Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Antimicrob Agents Chemother ; 68(5): e0169023, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38501806

RESUMO

Malaria tropica, caused by the parasite Plasmodium falciparum (P. falciparum), remains one of the greatest public health burdens for humankind. Due to its pivotal role in parasite survival, the energy metabolism of P. falciparum is an interesting target for drug design. To this end, analysis of the central metabolite adenosine triphosphate (ATP) is of great interest. So far, only cell-disruptive or intensiometric ATP assays have been available in this system, with various drawbacks for mechanistic interpretation and partly inconsistent results. To address this, we have established fluorescent probes, based on Förster resonance energy transfer (FRET) and known as ATeam, for use in blood-stage parasites. ATeams are capable of measuring MgATP2- levels in a ratiometric manner, thereby facilitating in cellulo measurements of ATP dynamics in real-time using fluorescence microscopy and plate reader detection and overcoming many of the obstacles of established ATP analysis methods. Additionally, we established a superfolder variant of the ratiometric pH sensor pHluorin (sfpHluorin) in P. falciparum to monitor pH homeostasis and control for pH fluctuations, which may affect ATeam measurements. We characterized recombinant ATeam and sfpHluorin protein in vitro and stably integrated the sensors into the genome of the P. falciparum NF54attB cell line. Using these new tools, we found distinct sensor response patterns caused by several different drug classes. Arylamino alcohols increased and redox cyclers decreased ATP; doxycycline caused first-cycle cytosol alkalization; and 4-aminoquinolines caused aberrant proteolysis. Our results open up a completely new perspective on drugs' mode of action, with possible implications for target identification and drug development.


Assuntos
Trifosfato de Adenosina , Antimaláricos , Transferência Ressonante de Energia de Fluorescência , Plasmodium falciparum , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/metabolismo , Plasmodium falciparum/genética , Trifosfato de Adenosina/metabolismo , Antimaláricos/farmacologia , Transferência Ressonante de Energia de Fluorescência/métodos , Corantes Fluorescentes/química , Humanos , Quinina/farmacologia , Doxiciclina/farmacologia , Artemisininas/farmacologia , Cloroquina/farmacologia , Concentração de Íons de Hidrogênio
2.
Int J Mol Sci ; 24(8)2023 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-37108438

RESUMO

During their life cycle, apicomplexan parasites pass through different microenvironments and encounter a range of ion concentrations. The discovery that the GPCR-like SR25 in Plasmodium falciparum is activated by a shift in potassium concentration indicates that the parasite can take advantage of its development by sensing different ionic concentrations in the external milieu. This pathway involves the activation of phospholipase C and an increase in cytosolic calcium. In the present report, we summarize the information available in the literature regarding the role of potassium ions during parasite development. A deeper understanding of the mechanisms that allow the parasite to cope with ionic potassium changes contributes to our knowledge about the cell cycle of Plasmodium spp.


Assuntos
Parasitos , Plasmodium , Toxoplasma , Animais , Toxoplasma/metabolismo , Parasitos/metabolismo , Plasmodium falciparum/metabolismo , Potássio/metabolismo , Proteínas de Protozoários/metabolismo
3.
Front Mol Biosci ; 9: 968248, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35992276

RESUMO

As part of their life-cycle, malaria parasites undergo rapid cell multiplication and division, with one parasite giving rise to over 20 new parasites within the course of 48 h. To support this, the parasite has an extremely high metabolic rate and level of protein biosynthesis. Underpinning these activities, the parasite encodes a number of chaperone/heat shock proteins, belonging to various families. Research over the past decade has revealed that these proteins are involved in a number of essential processes within the parasite, or within the infected host cell. Due to this, these proteins are now being viewed as potential targets for drug development, and we have begun to characterize their properties in more detail. In this article we summarize the current state of knowledge about one particular chaperone family, that of the HSP70, and highlight their importance, function, and potential co-chaperone interactions. This is then discussed with regard to the suitability of these proteins and interactions for drug development.

4.
Cell Rep ; 39(2): 110691, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35417716

RESUMO

The innate immune recognition of the malaria-causing pathogen Plasmodium falciparum (P. falciparum) is not fully explored. Here, we identify the nucleoside 5'-methylthioinosine (MTI), a Plasmodium-specific intermediate of the purine salvage pathway, as a pathogen-derived Toll-like receptor 8 (TLR8) agonist. Co-incubation of MTI with the TLR8 enhancer poly(dT) as well as synthetic or P. falciparum-derived RNA strongly increase its stimulatory activity. Of note, MTI generated from methylthioadenosine (MTA) by P. falciparum lysates activates TLR8 when MTI metabolism is inhibited by immucillin targeting the purine nucleoside phosphorylase (PfPNP). Importantly, P. falciparum-infected red blood cells incubated with MTI or cultivated with MTA and immucillin lead to TLR8-dependent interleukin-6 (IL-6) production in human monocytes. Our data demonstrate that the nucleoside MTI is a natural human TLR8 ligand with possible in vivo relevance for innate sensing of P. falciparum.


Assuntos
Malária Falciparum , Metiltioinosina , Receptor 8 Toll-Like , Humanos , Metiltioinosina/análogos & derivados , Nucleosídeos , Plasmodium falciparum/metabolismo , Purina-Núcleosídeo Fosforilase/metabolismo , Purinas , Receptor 8 Toll-Like/metabolismo
5.
Antioxid Redox Signal ; 37(1-3): 1-18, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35072524

RESUMO

Aims: Genetically encoded green fluorescent protein (GFP)-based redox biosensors are widely used to monitor specific and dynamic redox processes in living cells. Over the last few years, various biosensors for a variety of applications were engineered and enhanced to match the organism and cellular environments, which should be investigated. In this context, the unicellular intraerythrocytic parasite Plasmodium, the causative agent of malaria, represents a challenge, as the small size of the organism results in weak fluorescence signals that complicate precise measurements, especially for cell compartment-specific observations. To address this, we have functionally and structurally characterized an enhanced redox biosensor superfolder roGFP2 (sfroGFP2). Results: SfroGFP2 retains roGFP2-like behavior, yet with improved fluorescence intensity (FI) in cellulo. SfroGFP2-based redox biosensors are pH insensitive in a physiological pH range and show midpoint potentials comparable with roGFP2-based redox biosensors. Using crystallography and rigidity theory, we identified the superfolding mutations as being responsible for improved structural stability of the biosensor in a redox-sensitive environment, thus explaining the improved FI in cellulo. Innovation: This work provides insight into the structure and function of GFP-based redox biosensors. It describes an improved redox biosensor (sfroGFP2) suitable for measuring oxidizing effects within small cells where applicability of other redox sensor variants is limited. Conclusion: Improved structural stability of sfroGFP2 gives rise to increased FI in cellulo. Fusion to hGrx1 (human glutaredoxin-1) provides the hitherto most suitable biosensor for measuring oxidizing effects in Plasmodium. This sensor is of major interest for studying glutathione redox changes in small cells, as well as subcellular compartments in general. Antioxid. Redox Signal. 37, 1-18.


Assuntos
Técnicas Biossensoriais , Glutationa , Plasmodium , Técnicas Biossensoriais/métodos , Glutationa/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Oxirredução , Plasmodium/isolamento & purificação
6.
Parasitol Int ; 87: 102513, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34785370

RESUMO

Plasmodium falciparum macrophage migration inhibitory factor (PfMIF) is a homologue of the multifunctional human host cytokine MIF (HsMIF). Upon schizont rupture it is released into the human blood stream where it acts as a virulence factor, modulating the host immune system. Whereas for HsMIF a tautomerase, an oxidoreductase, and a nuclease activity have been identified, the latter has not yet been studied for PfMIF. Furthermore, previous studies identified PfMIF as a target for several redox post-translational modifications. Therefore, we analysed the impact of S-glutathionylation and S-nitrosation on the protein's functions. To determine the impact of the four cysteines of PfMIF we produced His-tagged cysteine to alanine mutants of PfMIF via site-directed mutagenesis. Recombinant proteins were analysed via mass spectrometry, and enzymatic assays. Here we show for the first time that PfMIF acts as a DNase of human genomic DNA and that this activity is greater than that shown by HsMIF. Moreover, we observed a significant decrease in the maximum velocity of the DCME tautomerase activity of PfMIF upon alanine replacement of Cys3, and Cys3/Cys4 double mutant. Lastly, using a yeast reporter system, we were able to verify binding of PfMIF to the human chemokine receptors CXCR4, and demonstrate a so-far overlooked binding to CXCR2, both of which function as non-cognate receptors for HsMIF. While S-glutathionylation and S-nitrosation of PfMIF did not impair the tautomerase activity of PfMIF, activation of these receptors was significantly decreased.


Assuntos
Cisteína/deficiência , Fatores Inibidores da Migração de Macrófagos/química , Fatores Inibidores da Migração de Macrófagos/genética , Plasmodium falciparum/química , Alanina/química , Cisteína/genética , Desoxirribonucleases/metabolismo , Humanos , Plasmodium falciparum/genética , Proteínas Recombinantes/genética
7.
PLoS Pathog ; 17(10): e1009969, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34614006

RESUMO

The pathology associated with malaria infection is largely due to the ability of infected human RBCs to adhere to a number of receptors on endothelial cells within tissues and organs. This phenomenon is driven by the export of parasite-encoded proteins to the host cell, the exact function of many of which is still unknown. Here we inactivate the function of one of these exported proteins, PFA66, a member of the J-domain protein family. Although parasites lacking this protein were still able to grow in cell culture, we observed severe defects in normal host cell modification, including aberrant morphology of surface knobs, disrupted presentation of the cytoadherence molecule PfEMP1, and a total lack of cytoadherence, despite the presence of the knob associated protein KAHRP. Complementation assays demonstrate that an intact J-domain is required for recovery to a wild-type phenotype and suggest that PFA66 functions in concert with a HSP70 to carry out host cell modification. Strikingly, this HSP70 is likely to be of host origin. ATPase assays on recombinant protein verify a functional interaction between PFA66 and residual host cell HSP70. Taken together, our data reveal a role for PFA66 in host cell modification, strongly implicate human HSP70s as being essential in this process and uncover a new KAHRP-independent molecular factor required for correct knob biogenesis.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Interações Hospedeiro-Parasita/fisiologia , Malária Falciparum/metabolismo , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/metabolismo , Humanos , Malária Falciparum/parasitologia , Malária Falciparum/patologia , Plasmodium falciparum/metabolismo , Virulência
8.
Adv Exp Med Biol ; 1340: 1-9, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34569019

RESUMO

Malaria did not die with the end of the age of western colonization but is still a major public health issue in large parts of the world. Despite repeated and concerted efforts to eradicate this disease, it has proved remarkably resilient, and constant vigilance and continuous research are required to discover new chinks in the parasite's armor and alleviate the suffering at both the individual and societal levels. A deeper understanding of the fundamental processes underlying parasite survival, propagation, virulence, and ability to cause disease is the key to the development of desperately needed new therapies and prophylactic drugs. Malaria parasites, by the nature of their lifecycle, are subject to a number of environmental and cellular stresses which they must overcome to survive. To this end, they express a number of heat shock proteins (HSPs), molecules specialized on buffering the effects of external stimuli, but which are also essential for normal cellular biochemistry. In this introductory chapter, I give a brief overview of the diversity of structure, function, and importance of these HSPs, and highlight some of the current and future research questions in this field. Additionally, this chapter acts as a bridge to the other chapters in this book. These chapters, I think you will agree, demonstrate that with regard to HSPs malaria parasites, as in so many things, obey the adage "Same same, but different."


Assuntos
Malária , Parasitos , Animais , Proteínas de Choque Térmico/genética , Malária/tratamento farmacológico , Plasmodium falciparum/genética , Proteínas de Protozoários/genética
9.
Mol Biochem Parasitol ; 242: 111362, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33513391

RESUMO

Plasmodium falciparum causes the deadliest form of malaria. Adequate redox control is crucial for this protozoan parasite to overcome oxidative and nitrosative challenges, thus enabling its survival. Sulfenylation is an oxidative post-translational modification, which acts as a molecular on/off switch, regulating protein activity. To obtain a better understanding of which proteins are redox regulated in malaria parasites, we established an optimized affinity capture protocol coupled with mass spectrometry analysis for identification of in vivo sulfenylated proteins. The non-dimedone based probe BCN-Bio1 shows reaction rates over 100-times that of commonly used dimedone-based probes, allowing for a rapid trapping of sulfenylated proteins. Mass spectrometry analysis of BCN-Bio1 labeled proteins revealed the first insight into the Plasmodium falciparum trophozoite sulfenylome, identifying 102 proteins containing 152 sulfenylation sites. Comparison with Plasmodium proteins modified by S-glutathionylation and S-nitrosation showed a high overlap, suggesting a common core of proteins undergoing redox regulation by multiple mechanisms. Furthermore, parasite proteins which were identified as targets for sulfenylation were also identified as being sulfenylated in other organisms, especially proteins of the glycolytic cycle. This study suggests that a number of Plasmodium proteins are subject to redox regulation and it provides a basis for further investigations into the exact structural and biochemical basis of regulation, and a deeper understanding of cross-talk between post-translational modifications.


Assuntos
Compostos Bicíclicos com Pontes/química , Sondas Moleculares/química , Plasmodium falciparum/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas de Protozoários/metabolismo , Ácidos Sulfênicos/metabolismo , Trofozoítos/metabolismo , Células Cultivadas , Cisteína/metabolismo , Eritrócitos/parasitologia , Ontologia Genética , Glutationa/metabolismo , Humanos , Espectrometria de Massas , Anotação de Sequência Molecular , Compostos Nitrosos/metabolismo , Oxirredução , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Coloração e Rotulagem/métodos , Trofozoítos/genética
10.
Sci Rep ; 11(1): 2057, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33479315

RESUMO

The host hormone melatonin is known to modulate the asexual cell-cycle of the human malaria parasite Plasmodium falciparum and the kinase PfPK7 is fundamental in the downstream signaling pathways. The nuclear protein PfMORC displays a histidine kinase domain and is involved in parasite cell cycle control. By using a real-time assay, we show a 24 h (h) rhythmic expression of PfMORC at the parasite asexual cycle and the expression is dramatically changed when parasites were treated with 100 nM melatonin for 17 h. Moreover, PfMORC expression was severely affected in PfPK7 knockout (PfPK7-) parasites following melatonin treatment. Parasites expressing 3D7morc-GFP shows nuclear localization of the protein during the asexual stage of parasite development. Although the PfMORC knockdown had no significant impact on the parasite proliferation in vitro it significantly changed the ratio of the different asexual intraerythrocytic stages of the parasites upon the addition of melatonin. Our data reveal that in addition to the upstream melatonin signaling pathways such as IP3 generation, calcium, and cAMP rise, a nuclear protein, PfMORC is essential for the hormone response in parasite synchronization.


Assuntos
Malária Falciparum/genética , Proteínas Nucleares/genética , Plasmodium falciparum/genética , Animais , Eritrócitos/parasitologia , Humanos , Malária Falciparum/parasitologia , Melatonina/genética , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/genética , Reprodução Assexuada/genética
11.
Sci Rep ; 10(1): 4193, 2020 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-32144363

RESUMO

The regulation of human Arf1 GTPase activity by ArfGEFs that stimulate GDP/GTP exchange and ArfGAPs that mediate GTP hydrolysis has attracted attention for the discovery of Arf1 inhibitors as potential anti-cancer agents. The malaria parasite Plasmodium falciparum encodes a Sec7 domain-containing protein - presumably an ArfGEF - and two putative ArfGAPs, as well as an Arf1 homologue (PfArf1) that is essential for blood-stage parasite viability. However, ArfGEF and ArfGAP-mediated activation/deactivation of PfArf1 has not been demonstrated. In this study, we established an in vitro colorimetric microtiter plate-based assay to detect the activation status of truncated human and P. falciparum Arf1 and used it to demonstrate the activation of both proteins by the Sec7 domain of ARNO, their deactivation by the GAP domain of human ArfGAP1 and the inhibition of the respective reactions by the compounds SecinH3 and QS11. In addition, we found that the GAP domains of both P. falciparum ArfGAPs have activities equivalent to that of human ArfGAP1, but are insensitive to QS11. Library screening identified a novel inhibitor which selectively inhibits one of the P. falciparum GAP domains (IC50 4.7 µM), suggesting that the assay format is suitable for screening compound collections for inhibitors of Arf1 regulatory proteins.


Assuntos
Proteínas de Bactérias/metabolismo , Bioensaio/métodos , Colorimetria/métodos , Proteínas Ativadoras de GTPase/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Bactérias/química , Proteínas Ativadoras de GTPase/química , Guanosina Trifosfato/metabolismo , Humanos , Hidrólise
12.
Cell Microbiol ; 22(3): e13146, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31734953

RESUMO

Patatin-like phospholipases (PNPLAs) are highly conserved enzymes of prokaryotic and eukaryotic organisms with major roles in lipid homeostasis. The genome of the malaria parasite Plasmodium falciparum encodes four putative PNPLAs with predicted functions during phospholipid degradation. We here investigated the role of one of the plasmodial PNPLAs, a putative PLA2 termed PNPLA1, during blood stage replication and gametocyte development. PNPLA1 is present in the asexual and sexual blood stages and here localizes to the cytoplasm. PNPLA1-deficiency due to gene disruption or conditional gene-knockdown had no effect on intraerythrocytic growth, gametocyte development and gametogenesis. However, parasites lacking PNPLA1 were impaired in gametocyte induction, while PNPLA1 overexpression promotes gametocyte formation. The loss of PNPLA1 further leads to transcriptional down-regulation of genes related to gametocytogenesis, including the gene encoding the sexual commitment regulator AP2-G. Additionally, lipidomics of PNPLA1-deficient asexual blood stage parasites revealed overall increased levels of major phospholipids, including phosphatidylcholine (PC), which is a substrate of PLA2 . PC synthesis is known to be pivotal for erythrocytic replication, while the reduced availability of PC precursors drives the parasite into gametocytogenesis; we thus hypothesize that the higher PC levels due to PNPLA1-deficiency prevent the blood stage parasites from entering the sexual pathway.


Assuntos
Fosfolipases/fisiologia , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/fisiologia , Animais , Citoplasma/genética , Citoplasma/metabolismo , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Genoma de Protozoário , Estágios do Ciclo de Vida , Metabolismo dos Lipídeos , Camundongos , Fosfolipases/genética , Plasmodium falciparum/enzimologia , Proteínas de Protozoários/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
13.
Biomolecules ; 9(7)2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31340488

RESUMO

The survival of the human malaria parasite Plasmodium falciparum under the physiologically distinct environments associated with their development in the cold-blooded invertebrate mosquito vectors and the warm-blooded vertebrate human host requires a genome that caters to adaptability. To this end, a robust stress response system coupled to an efficient protein quality control system are essential features of the parasite. Heat shock proteins constitute the main molecular chaperone system of the cell, accounting for approximately two percent of the malaria genome. Some heat shock proteins of parasites constitute a large part (5%) of the 'exportome' (parasite proteins that are exported to the infected host erythrocyte) that modify the host cell, promoting its cyto-adherence. In light of their importance in protein folding and refolding, and thus the survival of the parasite, heat shock proteins of P. falciparum have been a major subject of study. Emerging evidence points to their role not only being cyto-protection of the parasite, as they are also implicated in regulating parasite virulence. In undertaking their roles, heat shock proteins operate in networks that involve not only partners of parasite origin, but also potentially functionally associate with human proteins to facilitate parasite survival and pathogenicity. This review seeks to highlight these interplays and their roles in parasite pathogenicity. We further discuss the prospects of targeting the parasite heat shock protein network towards the developments of alternative antimalarial chemotherapies.


Assuntos
Proteínas de Choque Térmico/metabolismo , Plasmodium falciparum/metabolismo , Animais , Humanos , Plasmodium falciparum/patogenicidade , Virulência
14.
PLoS Pathog ; 14(8): e1007249, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30133543

RESUMO

The complex life-cycle of the human malaria parasite Plasmodium falciparum requires a high degree of tight coordination allowing the parasite to adapt to changing environments. One of the major challenges for the parasite is the human-to-mosquito transmission, which starts with the differentiation of blood stage parasites into the transmissible gametocytes, followed by the rapid conversion of the gametocytes into gametes, once they are taken up by the blood-feeding Anopheles vector. In order to pre-adapt to this change of host, the gametocytes store transcripts in stress granules that encode proteins needed for parasite development in the mosquito. Here we report on a novel stress granule component, the seven-helix protein 7-Helix-1. The protein, a homolog of the human stress response regulator LanC-like 2, accumulates in stress granules of female gametocytes and interacts with ribonucleoproteins, such as CITH, DOZI, and PABP1. Malaria parasites lacking 7-Helix-1 are significantly impaired in female gametogenesis and thus transmission to the mosquito. Lack of 7-Helix-1 further leads to a deregulation of components required for protein synthesis. Consistently, inhibitors of translation could mimic the 7-Helix-1 loss-of-function phenotype. 7-Helix-1 forms a complex with the RNA-binding protein Puf2, a translational regulator of the female-specific antigen Pfs25, as well as with pfs25-coding mRNA. In accord, gametocytes deficient of 7-Helix-1 exhibit impaired Pfs25 synthesis. Our data demonstrate that 7-Helix-1 constitutes stress granules crucial for regulating the synthesis of proteins needed for life-cycle progression of Plasmodium in the mosquito vector.


Assuntos
Anopheles/parasitologia , Malária Falciparum/transmissão , Proteínas de Membrana/fisiologia , Plasmodium falciparum , Biossíntese de Proteínas , Animais , Grânulos Citoplasmáticos/metabolismo , Feminino , Humanos , Estágios do Ciclo de Vida/genética , Malária Falciparum/parasitologia , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Organismos Geneticamente Modificados , Proteínas de Ligação a Fosfato , Plasmodium falciparum/genética , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/metabolismo , Biossíntese de Proteínas/genética , Processamento de Proteína Pós-Traducional , Estrutura Secundária de Proteína , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/fisiologia , Homologia de Sequência , Estresse Fisiológico
15.
PLoS One ; 13(4): e0195823, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29672587

RESUMO

An increasing number of monoclonal antibodies and derivatives such as antibody-drug conjugates (ADC) are of the IgG1 and IgG4 isotype with distinct structural and functional properties. In cases where antibody-mediated cytotoxicity is not desired, IgG4 is often used, as its Fc region is relatively poor at inducing antibody-dependent cell-mediated or complement-dependent cytotoxicity. IgG4 ADCs with highly cytotoxic drugs against proliferating target cells but which lack or have diminished antibody effector functions against quiescent cells may have a favorable safety profile compared to IgG1. Another unique property of the IgG4 subclass is the capability to exchange half antibodies in vivo creating randomly bispecific antibodies. To investigate the functional properties of process-derived antibody species, and determine the influence of shuffling on the therapeutic efficacy, several model antibodies on the basis of the anti-CD138 antibody-drug conjugate BT062 (Indatuximab ravtansine) were generated: (I) A wild type nBT062, (II) a stable nBT062 comprising mutations to prevent half-antibody exchange, (III) a half nBT062 lacking covalent binding between two heavy chains and (IV) a stabilized, bispecific nBT062-natalizumab antibody with a second, monovalent specificity against CD49d. All nBT062 model variants were capable of CD138-specific binding and antigen-mediated internalization into cells. Furthermore, all nBT062 models inhibited tumor growth in vitro after conjugation with the maytansinoid DM4. The in vivo effects of the different molecular variants were assessed in the MAXF1322 xenograft model. The bispecific nBT062-natalizumab-DM4 demonstrated the least efficacy and was only moderately active even without the co-administration of a human IgG preparation. Wild type, stable and half nBT062-DM4 models demonstrated great anti-tumor activities. The efficacy of wild type and half nBT062-DM4 was reduced in the presence of IgG, while stable nBT062-DM4 was only marginally influenced. These pre-clinical data demonstrate the advantage of introducing half-antibody exchange-preventing mutations into therapeutic IgG4-based antibody drug-conjugates.


Assuntos
Anticorpos Biespecíficos/farmacologia , Anticorpos Monoclonais/farmacologia , Imunoconjugados/farmacologia , Imunoglobulina G/farmacologia , Animais , Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Antígenos/imunologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Variação Genética , Humanos , Imunoconjugados/imunologia , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Camundongos , Mutação , Ligação Proteica , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Int J Med Microbiol ; 308(1): 148-154, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29089241

RESUMO

Human red blood cells infected with the malaria parasite Plasmodium falciparum show an increased permeability to a number of solutes. We have previously demonstrated that such infected cells take up glutamate via a member of the excitatory amino acid transporter protein family (EAAT), namely EAAT3. Babesia divergens is a parasite that also infects human erythrocytes, and also induces increased solute permeability, including for glutamate. Here we have investigated whether glutamate uptake in B. divergens infected human red blood cells is also dependent on EAAT3 activity. We find that, although B. divergens infected cells do take up glutamate, this uptake is independent on EAAT3. Thus, though infecting the same host cell, two related parasites have developed distinct pathways to obtain access to nutrients from the extracellular milieu.


Assuntos
Babesia/fisiologia , Eritrócitos/parasitologia , Transportador 3 de Aminoácido Excitatório/metabolismo , Ácido Glutâmico/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Colina/farmacologia , Membrana Eritrocítica/efeitos dos fármacos , Membrana Eritrocítica/fisiologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/fisiologia , Transportador 3 de Aminoácido Excitatório/antagonistas & inibidores , Glutamatos/farmacologia , Nitrobenzoatos/farmacologia
17.
Int J Med Microbiol ; 308(1): 3-12, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28750796

RESUMO

The malaria parasite P. falciparum exports a large number of proteins to its host cell, the mature human erythrocyte. Although the function of the majority of these proteins is not well understood, many exported proteins appear to play a role in modification of the erythrocyte following invasion. Protein export to the erythrocyte is a secretory process that begins with entry to the endoplasmic reticulum. For most exported proteins, this step is mediated by hydrophobic signal peptides found towards the N-terminal end of proteins. The signal peptides present on P. falciparum exported proteins often differ in length from those found in other systems, and generally contain a highly extended N-terminal region. Here we have investigated the function of these extended N-terminal regions, using the exported parasite protein GBP130 as a model. Surprisingly, several deletions of the extended N-terminal regions of the GBP130 signal peptide have no effect on the ability of the signal peptide to direct a fluorescent reporter to the secretory pathway. Addition of the same N-terminal extension to a canonical signal peptide does not affect transport of either soluble or membrane proteins to their correct respective subcellular localisations. Finally, we show that extended signal peptides are able to complement canonical signal peptides in driving protein traffic to the apicoplast of the parasite, and are also functional in a mammalian cell system. Our study is the first detailed analysis of an extended P. falciparum signal peptide and suggests that N-terminal extensions of exported Plasmodium falciparum proteins are not required for entry to the secretory system, and are likely to be involved in other, so far unknown, processes.


Assuntos
Plasmodium falciparum/metabolismo , Sinais Direcionadores de Proteínas/fisiologia , Proteínas de Protozoários/metabolismo , Apicoplastos/metabolismo , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Células HEK293 , Humanos , Proteínas de Membrana/metabolismo , Mutação , Sinais Direcionadores de Proteínas/genética , Transporte Proteico , Proteínas de Protozoários/química , Via Secretória , Solubilidade
18.
Genome Biol Evol ; 9(11): 3108-3121, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29126146

RESUMO

Apicomplexans are successful parasites responsible for severe human diseases including malaria, toxoplasmosis, and cryptosporidiosis. For many years, it has been discussed whether these parasites are in possession of peroxisomes, highly variable eukaryotic organelles usually involved in fatty acid degradation and cellular detoxification. Conflicting experimental data has been published. With the age of genomics, ever more high quality apicomplexan genomes have become available, that now allow a new assessment of the dispute. Here, we provide bioinformatic evidence for the presence of peroxisomes in Toxoplasma gondii and other coccidians. For these organisms, we have identified a complete set of peroxins, probably responsible for peroxisome biogenesis, division, and protein import. Moreover, via a global screening for peroxisomal targeting signals, we were able to show that a complete set of fatty acid ß-oxidation enzymes is equipped with either PTS1 or PTS2 sequences, most likely mediating transport of these factors to putative peroxisomes in all investigated Coccidia. Our results further imply a life cycle stage-specific presence of peroxisomes in T. gondii and suggest several independent losses of peroxisomes during the evolution of apicomplexan parasites.


Assuntos
Evolução Biológica , Coccídios/citologia , Peroxissomos/genética , Toxoplasma/citologia , Coccídios/crescimento & desenvolvimento , Coccídios/metabolismo , Estágios do Ciclo de Vida , Oxirredutases , Peroxissomos/química , Sinais Direcionadores de Proteínas , Proteínas de Protozoários/química , Toxoplasma/crescimento & desenvolvimento , Toxoplasma/metabolismo
19.
Sci Rep ; 7(1): 10449, 2017 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-28874682

RESUMO

Redox balance is essential for the survival, growth and multiplication of malaria parasites and oxidative stress is involved in the mechanism of action of many antimalarial drugs. Hydrogen peroxide (H2O2) plays an important role in redox signalling and pathogen-host cell interactions. For monitoring intra- and subcellular redox events, highly sensitive and specific probes are required. Here, we stably expressed the ratiometric H2O2 redox sensor roGFP2-Orp1 in the cytosol and the mitochondria of Plasmodium falciparum (P. falciparum) NF54-attB blood-stage parasites and evaluated its sensitivity towards oxidative stress, selected antimalarial drugs, and novel lead compounds. In both compartments, the sensor showed reproducible sensitivity towards H2O2 in the low micromolar range and towards antimalarial compounds at pharmacologically relevant concentrations. Upon short-term exposure (4 h), artemisinin derivatives, quinine and mefloquine impacted H2O2 levels in mitochondria, whereas chloroquine and a glucose-6-phosphate dehydrogenase (G6PD) inhibitor affected the cytosol; 24 h exposure to arylmethylamino steroids and G6PD inhibitors revealed oxidation of mitochondria and cytosol, respectively. Genomic integration of an H2O2 sensor expressed in subcellular compartments of P. falciparum provides the basis for studying complex parasite-host cell interactions or drug effects with spatio-temporal resolution while preserving cell integrity, and sets the stage for high-throughput approaches to identify antimalarial agents perturbing redox equilibrium.


Assuntos
Peróxido de Hidrogênio/metabolismo , Malária/parasitologia , Oxirredução , Plasmodium/metabolismo , Antimaláricos/farmacologia , Técnicas Biossensoriais , Expressão Gênica , Genes Reporter , Imagem Molecular , Sondas Moleculares , Plasmodium/efeitos dos fármacos , Plasmodium/genética
20.
PLoS One ; 12(7): e0181656, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28732045

RESUMO

Malaria is caused by five different Plasmodium spp. in humans each of which modifies the host erythrocyte to survive and replicate. The two main causes of malaria, P. falciparum and P. vivax, differ in their ability to cause severe disease, mainly due to differences in the cytoadhesion of infected erythrocytes (IE) in the microvasculature. Cytoadhesion of P. falciparum in the brain leads to a large number of deaths each year and is a consequence of exported parasite proteins, some of which modify the erythrocyte cytoskeleton while others such as PfEMP1 project onto the erythrocyte surface where they bind to endothelial cells. Here we investigate the effects of knocking out an exported Hsp70-type chaperone termed Hsp70-x that is present in P. falciparum but not P. vivax. Although the growth of Δhsp70-x parasites was unaffected, the export of PfEMP1 cytoadherence proteins was delayed and Δhsp70-x IE had reduced adhesion. The Δhsp70-x IE were also more rigid than wild-type controls indicating changes in the way the parasites modified their host erythrocyte. To investigate the cause of this, transcriptional and translational changes in exported and chaperone proteins were monitored and some changes were observed. We propose that PfHsp70-x is not essential for survival in vitro, but may be required for the efficient export and functioning of some P. falciparum exported proteins.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Malária Falciparum/parasitologia , Chaperonas Moleculares/metabolismo , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/metabolismo , Virulência/fisiologia , Animais , Adesão Celular/fisiologia , Células Endoteliais/metabolismo , Células Endoteliais/parasitologia , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/parasitologia , Malária Falciparum/sangue , Malária Falciparum/metabolismo , Parasitos/metabolismo , Parasitos/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA