Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Cell ; 183(3): 636-649.e18, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-33031745

RESUMO

Cytoplasmic accumulation of TDP-43 is a disease hallmark for many cases of amyotrophic lateral sclerosis (ALS), associated with a neuroinflammatory cytokine profile related to upregulation of nuclear factor κB (NF-κB) and type I interferon (IFN) pathways. Here we show that this inflammation is driven by the cytoplasmic DNA sensor cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS) when TDP-43 invades mitochondria and releases DNA via the permeability transition pore. Pharmacologic inhibition or genetic deletion of cGAS and its downstream signaling partner STING prevents upregulation of NF-κB and type I IFN induced by TDP-43 in induced pluripotent stem cell (iPSC)-derived motor neurons and in TDP-43 mutant mice. Finally, we document elevated levels of the specific cGAS signaling metabolite cGAMP in spinal cord samples from patients, which may be a biomarker of mtDNA release and cGAS/STING activation in ALS. Our results identify mtDNA release and cGAS/STING activation as critical determinants of TDP-43-associated pathology and demonstrate the potential for targeting this pathway in ALS.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , DNA Mitocondrial/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Membrana/metabolismo , Poro de Transição de Permeabilidade Mitocondrial/metabolismo , Nucleotidiltransferases/metabolismo , Alarminas/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Citoplasma/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Inflamação/metabolismo , Interferon Tipo I/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , NF-kappa B/metabolismo , Degeneração Neural/patologia , Fosfotransferases (Aceptor do Grupo Álcool) , Subunidades Proteicas/metabolismo , Transdução de Sinais
2.
PLoS Biol ; 21(4): e3002066, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37053271

RESUMO

With emerging resistance to frontline treatments, it is vital that new antimalarial drugs are identified to target Plasmodium falciparum. We have recently described a compound, MMV020291, as a specific inhibitor of red blood cell (RBC) invasion, and have generated analogues with improved potency. Here, we generated resistance to MMV020291 and performed whole genome sequencing of 3 MMV020291-resistant populations. This revealed 3 nonsynonymous single nucleotide polymorphisms in 2 genes; 2 in profilin (N154Y, K124N) and a third one in actin-1 (M356L). Using CRISPR-Cas9, we engineered these mutations into wild-type parasites, which rendered them resistant to MMV020291. We demonstrate that MMV020291 reduces actin polymerisation that is required by the merozoite stage parasites to invade RBCs. Additionally, the series inhibits the actin-1-dependent process of apicoplast segregation, leading to a delayed death phenotype. In vitro cosedimentation experiments using recombinant P. falciparum proteins indicate that potent MMV020291 analogues disrupt the formation of filamentous actin in the presence of profilin. Altogether, this study identifies the first compound series interfering with the actin-1/profilin interaction in P. falciparum and paves the way for future antimalarial development against the highly dynamic process of actin polymerisation.


Assuntos
Antimaláricos , Malária Falciparum , Humanos , Plasmodium falciparum/metabolismo , Actinas/genética , Actinas/metabolismo , Profilinas/genética , Profilinas/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Malária Falciparum/tratamento farmacológico , Malária Falciparum/prevenção & controle , Malária Falciparum/genética , Eritrócitos/parasitologia , Antimaláricos/farmacologia
3.
PLoS Biol ; 20(5): e3001638, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35552541

RESUMO

Cryptosporidium is a leading cause of death from childhood diarrhea, but its biology is poorly understood. A recent study in PLOS Biology reveals hitherto unknown aspects of the parasite's life cycle that may lead to improvements in ex vivo culture.


Assuntos
Criptosporidiose , Cryptosporidium parvum , Cryptosporidium , Animais , Cryptosporidium/genética , Feminino , Células Germinativas , Estágios do Ciclo de Vida , Masculino
4.
Nature ; 565(7737): 118-121, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30542156

RESUMO

Plasmodium falciparum causes the severe form of malaria that has high levels of mortality in humans. Blood-stage merozoites of P. falciparum invade erythrocytes, and this requires interactions between multiple ligands from the parasite and receptors in hosts. These interactions include the binding of the Rh5-CyRPA-Ripr complex with the erythrocyte receptor basigin1,2, which is an essential step for entry into human erythrocytes. Here we show that the Rh5-CyRPA-Ripr complex binds the erythrocyte cell line JK-1 significantly better than does Rh5 alone, and that this binding occurs through the insertion of Rh5 and Ripr into host membranes as a complex with high molecular weight. We report a cryo-electron microscopy structure of the Rh5-CyRPA-Ripr complex at subnanometre resolution, which reveals the organization of this essential invasion complex and the mode of interactions between members of the complex, and shows that CyRPA is a critical mediator of complex assembly. Our structure identifies blades 4-6 of the ß-propeller of CyRPA as contact sites for Rh5 and Ripr. The limited contacts between Rh5-CyRPA and CyRPA-Ripr are consistent with the dissociation of Rh5 and Ripr from CyRPA for membrane insertion. A comparision of the crystal structure of Rh5-basigin with the cryo-electron microscopy structure of Rh5-CyRPA-Ripr suggests that Rh5 and Ripr are positioned parallel to the erythrocyte membrane before membrane insertion. This provides information on the function of this complex, and thereby provides insights into invasion by P. falciparum.


Assuntos
Antígenos de Protozoários/ultraestrutura , Proteínas de Transporte/ultraestrutura , Microscopia Crioeletrônica , Complexos Multiproteicos/química , Complexos Multiproteicos/ultraestrutura , Plasmodium falciparum , Proteínas de Protozoários/ultraestrutura , Animais , Antígenos de Protozoários/química , Antígenos de Protozoários/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Drosophila , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/parasitologia , Humanos , Modelos Moleculares , Complexos Multiproteicos/metabolismo , Plasmodium falciparum/química , Plasmodium falciparum/patogenicidade , Plasmodium falciparum/ultraestrutura , Ligação Proteica , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo
5.
J Biol Chem ; 299(3): 103006, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36775128

RESUMO

Cryptosporidium parvum is a zoonotic apicomplexan parasite and a common cause of diarrheal disease worldwide. The development of vaccines to prevent or limit infection remains an important goal for tackling cryptosporidiosis. At present, the only approved vaccine against any apicomplexan parasite targets a conserved adhesin possessing a thrombospondin repeat domain. C. parvum possesses 12 orthologous thrombospondin repeat domain-containing proteins known as CpTSP1-12, though little is known about these potentially important antigens. Here, we explore the architecture and conservation of the CpTSP protein family, as well as their abundance at the protein level within the sporozoite stage of the life cycle. We examine the glycosylation states of these proteins using a combination of glycopeptide enrichment techniques to demonstrate that these proteins are modified with C-, O-, and N-linked glycans. Using expansion microscopy, and an antibody against the C-linked mannose that is unique to the CpTSP protein family within C. parvum, we show that these proteins are found both on the cell surface and in structures that resemble the secretory pathway of C. parvum sporozoites. Finally, we generated a polyclonal antibody against CpTSP1 to show that it is found at the cell surface and within micronemes, in a pattern reminiscent of other apicomplexan motility-associated adhesins, and is present both in sporozoites and meronts. This work sheds new light on an understudied family of C. parvum proteins that are likely to be important to both parasite biology and the development of vaccines against cryptosporidiosis.


Assuntos
Criptosporidiose , Cryptosporidium parvum , Cryptosporidium , Animais , Humanos , Cryptosporidium parvum/metabolismo , Criptosporidiose/parasitologia , Criptosporidiose/prevenção & controle , Glicosilação , Cryptosporidium/metabolismo , Proteínas de Protozoários/química , Esporozoítos , Trombospondinas/metabolismo
6.
J Neuroinflammation ; 21(1): 14, 2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38195485

RESUMO

Traumatic brain injury (TBI) is a key contributor to global morbidity that lacks effective treatments. Microbial infections are common in TBI patients, and their presence could modify the physiological response to TBI. It is estimated that one-third of the human population is incurably infected with the feline-borne parasite, Toxoplasma gondii, which can invade the central nervous system and result in chronic low-grade neuroinflammation, oxidative stress, and excitotoxicity-all of which are also important pathophysiological processes in TBI. Considering the large number of TBI patients that have a pre-existing T. gondii infection prior to injury, and the potential mechanistic synergies between the conditions, this study investigated how a pre-existing T. gondii infection modified TBI outcomes across acute, sub-acute and chronic recovery in male and female mice. Gene expression analysis of brain tissue found that neuroinflammation and immune cell markers were amplified in the combined T. gondii + TBI setting in both males and females as early as 2-h post-injury. Glutamatergic, neurotoxic, and oxidative stress markers were altered in a sex-specific manner in T. gondii + TBI mice. Structural MRI found that male, but not female, T. gondii + TBI mice had a significantly larger lesion size compared to their uninfected counterparts at 18-weeks post-injury. Similarly, diffusion MRI revealed that T. gondii + TBI mice had exacerbated white matter tract abnormalities, particularly in male mice. These novel findings indicate that a pre-existing T. gondii infection affects the pathophysiological aftermath of TBI in a sex-dependent manner, and may be an important modifier to consider in the care and prognostication of TBI patients.


Assuntos
Lesões Encefálicas Traumáticas , Lesões Encefálicas , Toxoplasmose , Humanos , Animais , Gatos , Feminino , Masculino , Camundongos , Doenças Neuroinflamatórias , Lesões Encefálicas/complicações , Lesões Encefálicas Traumáticas/complicações , Toxoplasmose/complicações , Encéfalo
7.
J Cell Sci ; 134(20)2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34523684

RESUMO

The voltage-dependent anion channel (VDAC) is a ubiquitous channel in the outer membrane of the mitochondrion with multiple roles in protein, metabolite and small molecule transport. In mammalian cells, VDAC protein, as part of a larger complex including the inositol triphosphate receptor, has been shown to have a role in mediating contacts between the mitochondria and endoplasmic reticulum (ER). We identify VDAC of the pathogenic apicomplexan Toxoplasma gondii and demonstrate its importance for parasite growth. We show that VDAC is involved in protein import and metabolite transfer to mitochondria. Further, depletion of VDAC resulted in significant morphological changes in the mitochondrion and ER, suggesting a role in mediating contacts between these organelles in T. gondii. This article has an associated First Person interview with the first author of the paper.


Assuntos
Toxoplasma , Animais , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Humanos , Mitocôndrias/metabolismo , Transporte Proteico , Toxoplasma/genética , Toxoplasma/metabolismo , Canais de Ânion Dependentes de Voltagem/genética , Canais de Ânion Dependentes de Voltagem/metabolismo
8.
Mol Microbiol ; 115(5): 916-929, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33278047

RESUMO

Toxoplasma and other apicomplexan parasites undergo a unique form of cellular locomotion referred to as "gliding motility." Gliding motility is crucial for parasite survival as it powers tissue dissemination, host cell invasion and egress. Distinct environmental cues lead to activation of gliding motility and have become a prominent focus of recent investigation. Progress has been made toward understanding what environmental cues are sensed and how these signals are transduced in order to regulate the machinery and cellular events powering gliding motility. In this review, we will discuss new findings and integrate these into our current understanding to propose a model of how environmental sensing is achieved to regulate gliding motility in Toxoplasma. Collectively, these findings also have implications for the understanding of gliding motility across Apicomplexa more broadly.


Assuntos
Toxoplasma/citologia , Toxoplasma/metabolismo , Toxoplasmose/parasitologia , Animais , Movimento Celular , Ecossistema , Humanos , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Toxoplasma/genética
9.
PLoS Biol ; 16(9): e2005642, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30208022

RESUMO

The phylum Apicomplexa comprises a group of obligate intracellular parasites that alternate between intracellular replicating stages and actively motile extracellular forms that move through tissue. Parasite cytosolic Ca2+ signalling activates motility, but how this is switched off after invasion is complete to allow for replication to begin is not understood. Here, we show that the cyclic adenosine monophosphate (cAMP)-dependent protein kinase A catalytic subunit 1 (PKAc1) of Toxoplasma is responsible for suppression of Ca2+ signalling upon host cell invasion. We demonstrate that PKAc1 is sequestered to the parasite periphery by dual acylation of PKA regulatory subunit 1 (PKAr1). Upon genetic depletion of PKAc1 we show that newly invaded parasites exit host cells shortly thereafter, in a perforin-like protein 1 (PLP-1)-dependent fashion. Furthermore, we demonstrate that loss of PKAc1 prevents rapid down-regulation of cytosolic [Ca2+] levels shortly after invasion. We also provide evidence that loss of PKAc1 sensitises parasites to cyclic GMP (cGMP)-induced Ca2+ signalling, thus demonstrating a functional link between cAMP and these other signalling modalities. Together, this work provides a new paradigm in understanding how Toxoplasma and related apicomplexan parasites regulate infectivity.


Assuntos
Sinalização do Cálcio , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Toxoplasma/enzimologia , Acilação , Animais , Cálcio/metabolismo , AMP Cíclico/metabolismo , Citosol/metabolismo , Fibroblastos/parasitologia , Interações Hospedeiro-Parasita , Humanos , Estágios do Ciclo de Vida , Camundongos , Parasitos/enzimologia , Parasitos/crescimento & desenvolvimento , Subunidades Proteicas/metabolismo , Proteínas de Protozoários , Transdução de Sinais , Toxoplasma/crescimento & desenvolvimento
10.
J Biol Chem ; 294(5): 1541-1553, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30514763

RESUMO

Toxoplasma gondii is a ubiquitous, obligate intracellular eukaryotic parasite that causes congenital birth defects, disease in immunocompromised individuals, and blindness. Protein glycosylation plays an important role in the infectivity and evasion of immune responses of many eukaryotic parasites and is also of great relevance to vaccine design. Here we demonstrate that micronemal protein 2 (MIC2), a motility-associated adhesin of T. gondii, has highly glycosylated thrombospondin repeat (TSR) domains. Using affinity-purified MIC2 and MS/MS analysis along with enzymatic digestion assays, we observed that at least seven C-linked and three O-linked glycosylation sites exist within MIC2, with >95% occupancy at these O-glycosylation sites. We found that addition of O-glycans to MIC2 is mediated by a protein O-fucosyltransferase 2 homolog (TgPOFUT2) encoded by the TGGT1_273550 gene. Even though POFUT2 homologs are important for stabilizing motility-associated adhesins and for host infection in other apicomplexan parasites, loss of TgPOFUT2 in T. gondii had only a modest impact on MIC2 levels and the wider parasite proteome. Consistent with this, both plaque formation and tachyzoite invasion were broadly similar in the presence or absence of TgPOFUT2. These findings indicate that TgPOFUT2 O-glycosylates MIC2 and that this glycan, in contrast to previous findings in another study, is dispensable in T. gondii tachyzoites and for T. gondii infectivity.


Assuntos
Fibroblastos/parasitologia , Fucosiltransferases/metabolismo , Interações Hospedeiro-Parasita , Proteínas de Membrana/metabolismo , Proteínas de Protozoários/metabolismo , Toxoplasma/patogenicidade , Toxoplasmose/parasitologia , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Glicosilação , Humanos , Proteoma/análise , Toxoplasmose/metabolismo
11.
J Biol Chem ; 294(22): 8959-8972, 2019 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-30992368

RESUMO

Protozoan parasites of the phylum Apicomplexa actively move through tissue to initiate and perpetuate infection. The regulation of parasite motility relies on cyclic nucleotide-dependent kinases, but how these kinases are activated remains unknown. Here, using an array of biochemical and cell biology approaches, we show that the apicomplexan parasite Toxoplasma gondii expresses a large guanylate cyclase (TgGC) protein, which contains several upstream ATPase transporter-like domains. We show that TgGC has a dynamic localization, being concentrated at the apical tip in extracellular parasites, which then relocates to a more cytosolic distribution during intracellular replication. Conditional TgGC knockdown revealed that this protein is essential for acute-stage tachyzoite growth, as TgGC-deficient parasites were defective in motility, host cell attachment, invasion, and subsequent host cell egress. We show that TgGC is critical for a rapid rise in cytosolic [Ca2+] and for secretion of microneme organelles upon stimulation with a cGMP agonist, but these deficiencies can be bypassed by direct activation of signaling by a Ca2+ ionophore. Furthermore, we found that TgGC is required for transducing changes in extracellular pH and [K+] to activate cytosolic [Ca2+] flux. Together, the results of our work implicate TgGC as a putative signal transducer that activates Ca2+ signaling and motility in Toxoplasma.


Assuntos
Adenosina Trifosfatases/metabolismo , Sinalização do Cálcio , Guanilato Ciclase/metabolismo , Proteínas de Protozoários/metabolismo , Toxoplasma/metabolismo , Adenosina Trifosfatases/genética , Cálcio/metabolismo , Ionóforos de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , GMP Cíclico/metabolismo , Citosol/metabolismo , Guanilato Ciclase/antagonistas & inibidores , Guanilato Ciclase/genética , Concentração de Íons de Hidrogênio , Oligonucleotídeos Antissenso/metabolismo , Potássio/metabolismo , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/genética , Pirazóis/farmacologia , Pirimidinonas/farmacologia , Toxoplasma/crescimento & desenvolvimento
12.
J Biol Chem ; 294(14): 5720-5734, 2019 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-30723156

RESUMO

The Plasmodium falciparum ATPase PfATP4 is the target of a diverse range of antimalarial compounds, including the clinical drug candidate cipargamin. PfATP4 was originally annotated as a Ca2+ transporter, but recent evidence suggests that it is a Na+ efflux pump, extruding Na+ in exchange for H+ Here we demonstrate that ATP4 proteins belong to a clade of P-type ATPases that are restricted to apicomplexans and their closest relatives. We employed a variety of genetic and physiological approaches to investigate the ATP4 protein of the apicomplexan Toxoplasma gondii, TgATP4. We show that TgATP4 is a plasma membrane protein. Knockdown of TgATP4 had no effect on resting pH or Ca2+ but rendered parasites unable to regulate their cytosolic Na+ concentration ([Na+]cyt). PfATP4 inhibitors caused an increase in [Na+]cyt and a cytosolic alkalinization in WT but not TgATP4 knockdown parasites. Parasites in which TgATP4 was knocked down or disrupted exhibited a growth defect, attributable to reduced viability of extracellular parasites. Parasites in which TgATP4 had been disrupted showed reduced virulence in mice. These results provide evidence for ATP4 proteins playing a key conserved role in Na+ regulation in apicomplexan parasites.


Assuntos
Membrana Celular/enzimologia , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Proteínas de Protozoários/metabolismo , Toxoplasma/enzimologia , Animais , Membrana Celular/genética , Citoplasma/genética , Citoplasma/metabolismo , Feminino , Técnicas de Silenciamento de Genes , ATPase Trocadora de Hidrogênio-Potássio/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Protozoários/genética , Sódio/metabolismo , Toxoplasma/genética , Toxoplasma/patogenicidade
13.
J Neuroinflammation ; 17(1): 222, 2020 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-32711529

RESUMO

Traumatic brain injury (TBI) is one of the leading causes of morbidity and mortality worldwide; however, treatment development is hindered by the heterogenous nature of TBI presentation and pathophysiology. In particular, the degree of neuroinflammation after TBI varies between individuals and may be modified by other factors such as infection. Toxoplasma gondii, a parasite that infects approximately one-third of the world's population, has a tropism for brain tissue and can persist as a life-long infection. Importantly, there is notable overlap in the pathophysiology between TBI and T. gondii infection, including neuroinflammation. This paper will review current understandings of the clinical problems, pathophysiological mechanisms, and functional outcomes of TBI and T. gondii, before considering the potential synergy between the two conditions. In particular, the discussion will focus on neuroinflammatory processes such as microglial activation, inflammatory cytokines, and peripheral immune cell recruitment that occur during T. gondii infection and after TBI. We will present the notion that these overlapping pathologies in TBI individuals with a chronic T. gondii infection have the strong potential to exacerbate neuroinflammation and related brain damage, leading to amplified functional deficits. The impact of chronic T. gondii infection on TBI should therefore be investigated in both preclinical and clinical studies as the possible interplay could influence treatment strategies.


Assuntos
Lesões Encefálicas Traumáticas/microbiologia , Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/fisiopatologia , Toxoplasmose/complicações , Toxoplasmose/patologia , Animais , Encéfalo/microbiologia , Encéfalo/patologia , Gatos , Humanos , Inflamação , Toxoplasma
14.
Mol Microbiol ; 109(4): 458-473, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29873127

RESUMO

Transmission of the malaria parasite Plasmodium falciparum involves infection of Anopheles mosquitoes. Here we characterize SOPT, a protein expressed in P. falciparum ookinetes that facilitates infection of the mosquito midgut. SOPT was identified on the basis that it contains a signal peptide, a PEXEL-like sequence and is expressed in asexual, ookinete and sporozoite stages, suggesting it is involved in infecting the human or mosquito host. SOPT is predicted to contain a subtilisin-like fold with a non-canonical catalytic triad and is orthologous to P. berghei PIMMS2. Localization studies reveal that SOPT is not exported to the erythrocyte but is expressed in ookinetes at the parasite periphery. SOPT-deficient parasites develop normally through the asexual and sexual stages and produce equivalent numbers of ookinetes to NF54 controls, however, they form fewer oocysts and sporozoites in mosquitoes. SOPT-deficient parasites were also unable to activate the immune-responsive midgut invasion marker SRPN6 after mosquito ingestion, suggesting they are defective for entry into the midgut. Disruption of SOPT in P. berghei (PIMMS2) did not affect other lifecycle stages or ookinete development but again resulted in fewer oocysts and sporozoites in mosquitoes. Collectively, this study shows that SOPT/PIMMS2 plays a conserved role in ookinetes of different Plasmodium species.


Assuntos
Anopheles/parasitologia , Sistema Digestório/parasitologia , Oocistos/crescimento & desenvolvimento , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/metabolismo , Esporozoítos/crescimento & desenvolvimento , Animais , Malária Falciparum/transmissão , Mosquitos Vetores/parasitologia , Subtilisina/metabolismo
15.
Artigo em Inglês | MEDLINE | ID: mdl-30559138

RESUMO

A series of 4-amino 2-anilinoquinazolines optimized for activity against the most lethal malaria parasite of humans, Plasmodium falciparum, was evaluated for activity against other human Plasmodium parasites and related apicomplexans that infect humans and animals. Four of the most promising compounds from the 4-amino 2-anilinoquinazoline series were equally as effective against the asexual blood stages of the zoonotic P. knowlesi, suggesting that they could also be effective against the closely related P. vivax, another important human pathogen. The 2-anilinoquinazoline compounds were also potent against an array of P. falciparum parasites resistant to clinically available antimalarial compounds, although slightly less so than against the drug-sensitive 3D7 parasite line. The apicomplexan parasites Toxoplasma gondii, Babesia bovis, and Cryptosporidium parvum were less sensitive to the 2-anilinoquinazoline series with a 50% effective concentration generally in the low micromolar range, suggesting that the yet to be discovered target of these compounds is absent or highly divergent in non-Plasmodium parasites. The 2-anilinoquinazoline compounds act as rapidly as chloroquine in vitro and when tested in rodents displayed a half-life that contributed to the compound's capacity to clear P. falciparum blood stages in a humanized mouse model. At a dose of 50 mg/kg of body weight, adverse effects to the humanized mice were noted, and evaluation against a panel of experimental high-risk off targets indicated some potential off-target activity. Further optimization of the 2-anilinoquinazoline antimalarial class will concentrate on improving in vivo efficacy and addressing adverse risk.


Assuntos
Compostos de Anilina/farmacologia , Antiparasitários/farmacologia , Babesia bovis/efeitos dos fármacos , Cryptosporidium parvum/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Quinazolinas/farmacologia , Toxoplasma/efeitos dos fármacos , Animais , Antimaláricos/farmacologia , Linhagem Celular , Cloroquina/farmacologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Testes de Sensibilidade Parasitária , Ratos , Ratos Sprague-Dawley
16.
Brain Behav Immun ; 80: 88-108, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30807837

RESUMO

Toxoplasma gondii (T. gondii) is a neurotropic parasite that is associated with various neuropsychiatric disorders. Rodents infected with T. gondii display a plethora of behavioural alterations, and Toxoplasma infection in humans has been strongly associated with disorders such as schizophrenia, in which impaired social behaviour is an important feature. Elucidating changes at the cellular level relevant to neuropsychiatric conditions can lead to effective therapies. Here, we compare changes in behaviour during an acute and chronic T. gondii infection in female mice. Further, we notice that during chronic phase of infection, mice display impaired sociability when exposed to a novel conspecific. Also, we show that T. gondii infected mice display impaired short-term social recognition memory. However, object recognition memory remains intact. Using c-Fos as a marker of neuronal activity, we show that infection leads to an impairment in neuronal activation in the medial prefrontal cortex, hippocampus as well as the amygdala when mice are exposed to a social environment and a change in functional connectivity between these regions. We found changes in synaptic proteins that play a role in the process of neuronal activation such as synaptophysin, PSD-95 and changes in downstream substrates of cell activity such as cyclic AMP, phospho-CREB and BDNF. Our results point towards an imbalance in neuronal activity that can lead to a wider range of neuropsychiatric problems upon T. gondii infection.


Assuntos
Cognição/fisiologia , Neurônios/metabolismo , Toxoplasmose/psicologia , Tonsila do Cerebelo/metabolismo , Animais , Comportamento Animal/fisiologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Feminino , Hipocampo/metabolismo , Memória de Curto Prazo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Córtex Pré-Frontal/metabolismo , Comportamento Social , Sulfadiazina/farmacologia , Toxoplasma/metabolismo , Toxoplasma/patogenicidade , Toxoplasmose/metabolismo , Toxoplasmose Animal/psicologia
17.
J Immunol ; 199(12): 4165-4179, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29084838

RESUMO

We describe an MHC class II (I-Ab)-restricted TCR transgenic mouse line that produces CD4+ T cells specific for Plasmodium species. This line, termed PbT-II, was derived from a CD4+ T cell hybridoma generated to blood-stage Plasmodium berghei ANKA (PbA). PbT-II cells responded to all Plasmodium species and stages tested so far, including rodent (PbA, P. berghei NK65, Plasmodium chabaudi AS, and Plasmodium yoelii 17XNL) and human (Plasmodium falciparum) blood-stage parasites as well as irradiated PbA sporozoites. PbT-II cells can provide help for generation of Ab to P. chabaudi infection and can control this otherwise lethal infection in CD40L-deficient mice. PbT-II cells can also provide help for development of CD8+ T cell-mediated experimental cerebral malaria (ECM) during PbA infection. Using PbT-II CD4+ T cells and the previously described PbT-I CD8+ T cells, we determined the dendritic cell (DC) subsets responsible for immunity to PbA blood-stage infection. CD8+ DC (a subset of XCR1+ DC) were the major APC responsible for activation of both T cell subsets, although other DC also contributed to CD4+ T cell responses. Depletion of CD8+ DC at the beginning of infection prevented ECM development and impaired both Th1 and follicular Th cell responses; in contrast, late depletion did not affect ECM. This study describes a novel and versatile tool for examining CD4+ T cell immunity during malaria and provides evidence that CD4+ T cell help, acting via CD40L signaling, can promote immunity or pathology to blood-stage malaria largely through Ag presentation by CD8+ DC.


Assuntos
Apresentação de Antígeno , Linfócitos T CD4-Positivos/imunologia , Antígenos CD40/imunologia , Células Dendríticas/imunologia , Malária/imunologia , Camundongos Transgênicos/imunologia , Parasitemia/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Antígenos de Protozoários/imunologia , Antígenos CD40/deficiência , Ligante de CD40/imunologia , Células Cultivadas , Cruzamentos Genéticos , Hibridomas , Ativação Linfocitária , Malária Cerebral/imunologia , Malária Cerebral/prevenção & controle , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/genética , Plasmodium berghei/imunologia , Quimera por Radiação
18.
Traffic ; 17(2): 102-16, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26566590

RESUMO

Upon infection, apicomplexan parasites quickly invade host cells and begin a replicative cycle rapidly increasing in number over a short period of time, leading to tissue lysis and disease. The secretory pathway of these highly polarized protozoan parasites tightly controls, in time and space, the biogenesis of specialized structures and organelles required for invasion and intracellular survival. In other systems, regulation of protein trafficking can occur by phosphorylation of vesicle fusion machinery. Previously, we have shown that Toxoplasma gondii αSNAP - a protein that controls the disassembly of cis-SNARE complexes--is phosphorylated. Here, we show that this post-translational modification is required for the correct function of αSNAP in controlling secretory traffic. We demonstrate that during intracellular development conditional expression of a non-phosphorylatable form of αSNAP results in Golgi fragmentation and vesiculation of all downstream secretory organelles. In addition, we show that the vestigial plastid (termed apicoplast), although reported not to be reliant on Golgi trafficking for biogenesis, is also affected upon overexpression of αSNAP and is much more sensitive to the levels of this protein than targeting to other organelles. This work highlights the importance of αSNAP and its phosphorylation in Toxoplasma organelle biogenesis and exposes a hereto fore-unexplored mechanism of regulation of vesicle fusion during secretory pathway trafficking in apicomplexan parasites.


Assuntos
Organelas/metabolismo , Fosforilação/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Via Secretória/fisiologia , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida/metabolismo , Toxoplasma/metabolismo , Complexo de Golgi/fisiologia , Biogênese de Organelas , Organelas/fisiologia , Transporte Proteico/fisiologia , Proteínas de Protozoários/metabolismo , Toxoplasma/fisiologia
19.
J Biol Chem ; 292(18): 7662-7674, 2017 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-28258212

RESUMO

Toxoplasma gondii, like all apicomplexan parasites, uses Ca2+ signaling pathways to activate gliding motility to power tissue dissemination and host cell invasion and egress. A group of "plant-like" Ca2+-dependent protein kinases (CDPKs) transduces cytosolic Ca2+ flux into enzymatic activity, but how they function is poorly understood. To investigate how Ca2+ signaling activates egress through CDPKs, we performed a forward genetic screen to isolate gain-of-function mutants from an egress-deficient cdpk3 knockout strain. We recovered mutants that regained the ability to egress from host cells that harbored mutations in the gene Suppressor of Ca2+-dependent Egress 1 (SCE1). Global phosphoproteomic analysis showed that SCE1 deletion restored many Δcdpk3-dependent phosphorylation events to near wild-type levels. We also show that CDPK3-dependent SCE1 phosphorylation is required to relieve its suppressive activity to potentiate egress. In summary, our work has uncovered a novel component and suppressor of Ca2+-dependent cell egress during Toxoplasma lytic growth.


Assuntos
Sinalização do Cálcio/fisiologia , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas Quinases/metabolismo , Proteínas de Protozoários/metabolismo , Toxoplasma/metabolismo , Proteínas de Ligação ao Cálcio/genética , Fosforilação/fisiologia , Proteínas Quinases/genética , Proteínas de Protozoários/genética , Toxoplasma/genética
20.
Cell Microbiol ; 19(4)2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27781359

RESUMO

Host cell invasion, exit and parasite dissemination is critical to the pathogenesis of apicomplexan parasites such as Toxoplasma gondii and Plasmodium spp. These processes are regulated by intracellular Ca2+ signaling although the temporal dynamics of Ca2+ fluxes and down-stream second messenger pathways are poorly understood. Here, we use a genetically encoded biosensor, GFP-Calmodulin-M13-6 (GCaMP6), to capture Ca2+ flux in live Toxoplasma and investigate the role of Ca2+ signaling in egress and motility. Our analysis determines how environmental cues and signal activation influence intracellular Ca2+ flux, allowing placement of effector molecules within this pathway. Importantly, we have identified key interrelationships between cGMP and Ca2+ signaling that are required for activation of egress and motility. Furthermore, we extend this analysis to show that the Ca2+ Dependent Protein Kinases-TgCDPK1 and TgCDPK3-play a role in signal quenching before egress. This work highlights the interrelationships of second messenger pathways of Toxoplasma in space and time, which is likely required for pathogenesis of all apicomplexan species.


Assuntos
Sinalização do Cálcio , Toxoplasma/patogenicidade , Toxoplasmose/parasitologia , Células Cultivadas , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Interações Hospedeiro-Parasita , Humanos , Toxoplasma/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA