Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
1.
J Vet Med Sci ; 85(11): 1146-1150, 2023 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-37766567

RESUMEN

Many parasite species migrate to another site of infection after entering the host body. Such parasite dynamics are closely related to pathogenicity, but it is not easy to observe such parasite behavior deep within the organs. In recent years, technology that can make organs transparent has been developed that enables us to observe deep within organs ex vivo while maintaining their three-dimensional structure. This review describes a series of attempts to apply this technology to understand the behavior of Toxoplasma gondii in the host body. A series of studies has shown that T. gondii tachyzoites that infect leukocytes can reach target organs far from the site of invasion via the circulatory system. In addition, infected leukocytes in the bloodstream adhere more readily to vascular endothelial cells than uninfected leukocytes and are more likely to remain inside the target organs. When infected leukocytes adhere to the vascular endothelial cells of the target organ, the tachyzoites inside the cells immediately escape and infect the parenchyma of the organs. As described above, organ transparency technology is a powerful tool for understanding the internal dynamics of parasites.


Asunto(s)
Parásitos , Toxoplasma , Animales , Células Endoteliales/parasitología , Leucocitos
2.
Proc Natl Acad Sci U S A ; 119(36): e2206327119, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-36037380

RESUMEN

Cerebral malaria (CM) is a life-threatening form of Plasmodium falciparum infection caused by brain inflammation. Brain endothelium dysfunction is a hallmark of CM pathology, which is also associated with the activation of the type I interferon (IFN) inflammatory pathway. The molecular triggers and sensors eliciting brain type I IFN cellular responses during CM remain largely unknown. We herein identified the stimulator of interferon response cGAMP interactor 1 (STING1) as the key innate immune sensor that induces Ifnß1 transcription in the brain of mice infected with Plasmodium berghei ANKA (Pba). This STING1/IFNß-mediated response increases brain CXCL10 governing the extent of brain leukocyte infiltration and blood-brain barrier (BBB) breakdown, and determining CM lethality. The critical role of brain endothelial cells (BECs) in fueling type I IFN-driven brain inflammation was demonstrated in brain endothelial-specific IFNß-reporter and STING1-deficient Pba-infected mice, which were significantly protected from CM lethality. Moreover, extracellular particles (EPs) released from Pba-infected erythrocytes activated the STING1-dependent type I IFN response in BECs, a response requiring intracellular acidification. Fractionation of the EPs enabled us to identify a defined fraction carrying hemoglobin degradation remnants that activates STING1/IFNß in the brain endothelium, a process correlated with heme content. Notably, stimulation of STING1-deficient BECs with heme, docking experiments, and in vitro binding assays unveiled that heme is a putative STING1 ligand. This work shows that heme resultant from the parasite heterotrophic activity operates as an alarmin, triggering brain endothelial inflammatory responses via the STING1/IFNß/CXCL10 axis crucial to CM pathogenesis and lethality.


Asunto(s)
Encéfalo , Hemo , Interferón beta , Malaria Cerebral , Proteínas de la Membrana , Animales , Encéfalo/parasitología , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Células Endoteliales/parasitología , Endotelio/inmunología , Endotelio/parasitología , Hemo/metabolismo , Interferón beta/inmunología , Malaria Cerebral/inmunología , Malaria Cerebral/parasitología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Plasmodium berghei/metabolismo , Activación Transcripcional/inmunología
3.
PLoS Negl Trop Dis ; 16(1): e0010074, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34986160

RESUMEN

The protozoan parasite, Trypanosoma cruzi, causes severe morbidity and mortality in afflicted individuals. Approximately 30% of T. cruzi infected individuals present with cardiac pathology. The invasive forms of the parasite are carried in the vascular system to infect other cells of the body. During transportation, the molecular mechanisms by which the parasite signals and interact with host endothelial cells (EC) especially heart endothelium is currently unknown. The parasite increases host thrombospondin-1 (TSP1) expression and activates the Wnt/ß-catenin and hippo signaling pathways during the early phase of infection. The links between TSP1 and activation of the signaling pathways and their impact on parasite infectivity during the early phase of infection remain unknown. To elucidate the significance of TSP1 function in YAP/ß-catenin colocalization and how they impact parasite infectivity during the early phase of infection, we challenged mouse heart endothelial cells (MHEC) from wild type (WT) and TSP1 knockout mice with T. cruzi and evaluated Wnt signaling, YAP/ß-catenin crosstalk, and how they affect parasite infection. We found that in the absence of TSP1, the parasite induced the expression of Wnt-5a to a maximum at 2 h (1.73±0.13), P< 0.001 and enhanced the level of phosphorylated glycogen synthase kinase 3ß at the same time point (2.99±0.24), P<0.001. In WT MHEC, the levels of Wnt-5a were toned down and the level of p-GSK-3ß was lowest at 2 h (0.47±0.06), P< 0.01 compared to uninfected control. This was accompanied by a continuous significant increase in the nuclear colocalization of ß-catenin/YAP in TSP1 KO MHEC with a maximum Pearson correlation coefficient of (0.67±0.02), P< 0.05 at 6 h. In WT MHEC, the nuclear colocalization of ß-catenin/YAP remained steady and showed a reduction at 6 h (0.29±0.007), P< 0.05. These results indicate that TSP1 plays an important role in regulating ß-catenin/YAP colocalization during the early phase of T. cruzi infection. Importantly, dysregulation of this crosstalk by pre-incubation of WT MHEC with a ß-catenin inhibitor, endo-IWR 1, dramatically reduced the level of infection of WT MHEC. Parasite infectivity of inhibitor treated WT MHEC was similar to the level of infection of TSP1 KO MHEC. These results indicate that the ß-catenin pathway induced by the parasite and regulated by TSP1 during the early phase of T. cruzi infection is an important potential therapeutic target, which can be explored for the prophylactic prevention of T. cruzi infection.


Asunto(s)
Enfermedad de Chagas/patología , Vía de Señalización Hippo/fisiología , Trombospondina 1/metabolismo , Vía de Señalización Wnt/fisiología , Proteínas Señalizadoras YAP/metabolismo , beta Catenina/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Animales , Células Endoteliales/parasitología , Endotelio/citología , Endotelio/parasitología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Corazón/parasitología , Ratones , Ratones Noqueados , Ratas , Trombospondina 1/genética , Trypanosoma cruzi/metabolismo , Proteína Wnt-5a/metabolismo , beta Catenina/antagonistas & inhibidores
4.
Infect Immun ; 89(10): e0017821, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34228487

RESUMEN

The Chagas disease parasite Trypanosoma cruzi must extravasate to home in on susceptible cells residing in most tissues. It remains unknown how T. cruzi undertakes this crucial step of its life cycle. We hypothesized that the pathogen exploits the endothelial cell programming leukocytes use to extravasate to sites of inflammation. Transendothelial migration (TEM) starts after inflammatory cytokines induce E-selectin expression and P-selectin translocation on endothelial cells (ECs), enabling recognition by leukocyte ligands that engender rolling cell adhesion. Here, we show that T. cruzi upregulates E- and P-selectins in cardiac ECs to which it binds in a ligand-receptor fashion, whether under static or shear flow conditions. Glycoproteins isolated from T. cruzi (TcEx) specifically recognize P-selectin in a ligand-receptor interaction. As with leukocytes, binding of P-selectin to T. cruzi or TcEx requires sialic acid and tyrosine sulfate, which are pivotal for downstream migration across ECs and extracellular matrix proteins. Additionally, soluble selectins, which bind T. cruzi, block transendothelial migration dose dependently, implying that the pathogen bears selectin-binding ligand(s) that start transmigration. Furthermore, function-blocking antibodies against E- and P-selectins, which act on endothelial cells and not T. cruzi, are exquisite in preventing TEM. Thus, our results show that selectins can function as mediators of T. cruzi transendothelial transmigration, suggesting a pathogenic mechanism that allows homing in of the parasite on targeted tissues. As selectin inhibitors are sought-after therapeutic targets for autoimmune diseases and cancer metastasis, they may similarly represent a novel strategy for Chagas disease therapy.


Asunto(s)
Selectina E/metabolismo , Células Endoteliales/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Selectina-P/metabolismo , Trypanosoma cruzi/metabolismo , Animales , Adhesión Celular/fisiología , Citocinas/metabolismo , Células Endoteliales/parasitología , Femenino , Humanos , Inflamación/metabolismo , Inflamación/parasitología , Leucocitos/metabolismo , Leucocitos/parasitología , Ligandos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Migración Transendotelial y Transepitelial/fisiología
5.
Toxins (Basel) ; 13(1)2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33467515

RESUMEN

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


Asunto(s)
Células Endoteliales/parasitología , Malaria Falciparum/parasitología , Necrosis/parasitología , Perforina/efectos adversos , Proteínas Protozoarias/efectos adversos , Animales , Apoptosis , Biomarcadores/metabolismo , Barrera Hematoencefálica , Calcio/metabolismo , Línea Celular , Permeabilidad de la Membrana Celular , Supervivencia Celular , Perros , Eritrocitos/parasitología , Proteína HMGB1/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células de Riñón Canino Madin Darby , Membranas Mitocondriales , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidad , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes
6.
Biochim Biophys Acta Mol Cell Res ; 1868(2): 118898, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33157166

RESUMEN

Intracellular pathogens like Toxoplasma gondii often target proteins and pathways critical for host cell survival and stress response. Molecular chaperones encoded by the evolutionary conserved Heat shock proteins (Hsps) maintain proteostasis and are vital to cell survival following exposure to any form of stress. A key protein of this family is Hsp70, an ATP-driven molecular chaperone, which is stress inducible and often indiscernible in normal cells. Role of this protein with respect to intracellular survival and multiplication of protozoan parasite like T. gondii remains to be examined. We find that T. gondii infection upregulates expression of host Hsp70. Hsp70 selective inhibitor 2-phenylethynesulfonamide (PES) attenuates intracellular T. gondii multiplication. Biotinylated PES confirms selective interaction of this small molecule inhibitor with Hsp70. We show that PES acts by disrupting Hsp70 chaperone function which leads to dysregulation of host autophagy. Silencing of host Hsp70 underscores its importance for intracellular multiplication of T. gondii, however, attenuation achieved using PES is not completely attributable to host Hsp70 indicating the presence of other intracellular targets of PES in infected host cells. We find that PES is also able to target T. gondii Hsp70 homologue which was shown using PES binding assay. Detailed molecular docking analysis substantiates PES targeting of TgHsp70 in addition to host Hsp70. While establishing the importance of protein quality control in infection, this study brings to the fore a unique opportunity of dual targeting of host and parasite Hsp70 demonstrating how structural conservation of these proteins may be exploited for therapeutic design.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Espacio Intracelular/metabolismo , Toxoplasma/fisiología , Toxoplasmosis/metabolismo , Animales , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Línea Celular , Células Endoteliales/parasitología , Proteínas HSP70 de Choque Térmico/genética , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Ratones , Microglía/parasitología , Simulación del Acoplamiento Molecular , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/parasitología , Sulfonamidas/farmacología , Toxoplasmosis/parasitología , Transfección
7.
PLoS Pathog ; 16(10): e1008917, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33017449

RESUMEN

Babesia bovis causes a pathogenic form of babesiosis in cattle. Following invasion of red blood cells (RBCs) the parasite extensively modifies host cell structural and mechanical properties via the export of numerous proteins. Despite their crucial role in virulence and pathogenesis, such proteins have not been comprehensively characterized in B. bovis. Here we describe the surface biotinylation of infected RBCs (iRBCs), followed by proteomic analysis. We describe a multigene family (mtm) that encodes predicted multi-transmembrane integral membrane proteins which are exported and expressed on the surface of iRBCs. One mtm gene was downregulated in blasticidin-S (BS) resistant parasites, suggesting an association with BS uptake. Induced knockdown of a novel exported protein encoded by BBOV_III004280, named VESA export-associated protein (BbVEAP), resulted in a decreased growth rate, reduced RBC surface ridge numbers, mis-localized VESA1, and abrogated cytoadhesion to endothelial cells, suggesting that BbVEAP is a novel virulence factor for B. bovis.


Asunto(s)
Babesia bovis/patogenicidad , Babesiosis/parasitología , Células Endoteliales/parasitología , Eritrocitos/parasitología , Animales , Babesia bovis/genética , Bovinos , Enfermedades de los Bovinos/parasitología , Proteínas de la Membrana , Parásitos/patogenicidad , Proteómica/métodos , Factores de Virulencia/genética
8.
Malar J ; 19(1): 376, 2020 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-33087130

RESUMEN

BACKGROUND: Cerebral malaria (CM) is associated with morbidity and mortality despite the use of potent anti-malarial agents. Brain endothelial cell activation and dysfunction from oxidative and inflammatory host responses and products released by Plasmodium falciparum-infected erythrocytes (IE), are likely the major contributors to the encephalopathy, seizures, and brain swelling that are associated with CM. The development of adjunctive therapy to reduce the pathological consequences of host response pathways could improve outcomes. A potentially protective role of the nuclear factor E2-related factor 2 (NRF2) pathway, which serves as a therapeutic target in brain microvascular diseases and central nervous system (CNS) inflammatory diseases such as multiple sclerosis was tested to protect endothelial cells in an in vitro culture system subjected to tumour necrosis factor (TNF) or infected red blood cell exposure. NRF2 is a transcription factor that mediates anti-oxidant and anti-inflammatory responses. METHODS: To accurately reflect clinically relevant parasite biology a unique panel of parasite isolates derived from patients with stringently defined CM was developed. The effect of TNF and these parasite lines on primary human brain microvascular endothelial cell (HBMVEC) activation in an in vitro co-culture model was tested. HBMVEC activation was measured by cellular release of IL6 and nuclear translocation of NFκB. The transcriptional and functional effects of dimethyl fumarate (DMF), an FDA approved drug which induces the NRF2 pathway, on host and parasite induced HBMVEC activation was characterized. In addition, the effect of DMF on parasite binding to TNF stimulated HBMVEC in a semi-static binding assay was examined. RESULTS: Transcriptional profiling demonstrates that DMF upregulates the NRF2-Mediated Oxidative Stress Response, ErbB4 Signaling Pathway, Peroxisome Proliferator-activated Receptor (PPAR) Signaling and downregulates iNOS Signaling and the Neuroinflammation Signaling Pathway on TNF activated HBMVEC. The parasite lines derived from eight paediatric CM patients demonstrated increased binding to TNF activated HBMVEC and varied in their binding and activation of HBMVEC. Overall DMF reduced both TNF and CM derived parasite activation of HBMVEC. CONCLUSIONS: These findings provide evidence that targeting the NRF2 pathway in TNF and parasite activated HBMVEC mediates multiple protective pathways and may represent a novel adjunctive therapy to improve infection outcomes in CM.


Asunto(s)
Antiinflamatorios/farmacología , Dimetilfumarato/farmacología , Células Endoteliales/parasitología , Malaria Cerebral/prevención & control , Malaria Falciparum/prevención & control , Plasmodium falciparum/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Antioxidantes/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/parasitología , Niño , Preescolar , Células Endoteliales/efectos de los fármacos , Humanos , Lactante , Plasmodium falciparum/fisiología
9.
Parasitol Res ; 119(11): 3793-3801, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32979103

RESUMEN

Hepatozoon species are the most widely known haemogregarines infecting a wide range of vertebrates, although predominately snakes. Herein, Hepatozoon bashtari n. sp., originally infecting the painted saw-scaled viper, Echis coloratus, in Saudi Arabia is described using both morphological features and molecular data from 18S rDNA sequences. The overall prevalence of infection was 60% (9/15) with parasitaemia ranging from 52 to 60%. Gamonts were entirely intraerythrocytic and were observed to cause considerable hypertrophy within the host cell. The mean size of mature gamonts was 15.4 × 3.3 µm. Merogonic stages were confined to the lung endothelial cells with monomorphic meronts. The average size of mature meronts was 32 × 12 µm and they were estimated to produce 13-16 merozoites each. The phylogenetic tree generated from SSU rDNA sequences revealed that Hepatozoon bashtari sp. n. clusters with the vast majority of other Hepatozoon species infecting snakes, lizards and geckos in various regions of the world, which would appear to support the hypothesis of prey-predator transmission of the genus Hepatozoon. Through a combination of morphological comparison with closely related Hepatozoon spp. and 18S rRNA gene sequence analysis, it is possible to confirm Hepatozoon bashtari sp. n. as a new species.


Asunto(s)
Coccidiosis , Eucoccidiida/clasificación , Viperidae/parasitología , Animales , Coccidiosis/epidemiología , Coccidiosis/parasitología , Células Endoteliales/parasitología , Eucoccidiida/citología , Eucoccidiida/genética , Pulmón/parasitología , Parasitemia/epidemiología , Filogenia , Prevalencia , ARN Ribosómico 18S/genética , Arabia Saudita/epidemiología , Especificidad de la Especie
10.
J Am Soc Mass Spectrom ; 31(9): 1815-1824, 2020 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-32830963

RESUMEN

The obligate intracellular apicomplexan parasites Toxoplasma gondii and Besnoitia besnoiti are important causes of disease in both humans and cattle. To date, effective specific treatments are lacking for both infections. To counteract severe symptoms leading to, e.g., disabilities and even abortion in the case of human toxoplasmosis and bovine besnoitiosis, novel targets are required for development of drugs and vaccines. A promising emerging technique for molecular characterization of organisms is high-resolution atmospheric-pressure scanning microprobe matrix-assisted laser desorption/ionization (AP-SMALDI) mass spectrometry imaging (MSI) which enables semiquantitative visualization of metabolite distributions. MSI was here used to trace and characterize lipid metabolites in primary bovine umbilical vein endothelial cells (BUVECs) upon infection with tachyzoites, an early and pathogenic fast-replicating life stage of T. gondii and B. besnoiti. A cell bulk, derived from noninfected controls and parasite-infected cell pellets, was analyzed by AP-SMALDI MSI in technical and biological triplicates. Multivariate statistical analysis including hierarchical clustering and principle component analysis revealed infection-specific metabolites in both positive- and negative-ion mode, identified by combining database search and LC-MS2 experiments. MSI analyses of host cell monolayers were conducted at 5 µm lateral resolution, allowing single apicomplexan-infected cells to be allocated. This is the first mass spectrometry imaging study on intracellular T. gondii and B. besnoiti infections and the first detailed metabolomic characterization of B. besnoiti tachyzoites. MSI was used here as an efficient tool to discriminate infected from noninfected cells at the single-cell level in vitro.


Asunto(s)
Coccidiosis , Espacio Intracelular/parasitología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Toxoplasmosis Animal , Animales , Bovinos , Células Cultivadas , Coccidiosis/diagnóstico por imagen , Coccidiosis/parasitología , Células Endoteliales/citología , Células Endoteliales/parasitología , Imagen Molecular , Sarcocystidae/patogenicidad , Análisis de la Célula Individual , Toxoplasma/patogenicidad , Toxoplasmosis Animal/diagnóstico por imagen , Toxoplasmosis Animal/parasitología
11.
Int J Mol Sci ; 21(14)2020 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-32664627

RESUMEN

The protozoan parasite Trypanosoma cruzi is the causative agent of Chagas disease. This neglected tropical disease causes severe morbidity and mortality in endemic regions. About 30% of T. cruzi infected individuals will present with cardiac complications. Invasive trypomastigotes released from infected cells can be carried in the vascular endothelial system to infect neighboring and distant cells. During the process of cellular infection, the parasite induces host cells, to increase the levels of host thrombospondin-1 (TSP-1), to facilitate the process of infection. TSP-1 plays important roles in the functioning of vascular cells, including vascular endothelial cells with important implications in cardiovascular health. Many signal transduction pathways, including the yes-associated protein 1 (YAP)/transcriptional coactivator, with PDZ-binding motif (TAZ) signaling, which are upstream of TSP-1, have been linked to the pathophysiology of heart damage. The molecular mechanisms by which T. cruzi signals, and eventually infects, heart endothelial cells remain unknown. To evaluate the importance of TSP-1 expression in heart endothelial cells during the process of T. cruzi infection, we exposed heart endothelial cells prepared from Wild Type and TSP-1 Knockout mouse to invasive T. cruzi trypomastigotes at multiple time points, and evaluated changes in the hippo signaling cascade using immunoblotting and immunofluorescence assays. We found that the parasite turned off the hippo signaling pathway in TSP-1KO heart endothelial cells. The levels of SAV1 and MOB1A increased to a maximum of 2.70 ± 0.23 and 5.74 ± 1.45-fold at 3 and 6 h, respectively, in TSP-1KO mouse heart endothelial cells (MHEC), compared to WT MHEC, following a parasite challenge. This was accompanied by a significant continuous increase in the nuclear translocation of downstream effector molecule YAP, to a maximum mean nuclear fluorescence intensity of 10.14 ± 0.40 at 6 h, compared to wild type cells. Furthermore, we found that increased nuclear translocated YAP significantly colocalized with the transcription co-activator molecule pan-TEAD, with a maximum Pearson's correlation coefficient of 0.51 ± 0.06 at 6 h, compared to YAP-Pan-TEAD colocalization in the WT MHEC, which decreased significantly, with a minimum Pearson's correlation coefficient of 0.30 ± 0.01 at 6 h. Our data indicate that, during the early phase of infection, upregulated TSP-1 is essential for the regulation of the hippo signaling pathway. These studies advance our understanding of the molecular interactions occurring between heart endothelial cells and T. cruzi, in the presence and absence of TSP-1, providing insights into processes linked to parasite dissemination and pathogenesis.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Células Endoteliales/parasitología , Mioblastos/parasitología , Miocardio/citología , Proteínas Protozoarias/fisiología , Trombospondina 1/fisiología , Trypanosoma cruzi/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Endoteliales/metabolismo , Técnicas de Inactivación de Genes , Ratones , Mioblastos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ratas , Transducción de Señal/fisiología , Trombospondina 1/deficiencia , Transactivadores/fisiología
12.
Microvasc Res ; 131: 104024, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32502488

RESUMEN

Congenital toxoplasmosis is a parasitic disease that occurs due vertical transmission of the protozoan Toxoplasma gondii (T. gondii) during pregnancy. The parasite crosses the placental barrier and reaches the developing brain, infecting progenitor, glial, neuronal and vascular cell types. Although the role of Radial glia (RG) neural stem cells in the development of the brain vasculature has been recently investigated, the impact of T. gondii infection in these events is not yet understood. Herein, we studied the role of T. gondii infection on RG cell function and its interaction with endothelial cells. By infecting isolated RG cultures with T. gondii tachyzoites, we observed a cytotoxic effect with reduced numbers of RG populations together with decrease neuronal and oligodendrocyte progenitor populations. Conditioned medium (CM) from RG control cultures increased ZO-1 protein levels and organization on endothelial bEnd.3 cells membranes, which was impaired by CM from infected RG, accompanied by decreased trans-endothelial electrical resistance (TEER). ELISA assays revealed reduced levels of anti-inflammatory cytokine TGF-ß1 in CM from T. gondii-infected RG cells. Treatment with recombinant TGF-ß1 concomitantly with CM from infected RG cultures led to restoration of ZO-1 staining in bEnd.3 cells. Congenital infection in Swiss Webster mice led to abnormalities in the cortical microvasculature in comparison to uninfected embryos. Our results suggest that infection of RG cells by T. gondii negatively modulates cytokine secretion, which might contribute to endothelial loss of barrier properties, thus leading to impairment of neurovascular interaction establishment.


Asunto(s)
Diferenciación Celular , Corteza Cerebral/irrigación sanguínea , Células Endoteliales/parasitología , Células Ependimogliales/parasitología , Microvasos/parasitología , Acoplamiento Neurovascular , Toxoplasma/patogenicidad , Toxoplasmosis Cerebral/parasitología , Toxoplasmosis Congénita/parasitología , Animales , Línea Celular , Modelos Animales de Enfermedad , Impedancia Eléctrica , Células Endoteliales/metabolismo , Células Endoteliales/patología , Células Ependimogliales/metabolismo , Células Ependimogliales/patología , Ratones Endogámicos C57BL , Microvasos/metabolismo , Microvasos/patología , Uniones Estrechas/metabolismo , Uniones Estrechas/parasitología , Uniones Estrechas/patología , Toxoplasmosis Cerebral/metabolismo , Toxoplasmosis Cerebral/patología , Toxoplasmosis Congénita/metabolismo , Toxoplasmosis Congénita/patología , Factor de Crecimiento Transformador beta1/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
13.
Parasitol Res ; 119(8): 2563-2577, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32548739

RESUMEN

Besnoitia besnoiti is an important obligate intracellular parasite of cattle which primarily infects host endothelial cells of blood vessels during the acute phase of infection. Similar to the closely related parasite Toxoplasma gondii, B. besnoiti has fast proliferating properties leading to rapid host cell lysis within 24-30 h p.i. in vitro. Some apicomplexan parasites were demonstrated to modulate the host cellular cell cycle to successfully perform their intracellular development. As such, we recently demonstrated that T. gondii tachyzoites induce G2/M arrest accompanied by chromosome missegregation, cell spindle alteration, formation of supernumerary centrosomes, and cytokinesis impairment when infecting primary bovine umbilical vein endothelial cells (BUVEC). Here, we follow a comparative approach by using the same host endothelial cell system for B. besnoiti infections. The current data showed that-in terms of host cell cycle modulation-infections of BUVEC by B. besnoiti tachyzoites indeed differ significantly from those by T. gondii. As such, cyclin expression patterns demonstrated a significant upregulation of cyclin E1 in B. besnoiti-infected BUVEC, thereby indicating parasite-driven host cell stasis at G1-to-S phase transition. In line, the mitotic phase of host cell cycle was not influenced since alterations of chromosome segregation, mitotic spindle formation, and cytokinesis were not observed. In contrast to respective T. gondii-related data, we furthermore found a significant upregulation of histone H3 (S10) phosphorylation in B. besnoiti-infected BUVEC, thereby indicating enhanced chromosome condensation to occur in these cells. In line to altered G1/S-transition, we here additionally showed that subcellular abundance of proliferating cell nuclear antigen (PCNA), a marker for G1 and S phase sub-stages, was affected by B. besnoiti since infected cells showed increased nuclear PCNA levels when compared with that of control cells.


Asunto(s)
Enfermedades de los Bovinos/fisiopatología , Coccidiosis/veterinaria , Puntos de Control de la Fase G2 del Ciclo Celular , Puntos de Control de la Fase M del Ciclo Celular , Sarcocystidae/fisiología , Animales , Apoptosis , Bovinos , Enfermedades de los Bovinos/parasitología , Coccidiosis/parasitología , Coccidiosis/fisiopatología , Células Endoteliales/citología , Células Endoteliales/parasitología
14.
Molecules ; 25(7)2020 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-32230894

RESUMEN

Malaria is a life-threatening disease and, what is more, the resistance to available antimalarial drugs is a recurring problem. The resistance of Plasmodium falciparum malaria parasites to previous generations of medicines has undermined malaria control efforts and reversed gains in child survival. This paper describes a continuation of our ongoing efforts to investigate the effects against Plasmodium falciparum strains and human microvascular endothelial cells (HMEC-1) of a series of methoxy p-benzyl-substituted thiazinoquinones designed starting from a pointed antimalarial lead candidate. The data obtained from the newly tested compounds expanded the structure-activity relationships (SARs) of the thiazinoquinone scaffold, indicating that antiplasmodial activity is not affected by the inductive effect but rather by the resonance effect of the introduced group at the para position of the benzyl substituent. Indeed, the current survey was based on the evaluation of antiparasitic usefulness as well as the selectivity on mammalian cells of the tested p-benzyl-substituted thiazinoquinones, upgrading the knowledge about the active thiazinoquinone scaffold.


Asunto(s)
Antimaláricos/farmacología , Células Endoteliales/efectos de los fármacos , Malaria/tratamiento farmacológico , Plasmodium falciparum/efectos de los fármacos , Quinonas/química , Quinonas/farmacología , Células Endoteliales/parasitología , Concentración 50 Inhibidora , Pruebas de Sensibilidad Parasitaria , Quinonas/síntesis química , Relación Estructura-Actividad
15.
Biomed Res Int ; 2020: 3932569, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32185202

RESUMEN

Pulmonary edema (PE) is a major cause of pulmonary manifestations of severe Plasmodium falciparum malaria and is usually associated with acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). The sphingosine kinase-1 (SphK-1)/sphingosine-1-phosphate receptor-3 (S1PR-3) pathway has recently been reported to affect the pathogenesis of lung injury, but the expression of these proteins in the lungs of severe P. falciparum malaria patients has not been investigated. The cellular expression of SphK-1 and S1PR-3 in lung tissues from autopsied patients with P. falciparum malaria was investigated using immunohistochemistry (IHC). Lung tissues from patients who died of severe P. falciparum malaria were classified into two groups based on histopathological findings: those with PE (18 patients) and those without PE (non-PE, 19 patients). Ten samples of normal lung tissues were used as the control group. The protein expression levels of SphK-1 and S1PR-3 were significantly upregulated in endothelial cells (ECs), alveolar epithelial cells, and alveolar macrophages (AMs) in the lungs of severe P. falciparum malaria patients with PE compared to those in the non-PE and control groups (all p < 0.001). In addition, the SphK-1 and S1PR-3 expression levels were significantly positively correlated in pulmonary ECs (r s = 0.922, p < 0.001), alveolar epithelial cells (r s = 0.995, p < 0.001), and AMs (r s = 0.969, p < 0.001). In conclusion, both the SphK-1 and S1PR-3 proteins were overexpressed in the lung tissues of severe P. falciparum malaria patients with PE, suggesting that SphK-1 and S1PR-3 mediate the pathogenesis of PE in severe malaria. Targeting the regulation of SphK-1 and/or S1PR-3 may be an approach to treat pulmonary complications in severe P. falciparum patients.


Asunto(s)
Malaria Falciparum/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Edema Pulmonar/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/parasitología , Adulto , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/parasitología , Células Endoteliales/metabolismo , Células Endoteliales/parasitología , Femenino , Humanos , Pulmón/metabolismo , Pulmón/parasitología , Malaria Falciparum/parasitología , Masculino , Plasmodium falciparum/patogenicidad , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/parasitología , Adulto Joven
16.
Parasitol Int ; 76: 102068, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32006675

RESUMEN

Successful excystation of sporulated Eimeria spp. oocysts is an important step to acquire large numbers of viable sporozoites for molecular, biochemical, immunological and in vitro experiments for detailed studies on complex host cell-parasite interactions. An improved method for excystation of sporulated oocysts and collection of infective E. bovis- and E. arloingi-sporozoites is here described. Eimeria spp. oocysts were treated for at least 20 h with sterile 0.02 M L-cysteine HCl/0.2 M NaHCO3 solution at 37 °C in 100% CO2 atmosphere. The last oocyst treatment was performed with a 0.4% trypsin 8% sterile bovine bile excystation solution, which disrupted oocyst walls with consequent activation of sporozoites within oocyst circumplasm, thereby releasing up to 90% of sporozoites in approximately 2 h of incubation (37 °C) with a 1:3 (oocysts:sporozoites) ratio. Free-released sporozoites were filtered in order to remove rests of oocysts, sporocysts and non-sporulated oocysts. Furthermore, live cell imaging 3D holotomographic microscopy (Nanolive®) analysis allowed visualization of differing sporozoite egress strategies. Sporozoites of both species were up to 99% viable, highly motile, capable of active host cell invasion and further development into trophozoite- as well as macroment-development in primary bovine umbilical vein endothelial cells (BUVEC). Sporozoites obtained by this new excystation protocol were cleaner at the time point of exposure of BUVEC monolayers and thus benefiting from the non-activation status of these highly immunocompetent cells through debris. Alongside, this protocol improved former described methods by being is less expensive, faster, accessible for all labs with minimum equipment, and without requirement of neither expensive buffer solutions nor sophisticated instruments such as ultracentrifuges.


Asunto(s)
Eimeria/fisiología , Parasitología/métodos , Animales , Bovinos , Células Endoteliales/parasitología , Microscopía/métodos , Microscopía/veterinaria , Esporozoítos/fisiología , Venas Umbilicales/parasitología
17.
Nano Lett ; 19(12): 8887-8895, 2019 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-31671939

RESUMEN

Cerebral malaria is a lethal complication of malaria infection characterized by central nervous system dysfunction and is often not effectively treated by antimalarial combination therapies. It has been shown that the sequestration of the parasite-infected red blood cells that interact with cerebral vessel endothelial cells and the damage of the blood-brain barrier (BBB) play critical roles in the pathogenesis. In this study, we developed a ferritin nanozyme (Fenozyme) composed of recombinant human ferritin (HFn) protein shells that specifically target BBB endothelial cells (BBB ECs) and the inner Fe3O4 nanozyme core that exhibits reactive oxygen species-scavenging catalase-like activity. In the experimental cerebral malaria (ECM) mouse model, administration of the Fenozyme, but not HFn, markedly ameliorated the damage of BBB induced by the parasite and improved the survival rate of infected mice significantly. Further investigations found that Fenozyme, as well as HFn, was able to polarize the macrophages in the liver to the M1 phenotype and promote the elimination of malaria in the blood. Thus, the catalase-like activity of the Fenozyme is required for its therapeutic effect in the mouse model. Moreover, the Fenozyme significantly alleviated the brain inflammation and memory impairment in ECM mice that had been treated with artemether, indicating that combining Fenozyme with an antimalarial drug is a novel strategy for the treatment of cerebral malaria.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Ferritinas/farmacología , Malaria Cerebral/prevención & control , Plasmodium berghei/metabolismo , Animales , Barrera Hematoencefálica/parasitología , Barrera Hematoencefálica/patología , Modelos Animales de Enfermedad , Células Endoteliales/parasitología , Células Endoteliales/patología , Ferritinas/genética , Humanos , Inflamación/metabolismo , Inflamación/parasitología , Inflamación/patología , Inflamación/prevención & control , Hígado/metabolismo , Hígado/parasitología , Hígado/patología , Macrófagos/metabolismo , Macrófagos/parasitología , Macrófagos/patología , Malaria Cerebral/metabolismo , Malaria Cerebral/patología , Ratones , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
18.
Parasit Vectors ; 12(1): 517, 2019 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-31685001

RESUMEN

BACKGROUND: Bovine besnoitiosis, caused by the cyst-forming apicomplexan parasite Besnoitia besnoiti, is a chronic and debilitating cattle disease that continues to spread in Europe in the absence of control tools. In this scenario, in vitro culture systems are valuable tools to carry out drug screenings and to unravel host-parasite interactions. However, studies performed in bovine target cells are scarce. METHODS: The objective of the present study was to obtain primary bovine aortic endothelial cells (BAECs) and fibroblast cell cultures, target cells during the acute and the chronic stage of the disease, respectively, from healthy bovine donors. Afterwards, expression of surface (CD31, CD34 and CD44) and intracellular markers (vimentin and cytokeratin) was studied to characterize cell populations by flow cytometry. Next, the lytic cycle of B. besnoiti tachyzoites was studied in both target cells. Invasion rates (IRs) were determined by immunofluorescence at several time points post-infection, and proliferation kinetics were studied by quantitative PCR (qPCR). Finally, the influence of bovine viral diarrhea virus (BVDV) co-infection on the host cell machinery, and consequently on B. besnoiti invasion and proliferation, was investigated in BAECs. RESULTS: Morphology and cytometry results confirmed the endothelial and fibroblast origins. CD31 was the surface marker that best discriminated between BAECs and fibroblasts, since fibroblasts lacked CD31 labelling. Expression of CD34 was weak in low-passage BAECs and absent in high-passage BAECs and fibroblasts. Positive labelling for CD44, vimentin and cytokeratin was observed in both BAECs and fibroblasts. Regarding the lytic cycle of the parasite, although low invasion rates (approximately 3-4%) were found in both cell culture systems, more invasion was observed in BAECs at 24 and 72 hpi. The proliferation kinetics did not differ between BAECs and fibroblasts. BVDV infection favoured early Besnoitia invasion but there was no difference in tachyzoite yields observed in BVDV-BAECs compared to BAECs. CONCLUSIONS: We have generated and characterized two novel standardized in vitro models for Besnoitia besnoiti infection based on bovine primary target BAECs and fibroblasts, and have shown the relevance of BVDV coinfections, which should be considered in further studies with other cattle pathogens.


Asunto(s)
Enfermedades de los Bovinos/parasitología , Coccidiosis/veterinaria , Células Endoteliales/parasitología , Fibroblastos/parasitología , Sarcocystidae/crecimiento & desarrollo , Animales , Antígenos CD34/metabolismo , Bovinos , Coccidiosis/parasitología , Receptores de Hialuranos/metabolismo , Estadios del Ciclo de Vida , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Factores de Tiempo
19.
Ann Parasitol ; 65(3): 193-204, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31578843

RESUMEN

Natural transmission of Trypanosoma cruzi to human is established when feces of hematophagous triatomines contaminated with insect-derived from metacyclic trypomastigotes get in contact with the skin, conjunctiva or even oral route. Article is aimed at updating the knowledge about the early interaction between insect-derived metacyclic trypomastigotes at the port of entry and the host. There are few works in the literature describing this first contact between host and natural insect-derived metacyclic trypomastigote. Although it is currently accepted that T. cruzi parasites can penetrate through the lesion left by the insect´s bite, pioneer data do not support this hypothesis as the main via; however, once in the dermis metacyclic trypomastigotes can spread rapidly and likely escape from inoculation site through endothelial cells and disseminate to the body via the bloodstream. A moderate inflammatory reaction took place in the skin at the port of entry within hours, the cytokines induces recruit of neutrophils predominantly, probably because triatomine feces microbiota is present in the inoculum that in some way, its presence modify the progress of the infection.


Asunto(s)
Enfermedad de Chagas , Células Endoteliales , Estadios del Ciclo de Vida , Trypanosoma cruzi , Animales , Enfermedad de Chagas/patología , Células Endoteliales/parasitología , Humanos , Insectos/parasitología , Estadios del Ciclo de Vida/fisiología
20.
Sci Rep ; 9(1): 13142, 2019 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-31511575

RESUMEN

Recent concepts suggest that both Plasmodium falciparum factors and coagulation contribute to endothelial activation and dysfunction in pediatric cerebral malaria (CM) pathology. However, there is still limited understanding of how these complex inflammatory stimuli are integrated by brain endothelial cells. In this study, we examined how mature-stage P. falciparum infected erythrocytes (IE) interact with tumor necrosis factor α (TNFα) and thrombin in the activation and permeability of primary human brain microvascular endothelial cell (HBMEC) monolayers. Whereas trophozoite-stage P. falciparum-IE have limited effect on the viability of HBMEC or the secretion of pro-inflammatory cytokines or chemokines, except at super physiological parasite-host cell ratios, schizont-stage P. falciparum-IE induced low levels of cell death. Additionally, schizont-stage parasites were more barrier disruptive than trophozoite-stage P. falciparum-IE and prolonged thrombin-induced barrier disruption in both resting and TNFα-activated HBMEC monolayers. These results provide evidence that parasite products and thrombin may interact to increase brain endothelial permeability.


Asunto(s)
Encéfalo/metabolismo , Células Endoteliales/metabolismo , Eritrocitos/metabolismo , Plasmodium falciparum/metabolismo , Trombina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/parasitología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/parasitología , Eritrocitos/parasitología , Humanos , Malaria Cerebral/metabolismo , Malaria Cerebral/parasitología , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/fisiología , Proteínas Protozoarias/metabolismo , Esquizontes/metabolismo , Esquizontes/fisiología , Trombina/farmacología , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...