that the parasitic DNA repair mechanism can be targeted to curb malaria. The small-molecule inhibitor of PfBlm tested in this study acts synergistically with two first-line malaria drugs, artemisinin (ART) and chloroquine, in both drug-sensitive and multidrug-resistant strains of P. falciparum, thus qualifying this chemical as a potential partner in ART-based combination therapy. Additionally, the identification of this new specific inhibitor of the Plasmodium homologous recombination (HR) mechanism will now allow us to investigate the role of HR in Plasmodium biology.


Assuntos
Artemisininas/farmacologia , Cloroquina/farmacologia , Reparo do DNA/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , RecQ Helicases/metabolismo , Antimaláricos/farmacologia , Inibidores Enzimáticos , Humanos , Malária Falciparum/parasitologia , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , RecQ Helicases/genética
15.
Indian J Med Microbiol ; 38(2): 213-215, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32883936

RESUMO

Prohibitins (PHBs) are evolutionarily conserved mitochondrial integral membrane proteins, shown to regulate mitochondrial structure and function, and can be classified into PHB1 and PHB2. PHB1 and PHB2 have been shown to interact with each other, and form heterodimers in mitochondrial inner membrane. Plasmodium falciparum has orthologues of PHB1 and PHB2 in its genome, and their role is unclear. Here, by homology modelling and yeast two-hybrid analysis, we show that putative Plasmodium PHBs (Pf PHB1 and Pf PHB2) interact with each other, which suggests that they could form supercomplexes of heterodimers in Plasmodium, the functional form required for optimum mitochondrial function.


Assuntos
Proteínas de Membrana/química , Plasmodium falciparum/química , Proteínas de Protozoários/química , Proteínas Repressoras/química , Mitocôndrias/metabolismo , Modelos Moleculares , Plasmodium falciparum/genética , Proibitinas , Conformação Proteica , Multimerização Proteica , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae , Técnicas do Sistema de Duplo-Híbrido
16.
Org Lett ; 21(22): 9040-9044, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31663760

RESUMO

The thioester surrogate 3,4-diaminobenzoic acid (Dbz) facilitates the efficient synthesis of peptide thioesters by Fmoc chemistry solid phase peptide synthesis and the optional attachment of a solubility tag at the C-terminus. The protection of the partially deactivated ortho-amine of Dbz is necessary to obtain contamination-free peptide synthesis. The reported carbamate protecting groups promote a serious side reaction, benzimidazolinone formation. Herein we introduce the Boc-protected Dbz that prevents the benzimidazolinone formation, leading to clean peptide o-aminoanilides suitable for the total chemical synthesis of proteins.


Assuntos
Anilidas/química , Peptídeos/química , Proteínas de Protozoários/síntese química , Ubiquitina/síntese química , Benzimidazóis
17.
mSphere ; 1(1)2016.
Artigo em Inglês | MEDLINE | ID: mdl-27303712

RESUMO

Plasmodium falciparum topoisomerase VIB (TopoVIB)-TopoVIA (TopoVIB-VIA) complex can be potentially exploited as a drug target against malaria due to its absence from the human genome. Previous work in our laboratory has suggested that P. falciparum TopoVIB (PfTopoVIB) might be a target of radicicol since treatment of parasite cultures with this antibiotic is associated with upregulation of Plasmodium TopoVIB at the transcript level as well as at the protein level. Further studies demonstrated that radicicol treatment impaired mitochondrial replication of human malaria parasite P. falciparum. However, the technical challenge associated with the expression of the above protein complex hampered its functional characterization. Using Saccharomyces cerevisiae as a heterologous system, we expressed PfTopoVIB (Myc-tagged) and PfTopoVIA (Flag-tagged) (PfTopoVIB-VIA) proteins. Yeast two-hybrid analysis showed the formation of PfTopoVIB homodimers and PfTopoVIB/PfTopoVIA heteromers. Our study demonstrated that PfTopoVIB and PfTopoVIA together can rescue the lethal phenotype of yeast ΔtopoII mutants, whereas Plasmodium topoisomerase VIB alone cannot. Using yeast cell-free extracts harboring the PfTopoVIB-VIA protein complex, we have performed a decatenation assay and observed that PfTopoVIB-VIA can decatenate DNA in an ATP- and Mg(2+)-dependent manner. The specificity of this enzyme is established by abrogation of its activity in the presence of PfTopoVIB-specific antibody. Our study results show that radicicol and etoposide can specifically inhibit PfTopoVIB-VIA decatenation activity whereas the gyrase inhibitor novobiocin cannot. Such a yeast-based assay system can be employed in screening specific inhibitors against Plasmodium VIB-VIA. IMPORTANCE In this study we characterize topoisomerase VI from Plasmodium falciparum using genetic and biochemical approaches. We use various inhibitors and identify radicicol as a specific inhibitor of its decatenation activity. We establish a very simple and economical biochemical assay system that can be exploited to screen inhibitors of PfTopoVI.

18.
Mol Biochem Parasitol ; 139(1): 33-9, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15610817

RESUMO

Although homologous recombination-mediated DNA rearrangements are quite widespread in Plasmodium falciparum, the molecular mechanisms involved are essentially unknown. Recent identification of PfRad51 in P. falciparum has suggested that it may play central role during homologous recombination and DNA rearrangements. Full-length recombinant PfRad51 was over expressed in Escherichia coli and purified to near homogeneity. Using optimized enzymatic activity conditions recombinant PfRad51 protein was shown to catalyze DNA strand-exchange reaction, a central step during homologous recombination. Unlike bacterial RecA protein, PfRad51 promoted strand-exchange reaction does not require ATP hydrolysis. The PfRad51 protein also catalyzed ssDNA-dependent ATP hydrolysis and the k(cat) values were similar to those reported for human Rad51. The demonstration of strand-exchange activity of PfRad51 protein, first such report in any protozoan parasite, suggests importance of similar recombination mechanism during DNA rearrangements associated with antigenic variation in P. falciparum.


Assuntos
Trifosfato de Adenosina/metabolismo , DNA de Protozoário/metabolismo , Plasmodium falciparum/enzimologia , Proteínas de Protozoários/metabolismo , Recombinases/metabolismo , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/isolamento & purificação , Adenosina Trifosfatases/metabolismo , Animais , Variação Antigênica , Clonagem Molecular , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Cinética , Proteínas de Protozoários/genética , Proteínas de Protozoários/isolamento & purificação , Rad51 Recombinase , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Recombinases/genética , Recombinases/isolamento & purificação , Especificidade por Substrato
19.
PLoS One ; 10(5): e0125358, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25938776

RESUMO

The eukaryotic Meiotic Recombination protein 11 (Mre11) plays pivotal roles in the DNA damage response (DDR). Specifically, Mre11 senses and signals DNA double strand breaks (DSB) and facilitates their repair through effector proteins belonging to either homologous recombination (HR) or non-homologous end joining (NHEJ) repair mechanisms. In the human malaria parasite Plasmodium falciparum, HR and alternative-NHEJ have been identified; however, little is known about the upstream factors involved in the DDR of this organism. In this report, we identify a putative ortholog of Mre11 in P. falciparum (PfalMre11) that shares 22% sequence similarity to human Mre11. Homology modeling reveals striking structural resemblance of the predicted PfalMre11 nuclease domain to the nuclease domain of Saccharomyces cerevisiae Mre11 (ScMre11). Complementation analyses reveal functional conservation of PfalMre11 nuclease activity as demonstrated by the ability of the PfalMre11 nuclease domain, in conjunction with the C-terminal domain of ScMre11, to functionally complement an mre11 deficient yeast strain. Functional complementation was virtually abrogated by an amino acid substitution in the PfalMre11 nuclease domain (D398N). PfalMre11 is abundant in the mitotically active trophozoite and schizont stages of P. falciparum and is up-regulated in response to DNA damage, suggesting a role in the DDR. PfalMre11 exhibits physical interaction with PfalRad50. In addition, yeast 2-hybrid studies show that PfalMre11 interacts with ScRad50 and ScXrs2, two important components of the well characterized Mre11-Rad50-Xrs2 complex which is involved in DDR signaling and repair in S. cerevisiae, further supporting a role for PfalMre11 in the DDR. Taken together, these findings provide evidence that PfalMre11 is an evolutionarily conserved component of the DDR in Plasmodium.


Assuntos
Evolução Biológica , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Anticorpos Antiprotozoários/imunologia , Dano ao DNA , Reparo do DNA por Junção de Extremidades , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/imunologia , Eritrócitos/parasitologia , Regulação da Expressão Gênica , Teste de Complementação Genética , Humanos , Estágios do Ciclo de Vida , Modelos Moleculares , Dados de Sequência Molecular , Plasmodium falciparum/crescimento & desenvolvimento , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia , Proteínas Recombinantes , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
20.
Int J Parasitol ; 33(12): 1385-92, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14527521

RESUMO

Rad51 protein, the eukaryotic homologue of Escherichia coli RecA protein plays a pivotal role in recombinational repair mechanism. We have identified a new homologue of Rad51 from the apicomplexan parasite Plasmodium falciparum, designated PfRad51. The PfRad51 gene codes for a 351 amino acid polypeptide with a predicted molecular mass of 38720, and shares 66-75% sequence identity within the catalytic region with Rad51 from human, yeast and other protozoan parasites such as Trypanosoma and Leishmania. The expression of PfRad51 transcript as well as protein in the intra-erythrocytic in vitro culture of P. flalciparum was found to be up-regulated in response to the DNA damaging agent methyl methanesulfonate, suggesting its functional involvement in recombinational repair process. PfRad51 is the first apicomplexan gene identified that codes for a recombination protein, and it offers an excellent model for studying DNA damage inducible gene expression in such parasites.


Assuntos
Reparo do DNA , Genes de Protozoários , Plasmodium falciparum/genética , Proteínas de Protozoários , RNA Mensageiro/análise , Recombinases/genética , Animais , Sequência de Bases , Northern Blotting/métodos , Western Blotting/métodos , Clonagem Molecular , Dano ao DNA , Expressão Gênica/efeitos dos fármacos , Humanos , Leishmania major/genética , Metanossulfonato de Metila/farmacologia , Dados de Sequência Molecular , Mutagênicos/farmacologia , Plasmodium falciparum/efeitos dos fármacos , Testes de Precipitina , Rad51 Recombinase , Saccharomyces/genética , Alinhamento de Sequência , Trypanosoma brucei brucei/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA