Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochem Pharmacol ; 203: 115154, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35798201

RESUMEN

The development of resistance to current antimalarial therapies remains a significant source of concern. To address this risk,newdrugswithnoveltargetsin distinct developmental stages ofPlasmodiumparasites are required. In the current study,we have targetedP. falciparumTubulin(PfTubulin)proteins which represent some of thepotentialdrug targetsfor malaria chemotherapy. PlasmodialMicrotubules (MTs) play a crucial role during parasite proliferation, growth, and transmission, which render them highlydesirabletargets for the development ofnext-generation chemotherapeutics. Towards this,we have evaluated the antimalarial activity ofTubulintargetingcompounds received from theMedicines for Malaria Venture (MMV)"Pathogen Box"against the human malaria parasite,P. falciparumincluding 3D7 (chloroquine and artemisinin sensitive strain), RKL-9 (chloroquine-resistant strain), and R539T (artemisinin-resistant strain). At nanomolar concentrations, the filtered-out compounds exhibitedpronouncedmultistage antimalarialeffects across the parasite life cycle, including intra-erythrocytic blood stages, liver stage parasites, gametocytes, and ookinetes. Concomitantly, these compoundswere found toimpedemale gamete ex-flagellation, thus showingtheir transmission-blocking potential. Target mining of these potent compounds, by combining in silico, biochemical and biophysical assays,implicatedPfTubulinas their moleculartarget, which may possibly act bydisruptingMT assembly dynamics by binding at the interface of α-ßTubulin-dimer.Further, the promising ADME profile of the parent scaffold supported its consideration as a lead compound for further development.Thus, our work highlights the potential of targetingPfTubulin proteins in discovering and developing next-generation, multistage antimalarial agents against Multi-Drug Resistant (MDR) malaria parasites.


Asunto(s)
Antimaláricos , Artemisininas , Malaria , Acceso a la Información , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Artemisininas/farmacología , Cloroquina/farmacología , Humanos , Malaria/tratamiento farmacológico , Plasmodium falciparum/metabolismo , Tubulina (Proteína)/metabolismo
2.
Emerg Microbes Infect ; 11(1): 938-955, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35264080

RESUMEN

The family of apicomplexan specific proteins contains caspases-like proteins called "metacaspases". These enzymes are present in the malaria parasite but absent in human; therefore, these can be explored as potential drug targets. We deleted the MCA-2 gene from Plasmodium berghei genome using a gene knockout strategy to decipher its precise function. This study has identified that MCA-2 plays an important role in parasite transmission since it is critical for the formation of gametocytes and for maintaining an appropriate number of infectious sporozoites required for sporogony. It is noticeable that a significant reduction in gametocyte, oocysts, ookinete and sporozoites load along with a delay in hepatocytes invasion were observed in the MCA-2 knockout parasite. Furthermore, a study found the two MCA-2 inhibitory molecules known as C-532 and C-533, which remarkably inhibited the MCA-2 activity, abolished the in vitro parasite growth, and also impaired the transmission cycle of P. falciparum and P. berghei in An. stephensi. Our findings indicate that the deletion of MCA-2 hampers the Plasmodium development during erythrocytic and exo-erythrocytic stages, and its inhibition by C-532 and C-533 critically affects the malaria transmission biology.


Asunto(s)
Malaria , Proteínas Protozoarias , Animales , Gametogénesis , Humanos , Malaria/parasitología , Plasmodium berghei/genética , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Esporozoítos/metabolismo
3.
Exp Cell Res ; 406(2): 112764, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34358525

RESUMEN

Protein kinases of both the parasite and the host are crucial in parasite invasion and survival and might act as drug targets against drug-resistant malaria. STK35L1 was among the top five hits in kinome-wide screening, suggesting its role in malaria's liver stage. However, the role of host STK35L1 in malaria remains elusive. In this study, we found that STK35L1 was highly upregulated during the infection of Plasmodium berghei (P. berghei) in HepG2 cells and mice liver, and knockdown of STK35L1 remarkably suppressed the sporozoites' infection in HepG2 cells. We showed that STAT3 is upregulated and phosphorylated during P. berghei sporozoites' infection, and STAT3 activation is required for both the upregulation of STK35L1 and STAT3. Furthermore, we found that ten cell cycle genes were upregulated in the sporozoite-infected hepatocytes. Knockdown of STK35L1 inhibited the basal expression of these genes except CDKN3 and GTSE1 in HepG2 cells. Thus, we identified STK35L1 as a host kinase that plays an obligatory role in malaria's liver stage and propose that it may serve as a potential drug target against drug-resistant malaria.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Hígado/parasitología , Malaria/parasitología , Plasmodium berghei/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Transcripción STAT3/metabolismo , Esporozoítos/fisiología , Animales , Proteínas de Ciclo Celular/genética , Femenino , Regulación de la Expresión Génica , Células Hep G2 , Humanos , Hígado/metabolismo , Malaria/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/genética , Factor de Transcripción STAT3/genética
4.
Toxins (Basel) ; 13(1)2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33467515

RESUMEN

Severe malaria caused by Plasmodium falciparum poses a major global health problem with high morbidity and mortality. P. falciparum harbors a family of pore-forming proteins (PFPs), known as perforin like proteins (PLPs), which are structurally equivalent to prokaryotic PFPs. These PLPs are secreted from the parasites and, they contribute to disease pathogenesis by interacting with host cells. The severe malaria pathogenesis is associated with the dysfunction of various barrier cells, including endothelial cells (EC). Several factors, including PLPs secreted by parasites, contribute to the host cell dysfunction. Herein, we have tested the hypothesis that PLPs mediate dysfunction of barrier cells and might have a role in disease pathogenesis. We analyzed various dysfunctions in barrier cells following rPLP2 exposure and demonstrate that it causes an increase in intracellular Ca2+ levels. Additionally, rPLP2 exposed barrier cells displayed features of cell death, including Annexin/PI positivity, depolarized the mitochondrial membrane potential, and ROS generation. We have further performed the time-lapse video microscopy of barrier cells and found that the treatment of rPLP2 triggers their membrane blebbing. The cytoplasmic localization of HMGB1, a marker of necrosis, further confirmed the necrotic type of cell death. This study highlights the role of parasite factor PLP in endothelial dysfunction and provides a rationale for the design of adjunct therapies against severe malaria.


Asunto(s)
Células Endoteliales/parasitología , Malaria Falciparum/parasitología , Necrosis/parasitología , Perforina/efectos adversos , Proteínas Protozoarias/efectos adversos , Animales , Apoptosis , Biomarcadores/metabolismo , Barrera Hematoencefálica , Calcio/metabolismo , Línea Celular , Permeabilidad de la Membrana Celular , Supervivencia Celular , Perros , Eritrocitos/parasitología , Proteína HMGB1/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células de Riñón Canino Madin Darby , Membranas Mitocondriales , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidad , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes
5.
J Biochem ; 170(5): 639-653, 2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-35670460

RESUMEN

Programmed cell death (PCD) is a multi-step process initiated by a set of proteases, which interacts and cleaves diverse proteins, thus modulating their biochemical and cellular functions. In metazoans, PCD is mediated by proteolytic enzymes called caspases, which triggered cell death by proteolysis of human Tudor staphylococcus nuclease (TSN). Non-metazoans lack a close homologue of caspases but possess an ancestral family of cysteine proteases termed 'metacaspases'. Studies supported that metacaspases are involved in PCD, but their natural substrates remain unknown. In this study, we performed the Plasmodium falciparum TSN (PfTSN) cleavage assay using wild and selected mutants of P. falciparum metacaspases-2 (PfMCA-2) in vitro and in vivo. Interestingly, PfMCA-2, cleaved a phylogenetically conserved protein, PfTSN at multiple sites. Deletion or substitution mutation in key interacting residues at the active site, Cys157 and His205 of PfMCA-2, impaired its enzymatic activity with the artificial substrate, z-GRR-AMC. However, the mutant Tyr224A did not affect the activity with z-GRR-AMC but abolished the cleavage of PfTSN. These results indicated that the catalytic dyad, Cys157 and His205 of PfMCA-2 was essential for its enzymatic activity with an artificial substrate, whereas Tyr224 and Cys157 residues were responsible for its interaction with the natural substrate and subsequent degradation of PfTSN. Our results suggested that MCA-2 interacts with TSN substrate in a non-canonical way using non-conserved or conformationally available residues for its binding and cleavage. In future, it would be interesting to explore how this interaction leads to the execution of PCD in the Plasmodium.


Asunto(s)
Caspasas , Plasmodium falciparum , Apoptosis/fisiología , Caspasas/genética , Caspasas/metabolismo , Dominio Catalítico , Muerte Celular , Humanos
6.
Front Immunol ; 12: 699887, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34987497

RESUMEN

The Plasmodium parasite has to cross various immunological barriers for successful infection. Parasites have evolved mechanisms to evade host immune responses, which hugely contributes to the successful infection and transmission by parasites. One way in which a parasite evades immune surveillance is by expressing molecular mimics of the host molecules in order to manipulate the host responses. In this study, we report a Plasmodium berghei hypothetical protein, PbTIP (PbANKA_124360.0), which is a Plasmodium homolog of the human T-cell immunomodulatory protein (TIP). The latter possesses immunomodulatory activities and suppressed the host immune responses in a mouse acute graft-versus-host disease (GvHD) model. The Plasmodium berghei protein, PbTIP, is expressed on the merozoite surface and exported to the host erythrocyte surface upon infection. It is shed in the blood circulation by the activity of an uncharacterized membrane protease(s). The shed PbTIP could be detected in the host serum during infection. Our results demonstrate that the shed PbTIP exhibits binding on the surface of macrophages and reduces their inflammatory cytokine response while upregulating the anti-inflammatory cytokines such as TGF-ß and IL-10. Such manipulated immune responses are observed in the later stage of malaria infection. PbTIP induced Th2-type gene transcript changes in macrophages, hinting toward its potential to regulate the host immune responses against the parasite. Therefore, this study highlights the role of a Plasmodium-released protein, PbTIP, in immune evasion using macrophages, which may represent the critical strategy of the parasite to successfully survive and thrive in its host. This study also indicates the human malaria parasite TIP as a potential diagnostic molecule that could be exploited in lateral flow-based immunochromatographic tests for malaria disease diagnosis.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Evasión Inmune/inmunología , Inmunidad Innata , Macrófagos/parasitología , Malaria/inmunología , Plasmodium berghei/inmunología , Proteínas Protozoarias/fisiología , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Citocinas/biosíntesis , Citocinas/genética , Membrana Eritrocítica/química , Eritrocitos/parasitología , Humanos , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Malaria/parasitología , Ratones , Ratones Endogámicos C57BL , Imitación Molecular , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/inmunología , Proteínas Protozoarias/inmunología , Células RAW 264.7 , Proteínas Recombinantes/farmacología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transcriptoma
7.
Artículo en Inglés | MEDLINE | ID: mdl-32266171

RESUMEN

The pore forming Plasmodium Perforin Like Proteins (PPLP), expressed in all stages of the parasite life cycle are critical for completion of the parasite life cycle. The high sequence similarity in the central Membrane Attack Complex/ Perforin (MACPF) domain among PLPs and their distinct functional overlaps define them as lucrative target for developing multi-stage antimalarial therapeutics. Herein, we evaluated the mechanism of Pan-active MACPF Domain (PMD), a centrally located and highly conserved region of PPLPs, and deciphered the inhibitory potential of specifically designed PMD inhibitors. The E. coli expressed rPMD interacts with erythrocyte membrane and form pores of ~10.5 nm height and ~24.3 nm diameter leading to hemoglobin release and dextran uptake. The treatment with PMD induced erythrocytes senescence which can be hypothesized to account for the physiological effect of disseminated PLPs in loss of circulating erythrocytes inducing malaria anemia. The anti-PMD inhibitors effectively blocked intraerythrocytic growth by suppressing invasion and egress processes and protected erythrocytes against rPMD induced senescence. Moreover, these inhibitors also blocked the hepatic stage and transmission stage parasite development suggesting multi-stage, transmission-blocking potential of these inhibitors. Concievably, our study has introduced a novel set of anti-PMD inhibitors with pan-inhibitory activity against all the PPLPs members which can be developed into potent cross-stage antimalarial therapeutics along with erythrocyte senescence protective potential to occlude PPLPs mediated anemia in severe malaria.


Asunto(s)
Escherichia coli , Plasmodium , Membrana Celular , Eritrocitos , Perforina , Plasmodium falciparum , Proteínas Protozoarias
8.
Biochem J ; 477(7): 1323-1344, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32202613

RESUMEN

Metacaspases are novel cysteine proteases found in apicomplexan whose function is poorly understood. Our earlier studies on Plasmodium falciparum metacaspase-2 (PfMCA-2) revealed that the caspase inhibitor, Z-FA-FMK efficiently inhibited PfMCA-2 activity and, expression, and significantly blocked in vitro progression of the parasite developmental cycle via apoptosis-like parasite death. Building on these findings, we synthesized a set of novel inhibitors based on structural modification of Z-FA-FMK with the amides of piperic acid and investigated their effect on PfMCA-2. One of these analogs, SS-5, specifically inhibited the activity and expression of PfMCA-2. The activities of some other known malarial proteases (falcipains, plasmepsins and vivapain), and human cathepsins-B, D and L, and caspase-3 and -7 were not inhibited by SS-5. SS-5 blocked the development of P. falciparum in vitro (IC50 1 µM) and caused prominent morphological distortions. Incubation with SS-5 led to persistent parasite oxidative stress accompanied by depolarization of mitochondrial potential and accumulation of intracellular Ca2+. SS-5 also inhibited the development of P. berghei in a murine model. Our results suggest that the inhibition of PfMCA-2 results in oxidative stress, leading to apoptosis-like parasite death. Thus, SS-5 offers a starting point for the optimization of new antimalarials, and PfMCA-2 could be a novel target for antimalarial drug discovery.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Dipéptidos/farmacología , Cetonas/farmacología , Plasmodium falciparum/enzimología , Amidas/química , Animales , Antimaláricos/química , Antimaláricos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Supervivencia Celular/efectos de los fármacos , Dipéptidos/química , Descubrimiento de Drogas/métodos , Ácidos Grasos Insaturados/química , Femenino , Células Hep G2 , Humanos , Cetonas/química , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Estrés Oxidativo/efectos de los fármacos
9.
Immun Inflamm Dis ; 8(1): 50-61, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31967737

RESUMEN

INTRODUCTION: Efforts are required at developing an effective vaccine that can inhibit malaria prevalence and transmission. Identifying the critical immunogenic antigens and understanding their interactions with host proteins forms a major focus of subunit vaccine development. Previously, our laboratory showed that SLTRiP conferred protection to the liver stage of Plasmodium growth in rodents. In the follow-up of earlier research, we demonstrate that SLTRiP-mediated protection is majorly concentrated in specific regions of protein. METHOD: To identify particular protective regions of protein, we synthesized multiple nonoverlapping fragments from SLTRiP protein. From this, we designed a panel of 8-20mer synthetic peptides, which were predicted using T-epitope-based prediction algorithm. We utilized the IFN-γ enzyme-linked immunosorbent spot assay to identify immunodominant peptides. The latter were used to immunize mice, and these mice were challenged to assess protection. RESULTS: The protective polypeptide fragment SLTRiP C3 and SLTRiP C4 were identified, by expressing and testing multiple fragments of PbSLTRiP protein. The immune responses generated by these fragments were compared to identify the immunodominant fragment. The T-epitopes were predicted from SLTRiP protein using computer-based algorithms. The in vitro immune responses generated by these peptides were compared with each other to identify the immunodominant T-epitope. Immunization using these peptides showed significant reduction in parasite numbers during liver stage. CONCLUSION: Our findings show that the protective efficacy shown by SLTRiP is localized in particular protein fragments. The peptides designed from such regions showed protective efficacy equivalent to whole protein. The sequence conservation analysis with human Plasmodium species also showed that these peptides were conserved. In conclusion, these peptides or their equivalent from other Plasmodium species could impart protection against malaria in their respective hosts too. Our studies provide a basis for the inclusion of these peptides in clinical vaccine constructs against malaria.


Asunto(s)
Antígenos de Protozoos/inmunología , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Vacunas contra la Malaria/inmunología , Secuencia de Aminoácidos , Animales , Antígenos de Protozoos/química , Mapeo Epitopo , Femenino , Malaria/inmunología , Malaria/parasitología , Malaria/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Plasmodium falciparum
10.
IUBMB Life ; 71(9): 1293-1301, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30865364

RESUMEN

Proliferative cell nuclear antigen (PCNA) is the processivity factor for various DNA polymerases and it functions in response to DNA damage in eukaryotic system. Plasmodium falciparum contains two PCNAs, while PCNA1 has been attributed to DNA replication, the role of PCNA2 has been assigned to DNA damage response in erythrocytic developmental stages. Although a recent transposon mediated knockout strategy qualified pcna2 as a nonessential gene in Plasmodium berghei, a conventional homologous recombination-based knockout strategy has not been employed for this gene yet. Moreover, the cellular dynamics of PCNA2 in extraerythrocytic stages still remain elusive in Plasmodium. We attempted multiple times to knock out PbPCNA2 from the parasite genome using homologous recombination strategy without much success. However, we were able to generate PbPCNA2-GFP tagged transgenic parasites confirming that the pcna2 locus is amenable to genetic manipulation. The GFP-tagged parasites showed similar growth phenotype, compared to wild-type parasites, in both erythrocytic and sporogonic cycle, suggesting that tagging had no effect on parasite physiology. PbPCNA2 expression was also observed during the sporogonic cycle in midgut oocyst and salivary gland sporozoites. The PbPCNA2 expression was upregulated in the presence of DNA damaging agents like hydroxyurea and methyl methanesulphonate. Our inability to knock out PCNA2 suggested its essentiality in the parasite development and elevated expression during DNA damaging condition hint at a critical role of the protein in parasite physiology. © 2019 IUBMB Life, 71(9):1293-1301, 2019.


Asunto(s)
Daño del ADN/genética , Plasmodium berghei/genética , Antígeno Nuclear de Célula en Proliferación/genética , Proteínas Protozoarias/genética , Animales , Replicación del ADN/genética , ADN Polimerasa Dirigida por ADN/genética , Regulación de la Expresión Génica/genética , Técnicas de Inactivación de Genes , Genoma/genética , Humanos , Plasmodium berghei/patogenicidad , Plasmodium falciparum/genética
11.
Cell Death Dis ; 10(3): 161, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30770800

RESUMEN

Recent studies have documented the diverse role of host immunity in infection by the protozoan parasite, Toxoplasma gondii. However, the contribution of the ß-catenin pathway in this process has not been explored. Here, we show that AKT-mediated phosphorylated ß-catenin supports T. gondii multiplication which is arrested in the deficiency of its phosphorylation domain at S552 position. The ß-catenin-TCF4 protein complex binds to the promoter region of IRF3 gene and initiates its transcription, which was also abrogated in ß-catenin knockout cells. TBK-independent phosphorylation of STING(S366) and its adaptor molecule TICAM2 by phospho-AKT(T308S473) augmented downstream IRF3-dependent IDO1 transcription, which was also dependent on ß-catenin. But, proteasomal degradation of IDO1 by its tyrosine phosphorylation (at Y115 and Y253) favoured parasite replication. In absence of IDO1, tryptophan was catabolized into melatonin, which supressed cellular reactive oxygen species (ROS) and boosted parasite growth. Conversely, when tyrosine phosphorylation was abolished by phosphosite mutations, IDO1 escaped its ubiquitin-mediated proteasomal degradation system (UPS) and the stable IDO1 prevented parasite replication by kynurenine synthesis. We propose that T. gondii selectively utilizes tryptophan to produce the antioxidant, melatonin, thus prolonging the survival of infected cells through functional AKT and ß-catenin activity for better parasite replication. Stable IDO1 in the presence of IFN-γ catabolized tryptophan into kynurenine, promoting cell death by suppressing phospho-AKT and phospho-ß-catenin levels, and circumvented parasite replication. Treatment of infected cells with kynurenine or its analogue, teriflunomide suppressed kinase activity of AKT, and phosphorylation of ß-catenin triggering caspase-3 dependent apoptosis of infected cells to inhibit parasite growth. Our results demonstrate that ß-catenin regulate phosphorylated STING-TICAM2-IRF3-IDO1 signalosome for a cell-intrinsic pro-parasitic role. We propose that the downstream IRF3-IDO1-reliant tryptophan catabolites and their analogues can act as effective immunotherapeutic molecules to control T. gondii replication by impairing the AKT and ß-catenin axis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Quinurenina/metabolismo , Proteínas de la Membrana/metabolismo , Toxoplasma/metabolismo , Toxoplasmosis/metabolismo , Triptófano/metabolismo , beta Catenina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Células CACO-2 , Crotonatos/farmacología , Técnicas de Inactivación de Genes , Humanos , Hidroxibutiratos , Quinurenina/farmacología , Ratones , Nitrilos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células RAW 264.7 , Toluidinas/farmacología , Toxoplasma/crecimiento & desarrollo , Toxoplasmosis/parasitología , Transcripción Genética , Transfección , beta Catenina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...