Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Trends Parasitol ; 39(11): 900-901, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37743113

ABSTRACT

The capacity to infect and survive in a wide variety of host cells is amongst the strategies that contribute to pathogen persistence. The recent study by Karagiannis et al. presents an unbiased approach to identify infected cells in a visceral leishmaniasis (VL) infection, which revealed parasites in unexpected host cells.

2.
Parasit Vectors ; 16(1): 152, 2023 Apr 28.
Article in English | MEDLINE | ID: mdl-37106356

ABSTRACT

BACKGROUND: Immune complexing of target antigen to high affinity host antibody is recognized to impact the sensitivity of commercial heartworm antigen tests. Published information describing the effect of heat on interfering canine host antibodies is lacking. Immune complex dissociation (ICD) by heat treatment of serum for samples initially testing negative for heartworm antigen increases sensitivity of commercial antigen tests, particularly for single sex or low adult infection intensities. In this study the stability and nature of the targeted epitope and mechanism of heat ICD were examined. METHODS: Canine IgG was isolated using protein-A columns from serum originating from four dogs evaluated after necropsy: one dog with evidence of previously cleared infection and three dogs with confirmed heartworm infections. These dogs were expected to have an excess of antibodies based on negative antigen test and to have no or low antigen optical density, respectively, following heat treatment. Interference of antigen detection on (non-heated) positive serum was evaluated, following 1:1 mixing of antibody/PBS solutions previously heated at 25 °C, 65 °C, 75 °C, 85 °C, 95 °C and 104 °C, compared to positive serum/PBS control measured by optical density using a commercial heartworm antigen ELISA and protein quantification. Live heartworms incubated in media for 72 h provided excretory/secretory antigen for antigen stability studies following heat, endopeptidase digestion and disulfide bond reduction. RESULTS: Mixing antigen-positive heartworm serum with antibody solutions demonstrated a significant inhibition of antigen detection for antibody solutions previously heated at 25 °C and 65 °C relative to positive serum/PBS control. Antigen detection optical density was restored at or above the control when positive serum was mixed with solutions previously heated at 75 °C, 85 °C, 95 °C and 104 °C. Significant changes occurred in protein levels for antibody solutions heated at 75 °C, 85 °C, 95 °C and 104 °C. Relative stability of antigen from live heartworms in culture was demonstrated following heat, chemical and enzymatic treatment. CONCLUSIONS: Significant changes in protein levels and antigen binding ability occurred in IgG solutions heated above 65 °C. The findings confirm heat denaturation of antibodies as the suspected mechanism of heat ICD at 104 °C for antigen diagnosis of heartworm. No significant change occurred in antigen detection following heat, chemical or enzymatic digestions supporting a heat-stable linear nature of the epitope.


Subject(s)
Dirofilaria immitis , Dirofilariasis , Dog Diseases , Dogs , Animals , Temperature , Antigens, Helminth , Antigen-Antibody Complex , Fever , Epitopes , Immunoglobulin G
3.
Article in English | MEDLINE | ID: mdl-35601996

ABSTRACT

Extracellular vesicles (EVs) are membrane-enclosed packets released from cells that can transfer bioactive molecules from cell to cell without direct contact with the target cells. This transfer of molecules can activate consequential processes in the recipient cells, including cell differentiation and migration that maintain tissue homeostasis or promote tissue pathology. One controversial aspect of the EV's biology that holds therapeutic promise is their capacity to engage defined cells at specific sites. On the one hand, persuasive studies have shown that EVs express surface molecules that ensure their tissue localization and enable cell-specific interactions, as demonstrated using in vitro and in vivo analyses. Therefore, this feature of EV biology is under investigation in translational studies to control malignancies and deliver chemicals and bioactive molecules to combat several diseases. On the other hand, some studies have shown that EVs fail to traffic in hosts in a targeted manner, which questions the potential role of EVs as vehicles for drug delivery and their capacity to serve as cell-free biomodulators. In this review, the biology of EV homing/tropism in mammalian hosts is discussed, and the biological characteristics that may result in their controversial characteristics are brought to the fore.

4.
Front Cell Infect Microbiol ; 11: 702676, 2021.
Article in English | MEDLINE | ID: mdl-34490142

ABSTRACT

Clinical manifestations of leishmaniasis range from self-healing, cutaneous lesions to fatal infections of the viscera. With no preventative Leishmania vaccine available, the frontline option against leishmaniasis is chemotherapy. Unfortunately, currently available anti-Leishmania drugs face several obstacles, including toxicity that limits dosing and emergent drug resistant strains in endemic regions. It is, therefore, imperative that more effective drug formulations with decreased toxicity profiles are developed. Previous studies had shown that 2-(((5-Methyl-2-thienyl)methylene)amino)-N-phenylbenzamide (also called Retro-2) has efficacy against Leishmania infections. Structure-activity relationship (SAR) analogs of Retro-2, using the dihydroquinazolinone (DHQZ) base structure, were subsequently described that are more efficacious than Retro-2. However, considering the hydrophobic nature of these compounds that limits their solubility and uptake, the current studies were initiated to determine whether the solubility of Retro-2 and its SAR analogs could be enhanced through encapsulation in amphiphilic polymer nanoparticles. We evaluated encapsulation of these compounds in the amphiphilic, thermoresponsive oligo(ethylene glycol) methacrylate-co-pentafluorostyrene (PFG30) copolymer that forms nanoparticle aggregates upon heating past temperatures of 30°C. The hydrophobic tracer, coumarin 6, was used to evaluate uptake of a hydrophobic molecule into PFG30 aggregates. Mass spectrometry analysis showed considerably greater delivery of encapsulated DHQZ analogs into infected cells and more rapid shrinkage of L. amazonensis communal vacuoles. Moreover, encapsulation in PFG30 augmented the efficacy of Retro-2 and its SAR analogs to clear both L. amazonensis and L. donovani infections. These studies demonstrate that encapsulation of compounds in PFG30 is a viable approach to dramatically increase bioavailability and efficacy of anti-Leishmania compounds.


Subject(s)
Leishmania , Leishmaniasis , Animals , Biological Availability , Leishmaniasis/drug therapy , Mice , Mice, Inbred BALB C , Polymers
5.
Front Immunol ; 12: 662944, 2021.
Article in English | MEDLINE | ID: mdl-33959131

ABSTRACT

Extracellular vesicles (EVs) have garnered significant interest in recent years due to their contributions to cell-to-cell communication and disease processes. EVs are composed of a complex profile of bioactive molecules, which include lipids, nucleic acids, metabolites, and proteins. Although the biogenesis of EVs released by cells under various normal and abnormal conditions has been well-studied, there is incomplete knowledge about how infection influences EV biogenesis. EVs from infected cells contain specific molecules of both host and pathogen origin that may contribute to pathogenesis and the elicitation of the host immune response. Intracellular pathogens exhibit diverse lifestyles that undoubtedly dictate the mechanisms by which their molecules enter the cell's exosome biogenesis schemes. We will discuss the current understanding of the mechanisms used during infection to traffic molecules from their vacuolar niche to host EVs by selected intravacuolar pathogens. We initially review general exosome biogenesis schemes and then discuss what is known about EV biogenesis in Mycobacterium, Plasmodium, Toxoplasma, and Leishmania infections, which are pathogens that reside within membrane delimited compartments in phagocytes at some time in their life cycle within mammalian hosts. The review includes discussion of the need for further studies into the biogenesis of EVs to better understand the contributions of these vesicles to host-pathogen interactions, and to uncover potential therapeutic targets to control these pathogens.


Subject(s)
Extracellular Vesicles/metabolism , Host-Pathogen Interactions/immunology , Virulence Factors/metabolism , Animals , Biological Transport , Cell Communication , Exosomes , Extracellular Vesicles/microbiology , Extracellular Vesicles/parasitology , Gene Expression Regulation , Host-Parasite Interactions , Host-Pathogen Interactions/genetics , Humans , Intracellular Space/immunology , Intracellular Space/metabolism , Intracellular Space/microbiology , Intracellular Space/parasitology , Protein Processing, Post-Translational , Signal Transduction
6.
Life Sci Alliance ; 3(12)2020 12.
Article in English | MEDLINE | ID: mdl-33122174

ABSTRACT

Leishmania donovani infection of macrophages results in quantitative and qualitative changes in the protein profile of extracellular vesicles (EVs) released by the infected host cells. We confirmed mass spectrometry results orthogonally by performing Western blots for several Leishmania-infected macrophage-enriched EVs (LieEVs) molecules. Several host cell proteins in LieEVs have been implicated in promoting vascular changes in other systems. We also identified 59 parasite-derived proteins in LieEVs, including a putative L. donovani homolog of mammalian vasohibins (LdVash), which in mammals promotes angiogenesis. We developed a transgenic parasite that expressed an endogenously tagged LdVash/mNeonGreen (mNG) and confirmed that LdVash/mNG is indeed expressed in infected macrophages and in LieEVs. We further observed that LieEVs induce endothelial cells to release angiogenesis promoting mediators including IL-8, G-CSF/CSF-3, and VEGF-A. In addition, LieEVs induce epithelial cell migration and tube formation by endothelial cells in surrogate angiogenesis assays. Taken together, these studies show that Leishmania infection alters the composition of EVs from infected cells and suggest that LieEVs may play a role in the promotion of vascularization of Leishmania infections.


Subject(s)
Extracellular Vesicles/physiology , Leishmaniasis/immunology , Macrophages/metabolism , Animals , Apoptosis/physiology , Cell Line , Cell Movement/physiology , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Leishmania donovani/metabolism , Leishmania donovani/pathogenicity , Leishmaniasis/metabolism , Macrophages/immunology , Mice , Parasites , Proteomics/methods , RAW 264.7 Cells
7.
Eur J Pharm Sci ; 150: 105335, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32272211

ABSTRACT

Leishmaniasis are a group of neglected infectious diseases caused by protozoa of the genus Leishmania with distinct presentations. The available leishmaniasis treatment options are either expensive and/or; cause adverse effects and some are ineffective for resistant Leishmania strains. Therefore, molecules derived from natural products as the monoterpene carvacrol, have attracted interest as promising anti-leishmania agents. However, the therapeutic use of carvacrol is limited due to its low aqueous solubility, rapid oxidation and volatilization. Thus, the development of nanostructured lipid carriers (NLCs) was proposed in the present study as a promising nanotechnology strategy to overcome these limitations and enable the use of carvacrol in leishmaniasis therapy. Carvacrol NLCs were obtained using a warm microemulsion method, and evaluated regarding the influence of lipid matrix and components concentration on the NLCs formation. NLCs were characterized by DSC and XRD as well. In addition, to the in vitro carvacrol release from NLCs, the in vitro cytotoxicity and leishmanicidal activity assays, and the in vivo pharmacokinetics evaluation of free and encapsulated carvacrol were performed. NLCs containing carvacrol were obtained successfully using a warm microemulsion dilution method. The NLCs formulation with the lowest particle size (98.42 ± 0.80 nm), narrowest size distribution (suitable for intravenous administration), and the highest encapsulation efficiency was produced by using beeswax as solid lipid (HLB=9) and 5% of lipids and surfactant. The in vitro release of carvacrol from NLCs was fitted to the Korsmeyer and Peppas, and Weibull models, demonstrating that the release mechanism is probably the Fickian diffusion type. Moreover, carvacrol encapsulation in NLCs provided a lower cytotoxicity in comparison to free carvacrol (p<0.05), increasing its in vitro leishmanicidal efficacy in the amastigote form. Finally, the in vivo pharmacokinetics of carvacrol after IV bolus administration suggests that this phenolic monoterpene undergoes enterohepatic circulation and therefore presented a long half-life (t1/2) and low clearance (Cl). In addition, C0, mean residence time (MRT) and Vdss of encapsulated carvacrol were higher than free carvacrol (p < 0.05), favoring a higher distribution of carvacrol in the target tissues. Thus, it is possible to conclude that the developed NLCs are a promising delivery system for leishmaniasis treatment.


Subject(s)
Antiprotozoal Agents/administration & dosage , Cymenes/administration & dosage , Drug Carriers/administration & dosage , Leishmania/drug effects , Nanostructures/administration & dosage , Animals , Antiprotozoal Agents/blood , Antiprotozoal Agents/chemistry , Antiprotozoal Agents/pharmacokinetics , Cell Survival/drug effects , Cymenes/blood , Cymenes/pharmacokinetics , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Liberation , Humans , Leishmaniasis/drug therapy , Lipids/administration & dosage , Lipids/chemistry , Lipids/pharmacokinetics , Macrophages, Peritoneal/drug effects , Male , Mice, Inbred BALB C , Nanostructures/chemistry , Rats, Wistar , THP-1 Cells
8.
Yale J Biol Med ; 92(3): 511-521, 2019 09.
Article in English | MEDLINE | ID: mdl-31543712

ABSTRACT

The Leishmania parasitophorous vacuole membrane (LPVM) sits at the interface of the parasite and its host. Evidence shows that molecules from the endocytic pathway as well as molecules from the secretory pathway are localized in the LPV and displayed on LPVM. In the review, we discuss our current understanding of the composition of the LPVM.


Subject(s)
Host-Parasite Interactions , Intracellular Membranes/metabolism , Leishmania/metabolism , Vacuoles/metabolism , Animals , Autophagy , Humans , Lipids/chemistry
9.
PLoS One ; 13(11): e0206920, 2018.
Article in English | MEDLINE | ID: mdl-30399177

ABSTRACT

Leishmaniasis is amongst the most important neglected diseases, afflicting more than 12 million people in 88 countries. There is an urgent need for safe orally bioavailable and cost-effective drugs for the treatment of leishmaniasis. It has recently been shown that Leishmania activates host macrophage serine/threonine kinase Akt, to promote survival of both parasites and infected cells. Here, we sought to evaluate a compound, Miransertib (ARQ 092), an orally bioavailable and selective allosteric Akt inhibitor currently in clinical trials for patients with PI3K/Akt-driven tumors or Proteus syndrome. Miransertib was tested against Leishmania donovani and Leishmania amazonensis, causative agents of visceral and cutaneous leishmaniasis, respectively. Cultured promastigotes were susceptible to Miransertib. In addition, Miransertib was markedly effective against intracellular amastigotes of L. donovani or L. amazonensis-infected macrophages. Miransertib also enhanced mTOR dependent autophagy in Leishmania-infected macrophages, which may represent one mechanism of Miransertib-mediated killing of intracellular Leishmania. Whereas parasite clearance in the spleen of mice infected with L. donovani and treated with Miransertib was comparable to that when treated with miltefosine, Miransertib caused a greater reduction in the parasite load in the liver. In the cutaneous leishmaniasis infection model, lesions were reduced by 40% as compared to mock treated mice. Together, these results provide direct evidence to support the conclusion that Miransertib is an excellent lead compound for the development of a new oral drug therapy for visceral and cutaneous leishmaniasis.


Subject(s)
Aminopyridines/administration & dosage , Imidazoles/administration & dosage , Leishmania donovani/drug effects , Leishmaniasis, Cutaneous/drug therapy , Leishmaniasis, Visceral/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Administration, Oral , Animals , Humans , Leishmania donovani/pathogenicity , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis, Visceral/parasitology , Macrophages/drug effects , Macrophages/parasitology , Mice , Parasite Load , Spleen/drug effects , Spleen/parasitology
10.
Cell Microbiol ; 20(11): e12889, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29993167

ABSTRACT

Miltefosine is an important drug for the treatment of leishmaniasis; however, its mechanism of action is still poorly understood. In these studies, we tested the hypothesis that like in cancer cells, miltefosine's efficacy in leishmaniasis is due to its inhibition of Akt activation in host cells. We show using pharmacologic agents that block Akt activation by different mechanisms and also using an inducible knockdown approach that miltefosine loses its efficacy when its access to Akt1 is limited. Interestingly, limitation of Akt activation results in clearance of established Leishmania infections. We then show, using fluorophore-tagged probes that bind to phosphoinositides, that Leishmania parasitophorous vacuole membranes (LPVMs) display the relevant phosphoinositides to which Akt can be recruited and activated continuously. Taken together, we propose that the acquisition of PI(4) P and the display of PI (3,4)P2 on LPVMs initiate the machinery that supports continuous Akt activation and sensitivity to miltefosine.


Subject(s)
Leishmania/drug effects , Leishmaniasis/drug therapy , Phosphatidylinositols/metabolism , Phosphorylcholine/analogs & derivatives , Proto-Oncogene Proteins c-akt/metabolism , Animals , Antiprotozoal Agents/pharmacology , Gene Knockdown Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Leishmaniasis/metabolism , Mice , Mice, Inbred BALB C , Molecular Targeted Therapy , Phosphorylcholine/pharmacology , Proto-Oncogene Proteins c-akt/genetics , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Transfection , Vacuoles/drug effects
11.
PLoS Negl Trop Dis ; 11(5): e0005556, 2017 May.
Article in English | MEDLINE | ID: mdl-28505157

ABSTRACT

In infected mammalian cells, Leishmania parasites reside within specialized compartments called parasitophorous vacuoles (LPVs). We have previously shown that Retro-2, a member of a novel class of small retrograde pathway inhibitors caused reduced LPV sizes and lower parasite numbers during experimental L. mexicana sp. infections. The purpose of this study was to determine if structural analogs of Retro-2cycl reported to have superior potency in the inhibition of retrograde pathway-dependent phenomena (i.e., polyomavirus cellular infection by polyomavrius and Shiga toxin trafficking in cells) are also more effective than the parent compound at controlling Leishmania infections. In addition to their effects on LPV development, we show that two optimized analogs of Retro-2cycl, DHQZ 36 and DHQZ 36.1 limit Leishmania amazonensis infection in macrophages at EC50 of 13.63+/-2.58µM and10.57+/-2.66µM, respectively, which is significantly lower than 40.15µM the EC50 of Retro-2cycl. In addition, these analogs caused a reversal in Leishmania induced suppression of IL-6 release by infected cells after LPS activation. Moreover, we show that in contrast to Retro-2cycl that is Leishmania static, the analogs can kill Leishmania parasites in axenic cultures, which is a desirable attribute for any drug to treat Leishmania infections. Together, these studies validate and extend the published structure-activity relationship analyses of Retro-2cycl.


Subject(s)
Benzamides/pharmacology , Interleukin-6/metabolism , Leishmania/drug effects , Macrophages/parasitology , Thiophenes/pharmacology , Vacuoles/parasitology , Animals , Leishmania/classification , Leishmaniasis/drug therapy , Mice , RAW 264.7 Cells
12.
Cell Immunol ; 309: 19-22, 2016 11.
Article in English | MEDLINE | ID: mdl-27622385

ABSTRACT

PI3K signaling plays a role in the host response to Leishmania infections. At the cellular level PI3K signaling is engaged by the parasite to control several cellular processes, which ensures parasite persistence. At the systemic level, there is evidence that recruitment of regulatory cells into lesions is impaired in the absence of robust PI3K signaling. In this mini-review the more recent studies that investigated the roles of PI3K signaling in Leishmania infections are discussed.


Subject(s)
Host-Parasite Interactions , Leishmania/immunology , Leishmaniasis/immunology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Animals , Humans , Immune Evasion
13.
Front Immunol ; 7: 146, 2016.
Article in English | MEDLINE | ID: mdl-27148265

ABSTRACT

Protein kinases play important roles in the regulation of cellular activities. In cells infected by pathogens, there is an increasing appreciation that dysregulated expression of protein kinases promotes the success of intracellular infections. In Leishmania-infected cells, expression and activation of protein kinases, such as the mitogen-activated protein kinases, kinases in the PI3-kinase signaling pathway, and kinases in the NF-κB-signaling pathway, are modulated in some manner. Several recent reviews have discussed our current understanding of the roles of these kinases in Leishmania infections. Apart from the kinases in the pathways enumerated above, there are other host cell protein kinases that are activated during the Leishmania infection of mammalian cells whose roles also appear to be significant. This review discusses recent observations on the Abl family of protein kinases and the protein kinase regulated by RNA in Leishmania infections.

14.
Microbes Infect ; 16(9): 721-6, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25107580

ABSTRACT

Parasites of the Leishmania genus are the causative agents of a complex disease called leishmaniasis. Many activities of infected cells including their responses to a range of stimuli are modulated by Leishmania parasites. This review will profile some of the parasite molecules that target host cell processes for which there has been recent progress.


Subject(s)
Host-Parasite Interactions , Leishmania/pathogenicity , Animals , Dendritic Cells/parasitology , Humans , Leishmania/physiology , Leishmaniasis/parasitology , Macrophages/parasitology
15.
PLoS Negl Trop Dis ; 8(7): e3000, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25033301

ABSTRACT

Professional phagocytes generate a myriad of antimicrobial molecules to kill invading microorganisms, of which nitrogen oxides are integral in controlling the obligate intracellular pathogen Leishmania. Although reactive nitrogen species produced by the inducible nitric oxide synthase (iNOS) can promote the clearance of intracellular parasites, some Leishmania species/stages are relatively resistant to iNOS-mediated antimicrobial activity. The underlying mechanism for this resistance remains largely uncharacterized. Here, we show that the amastigote form of L. amazonensis is hyper-resistant to the antimicrobial actions of cytokine-activated murine and human macrophages as compared to its promastigote counterpart. Amastigotes exhibit a marked ability to directly counter the cytotoxicity of peroxynitrite (ONOO-), a leishmanicidal oxidant that is generated during infection through the combined enzymatic activities of NADPH oxidase and iNOS. The enhanced antinitrosative defense of amastigotes correlates with the increased expression of a tryparedoxin peroxidase (TXNPx) isoform that is also upregulated in response to iNOS enzymatic activity within infected macrophages. Accordingly, ectopic over-expression of the TXNPx isoform by L. amazonensis promastigotes significantly enhances parasite resistance against ONOO- cytotoxicity. Moreover, TXNPx-overexpressing parasites exhibit greater intra-macrophage survival, and increased parasite growth and lesion development in a murine model of leishmaniasis. Our investigations indicate that TXNPx isoforms contribute to Leishmania's ability to adapt to and antagonize the hostile microenvironment of cytokine-activated macrophages, and provide a mechanistic explanation for persistent infection in experimental and human leishmaniasis.


Subject(s)
Leishmania , Macrophages/parasitology , Peroxidases/immunology , Protozoan Proteins/immunology , Animals , Humans , Leishmania/enzymology , Leishmania/metabolism , Leishmania/pathogenicity , Leishmania/physiology , Mice
16.
Front Immunol ; 4: 156, 2013.
Article in English | MEDLINE | ID: mdl-23801993

ABSTRACT

Leishmaniasis is a complex disease that is caused by parasites of the Leishmania genus. Leishmania are further classified into several complexes, each of which can engage in distinct interactions with mammalian hosts resulting in differing disease presentations. It is therefore not unexpected that host immune responses to Leishmania are variable. The induction of interferon gamma (IFN-γ) and response to it in these infections has received considerable attention. In this review, we summarize our current understanding of some of the host responses during Leishmania infections that are regulated by IFN-γ. In addition, studies that explore the nature of parasite-derived molecular mediators that might affect the host response to IFN-γ are also discussed.

17.
Cell Microbiol ; 14(11): 1676-86, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22862745

ABSTRACT

A subgroup of intracellular pathogens reside and replicate within membrane-bound compartments often termed pathogen-containing compartments (PCC). PCCs navigate around a wide range of host cell vesicles and organelles. In light of the perils of engaging with vesicles of the endocytic pathway, most PCCs modulate their interactions with endocytic vesicles while a few avoid those interactions. The secretory pathway constitutes another important grouping of vesicles and organelles in host cells. Although the negative consequences of engaging with the secretory pathway are not known, there is evidence that PCCs interact differentially with vesicles and organelles in this pathway as well. In this review, we consider three prokaryote pathogens and two protozoan parasites for which there is information on the interactions of their PCCs with the secretory pathway. Current understandings of the molecular interactions as well as the metabolic benefits that accompany those interactions are discussed. Not unexpectedly, our understanding of the extent of these interactions is variable. An underlying theme that is brought to the fore is that PCCs establish preferential interactions with distinct compartments of the secretory pathway.


Subject(s)
Bacteria/pathogenicity , Endosomes/microbiology , Endosomes/parasitology , Host-Pathogen Interactions , Leishmania/pathogenicity , Secretory Pathway , Toxoplasma/pathogenicity , Animals , Humans , Models, Biological
18.
Am J Pathol ; 181(4): 1348-55, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22885104

ABSTRACT

Our previous observations established a role for syntaxin-5 in the development of Leishmania parasitophorous vacuoles (LPVs). In this study, we took advantage of the recent identification of Retro-2, a small organic molecule that can cause the redistribution of syntaxin-5; we show herein that Retro-2 blocks LPV development within 2 hours of adding it to cells infected with Leishmania amazonensis. In infected cells incubated for 48 hours with Retro-2, LPV development was significantly limited; furthermore, infected cells harbored four to five times fewer parasites than infected cells incubated in vehicle alone. In vivo studies revealed that Retro-2 curbed experimental L. amazonensis infections in a dose-dependent manner. Retro-2 did not have any appreciable effect on the host cell physiological characteristics; furthermore, it had no apparent toxicity in experimental animals. An unexpected, but welcome, finding was that Retro-2 inhibited the replication of Leishmania parasites in axenic cultures. This study is significant because it identifies an endoplasmic reticulum/Golgi SNARE as a potential target for the control of Leishmania infections; moreover, it suggests that small organic molecules can be identified that can selectively disrupt the vesicle fusion machinery that promotes the development of pathogen-containing compartments without exerting toxic effects on the host.


Subject(s)
Leishmania/physiology , Leishmaniasis/pathology , Leishmaniasis/parasitology , Macrophages, Peritoneal/parasitology , Qa-SNARE Proteins/antagonists & inhibitors , Vacuoles/metabolism , Animals , Axenic Culture , Benzamides/pharmacology , Cell Line , Interleukin-6/metabolism , Leishmania/drug effects , Leishmania/growth & development , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , Protein Transport/drug effects , Qa-SNARE Proteins/metabolism , R-SNARE Proteins/metabolism , Thiophenes/pharmacology , Vacuoles/drug effects
19.
Cell Microbiol ; 14(6): 937-48, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22309219

ABSTRACT

Parasitophorous vacuoles (PV) that harbour Leishmania parasites acquire some characteristics from fusion with host cell vesicles. Recent studies have shown that PVs acquire and display resident endoplasmic reticulum (ER) molecules. We investigated the importance of ER molecules to PV biology by assessing the consequence of blocking the fusion of PVs with vesicles that originate from the early secretory pathway. This was achieved by targeting the N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) that mediate the fusion of early secretory vesicles. In the presence of dominant negative variants of sec22b or some of its known cognate partners, D12 and syntaxin 18, PVs failed to distend and harboured fewer parasites. These observations were confirmed in studies in which each of the SNAREs listed above including the intermediate compartment ER/Golgi SNARE, syntaxin 5, was knocked down. The knock-down of these SNARES had little or no measurable effect on the morphology of the ER or on activated secretion even though they resulted in a more significant reduction of PV size. Moreover, the knock-down of the ER/Golgi SNAREs resulted in significant reduction in parasite replication. Taken together, these studies provide further evidence that PVs acquire ER components by fusing with vesicles derived from the early secretory pathway; disruption of this interaction results in inhibition of the development of PVs as well as the limitation of parasite replication within infected cells.


Subject(s)
Endoplasmic Reticulum/parasitology , Host-Parasite Interactions , Leishmania/physiology , Macrophages/parasitology , Membrane Fusion , Vacuoles/parasitology , Animals , Cell Line , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/physiology , Gene Knockdown Techniques , Intracellular Membranes/physiology , Leishmania/growth & development , Leishmaniasis/parasitology , Mice , RNA Interference , SNARE Proteins/genetics , SNARE Proteins/metabolism , Vacuoles/metabolism , Vacuoles/physiology
20.
PLoS Negl Trop Dis ; 4(10)2010 Oct 05.
Article in English | MEDLINE | ID: mdl-20957202

ABSTRACT

Although Leishmania parasites have been shown to modulate their host cell's responses to multiple stimuli, there is limited evidence that parasite molecules are released into infected cells. In this study, we present an implementation of the change mediated antigen technology (CMAT) to identify parasite molecules that are preferentially expressed in infected cells. Sera from mice immunized with cell lysates prepared from L. donovani or L. pifanoi-infected macrophages were adsorbed with lysates of axenically grown amastigotes of L. donovani or L. pifanoi, respectively, as well as uninfected macrophages. The sera were then used to screen inducible parasite expression libraries constructed with genomic DNA. Eleven clones from the L. pifanoi and the L. donovani screen were selected to evaluate the characteristics of the molecules identified by this approach. The CMAT screen identified genes whose homologs encode molecules with unknown function as well as genes that had previously been shown to be preferentially expressed in the amastigote form of the parasite. In addition a variant of Tryparedoxin peroxidase that is preferentially expressed within infected cells was identified. Antisera that were then raised to recombinant products of the clones were used to validate that the endogenous molecules are preferentially expressed in infected cells. Evaluation of the distribution of the endogenous molecules in infected cells showed that some of these molecules are secreted into parasitophorous vacuoles (PVs) and that they then traffic out of PVs in vesicles with distinct morphologies. This study is a proof of concept study that the CMAT approach can be applied to identify putative Leishmania parasite effectors molecules that are preferentially expressed in infected cells. In addition we provide evidence that Leishmania molecules traffic out of the PV into the host cell cytosol and nucleus.


Subject(s)
Antigens, Protozoan/biosynthesis , Gene Expression Profiling/methods , Host-Parasite Interactions , Leishmania/genetics , Leishmania/pathogenicity , Protozoan Proteins/biosynthesis , Animals , Antibodies, Protozoan/immunology , Antigens, Protozoan/genetics , Cells, Cultured , Gene Library , Macrophages/parasitology , Mice , Protozoan Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...