Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 153(5): 1120-33, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23683579

RESUMEN

Cell-cell communication is an important mechanism for information exchange promoting cell survival for the control of features such as population density and differentiation. We determined that Plasmodium falciparum-infected red blood cells directly communicate between parasites within a population using exosome-like vesicles that are capable of delivering genes. Importantly, communication via exosome-like vesicles promotes differentiation to sexual forms at a rate that suggests that signaling is involved. Furthermore, we have identified a P. falciparum protein, PfPTP2, that plays a key role in efficient communication. This study reveals a previously unidentified pathway of P. falciparum biology critical for survival in the host and transmission to mosquitoes. This identifies a pathway for the development of agents to block parasite transmission from the human host to the mosquito.


Asunto(s)
Comunicación Celular , Eritrocitos/patología , Eritrocitos/parasitología , Malaria Falciparum/patología , Malaria Falciparum/parasitología , Plasmodium falciparum/fisiología , Actinas/antagonistas & inhibidores , Animales , Culicidae/parasitología , Resistencia a Medicamentos , Exosomas/parasitología , Humanos , Microtúbulos/efectos de los fármacos , Plásmidos/genética , Plasmodium falciparum/crecimiento & desarrollo , Transducción de Señal , Trofozoítos/fisiología
2.
Traffic ; 25(4): e12935, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38629580

RESUMEN

The protozoan parasites Plasmodium falciparum, Leishmania spp. and Trypanosoma cruzi continue to exert a significant toll on the disease landscape of the human population in sub-Saharan Africa and Latin America. Control measures have helped reduce the burden of their respective diseases-malaria, leishmaniasis and Chagas disease-in endemic regions. However, the need for new drugs, innovative vaccination strategies and molecular markers of disease severity and outcomes has emerged because of developing antimicrobial drug resistance, comparatively inadequate or absent vaccines, and a lack of trustworthy markers of morbid outcomes. Extracellular vesicles (EVs) have been widely reported to play a role in the biology and pathogenicity of P. falciparum, Leishmania spp. and T. cruzi ever since they were discovered. EVs are secreted by a yet to be fully understood mechanism in protozoans into the extracellular milieu and carry a cargo of diverse molecules that reflect the originator cell's metabolic state. Although our understanding of the biogenesis and function of EVs continues to deepen, the question of how EVs in P. falciparum, Leishmania spp. and T. cruzi can serve as targets for a translational agenda into clinical and public health interventions is yet to be fully explored. Here, as a consortium of protozoan researchers, we outline a plan for future researchers and pose three questions to direct an EV's translational agenda in P. falciparum, Leishmania spp. and T. cruzi. We opine that in the long term, executing this blueprint will help bridge the current unmet needs of these medically important protozoan diseases in sub-Saharan Africa and Latin America.


Asunto(s)
Enfermedad de Chagas , Vesículas Extracelulares , Leishmania , Parásitos , Trypanosoma cruzi , Animales , Humanos , Enfermedad de Chagas/epidemiología , Enfermedad de Chagas/parasitología
3.
PLoS Pathog ; 19(2): e1011140, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36821560

RESUMEN

Extracellular vesicles (EVs) provide a central mechanism of cell-cell communication. While EVs are found in most organisms, their pathogenesis-promoting roles in parasites are of particular interest given the potential for medical insight and consequential therapeutic intervention. Yet, a key feature of EVs in human parasitic protozoa remains elusive: their mechanisms of biogenesis. Here, we survey the current knowledge on the biogenesis pathways of EVs secreted by the four main clades of human parasitic protozoa: apicomplexans, trypanosomatids, flagellates, and amoebae. In particular, we shine a light on findings pertaining to the Endosomal Sorting Complex Required for Transport (ESCRT) machinery, as in mammals it plays important roles in EV biogenesis. This review highlights the diversity in EV biogenesis in protozoa, as well as the related involvement of the ESCRT system in these unique organisms.


Asunto(s)
Vesículas Extracelulares , Parásitos , Animales , Humanos , Parásitos/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Vesículas Extracelulares/metabolismo , Transporte Biológico , Transporte de Proteínas , Mamíferos
4.
EMBO Rep ; 24(5): e56114, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-36929726

RESUMEN

Vesicular transport is a means of communication. While cells can communicate with each other via secretion of extracellular vesicles, less is known regarding organelle-to organelle communication, particularly in the case of mitochondria. Mitochondria are responsible for the production of energy and for essential metabolic pathways in the cell, as well as fundamental processes such as apoptosis and aging. Here, we show that functional mitochondria isolated from Saccharomyces cerevisiae release vesicles, independent of the fission machinery. We isolate these mitochondrial-derived vesicles (MDVs) and find that they are relatively uniform in size, of about 100 nm, and carry selective protein cargo enriched for ATP synthase subunits. Remarkably, we further find that these MDVs harbor a functional ATP synthase complex. We demonstrate that these vesicles have a membrane potential, produce ATP, and seem to fuse with naive mitochondria. Our findings reveal a possible delivery mechanism of ATP-producing vesicles, which can potentially regenerate ATP-deficient mitochondria and may participate in organelle-to-organelle communication.


Asunto(s)
Mitocondrias , Saccharomyces cerevisiae , Potenciales de la Membrana , Mitocondrias/metabolismo , Transporte Biológico , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Adenosina Trifosfato/metabolismo
5.
EMBO Rep ; 23(7): e54755, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35642585

RESUMEN

Malaria is the most serious mosquito-borne parasitic disease, caused mainly by the intracellular parasite Plasmodium falciparum. The parasite invades human red blood cells and releases extracellular vesicles (EVs) to alter its host responses. It becomes clear that EVs are generally composed of sub-populations. Seeking to identify EV subpopulations, we subject malaria-derived EVs to size-separation analysis, using asymmetric flow field-flow fractionation. Multi-technique analysis reveals surprising characteristics: we identify two distinct EV subpopulations differing in size and protein content. Small EVs are enriched in complement-system proteins and large EVs in proteasome subunits. We then measure the membrane fusion abilities of each subpopulation with three types of host cellular membranes: plasma, late and early endosome. Remarkably, small EVs fuse to early endosome liposomes at significantly greater levels than large EVs. Atomic force microscope imaging combined with machine-learning methods further emphasizes the difference in biophysical properties between the two subpopulations. These results shed light on the sophisticated mechanism by which malaria parasites utilize EV subpopulations as a communication tool to target different cellular destinations or host systems.


Asunto(s)
Vesículas Extracelulares , Malaria , Parásitos , Animales , Eritrocitos/parasitología , Vesículas Extracelulares/metabolismo , Humanos , Plasmodium falciparum
6.
Circulation ; 143(25): 2475-2493, 2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-33793321

RESUMEN

BACKGROUND: The role of epicardial fat (eFat)-derived extracellular vesicles (EVs) in the pathogenesis of atrial fibrillation (AF) has never been studied. We tested the hypothesis that eFat-EVs transmit proinflammatory, profibrotic, and proarrhythmic molecules that induce atrial myopathy and fibrillation. METHODS: We collected eFat specimens from patients with (n=32) and without AF (n=30) during elective heart surgery. eFat samples were grown as organ cultures, and the culture medium was collected every 2 days. We then isolated and purified eFat-EVs from the culture medium, and analyzed the EV number, size, morphology, specific markers, encapsulated cytokines, proteome, and microRNAs. Next, we evaluated the biological effects of unpurified and purified EVs on atrial mesenchymal stromal cells and endothelial cells in vitro. To establish a causal association between eFat-EVs and vulnerability to AF, we modeled AF in vitro using induced pluripotent stem cell-derived cardiomyocytes. RESULTS: Microscopic examination revealed excessive inflammation, fibrosis, and apoptosis in fresh and cultured eFat tissues. Cultured explants from patients with AF secreted more EVs and harbored greater amounts of proinflammatory and profibrotic cytokines, and profibrotic microRNA, as well, than those without AF. The proteomic analysis confirmed the distinctive profile of purified eFat-EVs from patients with AF. In vitro, purified and unpurified eFat-EVs from patients with AF had a greater effect on proliferation and migration of human mesenchymal stromal cells and endothelial cells, compared with eFat-EVs from patients without AF. Last, whereas eFat-EVs from patients with and without AF shortened the action potential duration of induced pluripotent stem cell-derived cardiomyocytes, only eFat-EVs from patients with AF induced sustained reentry (rotor) in induced pluripotent stem cell-derived cardiomyocytes. CONCLUSIONS: We show, for the first time, a distinctive proinflammatory, profibrotic, and proarrhythmic signature of eFat-EVs from patients with AF. Our findings uncover another pathway by which eFat promotes the development of atrial myopathy and fibrillation.


Asunto(s)
Tejido Adiposo/patología , Fibrilación Atrial/etiología , Fibrilación Atrial/patología , Vesículas Extracelulares/patología , Miocitos Cardíacos/patología , Pericardio/patología , Tejido Adiposo/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Fibrilación Atrial/metabolismo , Células Cultivadas , Vesículas Extracelulares/metabolismo , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/metabolismo , Técnicas de Cultivo de Órganos , Pericardio/metabolismo , Proteómica/métodos , Ratas
7.
Faraday Discuss ; 240(0): 127-141, 2022 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-35938388

RESUMEN

Malaria is a potentially fatal infectious disease caused by the obligate intracellular parasite Plasmodium falciparum. The parasite infects human red blood cells (RBC) and derives nutrition by catabolism of hemoglobin. As amino acids are assimilated from the protein component, the toxic heme is released. Molecular heme is detoxified by rapid sequestration to physiologically insoluble hemozoin crystals within the parasite's digestive vacuole (DV). Common antimalarial drugs interfere with this crystallization process, leaving the parasites vulnerable to the by-product of their own metabolism. A fundamental debate with important implications on drug mechanism regards the chemical environment of crystallization in situ, whether aqueous or lipid. This issue had been addressed previously by cryogenic soft X-ray tomography. We employ cryo-scanning transmission electron tomography (CSTET) to probe parasite cells throughout the life cycle in a fully hydrated, vitrified state at higher resolution. During the acquisition of CSTET data, Bragg diffraction from the hemozoin provides a uniquely clear view of the crystal boundary at nanometer resolution. No intermediate medium, such as a lipid coating or shroud, could be detected surrounding the crystals. The present study describes a unique application of CSTET in the study of malaria. The findings can be extended to evaluate new drug candidates affecting hemozoin crystal growth.


Asunto(s)
Tomografía con Microscopio Electrónico , Malaria , Humanos , Hemo/química , Hemo/metabolismo , Malaria/parasitología , Lípidos/química
8.
EMBO Rep ; 21(1): e47882, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31825165

RESUMEN

During the chronic stage of Schistosoma infection, the female lays fertile eggs, triggering a strong anti-parasitic type 2 helper T-cell (Th2) immune response. It is unclear how this Th2 response gradually declines even though the worms live for years and continue to produce eggs. Here, we show that Schistosoma mansoni downregulates Th2 differentiation in an antigen-presenting cell-independent manner, by modulating the Th2-specific transcriptional program. Adult schistosomes secrete miRNA-harboring extracellular vesicles that are internalized by Th cells in vitro. Schistosomal miRNAs are found also in T helper cells isolated from Peyer's patches and mesenteric lymph nodes of infected mice. In T helper cells, the schistosomal miR-10 targets MAP3K7 and consequently downmodulates NF-κB activity, a critical transcription factor for Th2 differentiation and function. Our results explain, at least partially, how schistosomes tune down the Th2 response, and provide further insight into the reciprocal geographic distribution between high prevalence of parasitic infections and immune disorders such as allergy. Furthermore, this worm-host crosstalk mechanism can be harnessed to develop diagnostic and therapeutic approaches for human schistosomiasis and Th2-associated diseases.


Asunto(s)
Vesículas Extracelulares , MicroARNs , Animales , Diferenciación Celular , Femenino , Ratones , MicroARNs/genética , Schistosoma mansoni/genética , Células Th2
9.
Proteins ; 88(1): 187-195, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31325330

RESUMEN

Many human pathogens use host cell-surface receptors to attach and invade cells. Often, the host-pathogen interaction affinity is low, presenting opportunities to block invasion using a soluble, high-affinity mimic of the host protein. The Plasmodium falciparum reticulocyte-binding protein homolog 5 (RH5) provides an exciting candidate for mimicry: it is highly conserved and its moderate affinity binding to the human receptor basigin (KD ≥1 µM) is an essential step in erythrocyte invasion by this malaria parasite. We used deep mutational scanning of a soluble fragment of human basigin to systematically characterize point mutations that enhance basigin affinity for RH5 and then used Rosetta to design a variant within the sequence space of affinity-enhancing mutations. The resulting seven-mutation design exhibited 1900-fold higher affinity (KD approximately 1 nM) for RH5 with a very slow binding off rate (0.23 h-1 ) and reduced the effective Plasmodium growth-inhibitory concentration by at least 10-fold compared to human basigin. The design provides a favorable starting point for engineering on-rate improvements that are likely to be essential to reach therapeutically effective growth inhibition.


Asunto(s)
Basigina/farmacología , Proteínas Portadoras/genética , Malaria Falciparum/tratamiento farmacológico , Eritrocitos/efectos de los fármacos , Humanos , Malaria Falciparum/genética , Malaria Falciparum/parasitología , Modelos Moleculares , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/patogenicidad , Unión Proteica/efectos de los fármacos , Proteínas Protozoarias/genética
10.
Cell Microbiol ; 21(7): e13021, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30835870

RESUMEN

Protozoan pathogens secrete nanosized particles called extracellular vesicles (EVs) to facilitate their survival and chronic infection. Here, we show the inhibition by Plasmodium berghei NK65 blood stage-derived EVs of the proliferative response of CD4+ T cells in response to antigen presentation. Importantly, these results were confirmed in vivo by the capacity of EVs to diminish the ovalbumin-specific delayed type hypersensitivity response. We identified two proteins associated with EVs, the histamine releasing factor (HRF) and the elongation factor 1α (EF-1α) that were found to have immunosuppressive activities. Interestingly, in contrast to WT parasites, EVs from genetically HRF- and EF-1α-deficient parasites failed to inhibit T cell responses in vitro and in vivo. At the level of T cells, we demonstrated that EVs from WT parasites dephosphorylate key molecules (PLCγ1, Akt, and ERK) of the T cell receptor signalling cascade. Remarkably, immunisation with EF-1α alone or in combination with HRF conferred a long-lasting antiparasite protection and immune memory. In conclusion, we identified a new mechanism by which P. berghei-derived EVs exert their immunosuppressive functions by altering T cell responses. The identification of two highly conserved immune suppressive factors offers new conceptual strategies to overcome EV-mediated immune suppression in malaria-infected individuals.


Asunto(s)
Biomarcadores de Tumor/genética , Vesículas Extracelulares/inmunología , Malaria/genética , Factor 1 de Elongación Peptídica/genética , Animales , Presentación de Antígeno/inmunología , Antígenos/genética , Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/parasitología , Proliferación Celular/genética , Vesículas Extracelulares/genética , Humanos , Evasión Inmune/genética , Evasión Inmune/inmunología , Malaria/parasitología , Malaria/patología , Plasmodium berghei/genética , Plasmodium berghei/patogenicidad , Linfocitos T/inmunología , Linfocitos T/parasitología , Proteína Tumoral Controlada Traslacionalmente 1
11.
PLoS Biol ; 15(6): e2002711, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28650960

RESUMEN

Necroptosis is a regulated, nonapoptotic form of cell death initiated by receptor-interacting protein kinase-3 (RIPK3) and mixed lineage kinase domain-like (MLKL) proteins. It is considered to be a form of regulated necrosis, and, by lacking the "find me" and "eat me" signals that are a feature of apoptosis, necroptosis is considered to be inflammatory. One such "eat me" signal observed during apoptosis is the exposure of phosphatidylserine (PS) on the outer plasma membrane. Here, we demonstrate that necroptotic cells also expose PS after phosphorylated mixed lineage kinase-like (pMLKL) translocation to the membrane. Necroptotic cells that expose PS release extracellular vesicles containing proteins and pMLKL to their surroundings. Furthermore, inhibition of pMLKL after PS exposure can reverse the process of necroptosis and restore cell viability. Finally, externalization of PS by necroptotic cells drives recognition and phagocytosis, and this may limit the inflammatory response to this nonapoptotic form of cell death. The exposure of PS to the outer membrane and to extracellular vesicles is therefore a feature of necroptotic cell death and may serve to provide an immunologically-silent window by generating specific "find me" and "eat me" signals.


Asunto(s)
Membrana Celular/metabolismo , Necrosis/metabolismo , Fagocitosis , Fosfatidilserinas/metabolismo , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/inmunología , Membrana Celular/ultraestructura , Permeabilidad de la Membrana Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Vesículas Extracelulares/efectos de los fármacos , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/ultraestructura , Humanos , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Necrosis/inmunología , Necrosis/patología , Necrosis/prevención & control , Fagocitosis/efectos de los fármacos , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/química , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Propiedades de Superficie/efectos de los fármacos
12.
Proteomics ; 19(1-2): e1800170, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30358122

RESUMEN

Genetic plasticity of prokaryotic microbial communities is largely dependent on the ongoing exchange of genetic determinants by Horizontal Gene Transfer (HGT). HGT events allow beneficial genetic transitions to occur throughout microbial life, thus promoting adaptation to changing environmental conditions. Here, the significance of secreted vesicles in mediating HGT between microorganisms is discussed, while focusing on the benefits gained by vesicle-mediated gene delivery and its occurrence under different environmental cues. The potential use of secreted DNA-harboring vesicles as a mechanism of currently unresolved HGT events in eukaryotic microbes is further discussed.


Asunto(s)
Vesículas Extracelulares/metabolismo , Transferencia de Gen Horizontal/fisiología , Evolución Molecular , Transferencia de Gen Horizontal/genética
13.
Semin Cell Dev Biol ; 67: 83-90, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28366828

RESUMEN

Infectious diseases are the leading cause of death of children worldwide, causing a tenacious and major public-health burden. The dynamic interplay between pathogens and their host is one of the most complicated themes of the disease progression. Pathogens excel in developing different means to facilitate cell-cell communication via secreted vesicles, among others. The released vesicles are involved in the transfer of biologically active molecules that induce phenotypic changes in the recipient cells. The messages within the vesicles are delivered to coordinate diverse processes, including virulence factor expression, differentiation state and control of their population density. Importantly, production of such vesicles promotes pathogen survival, as it provides a secure means of pathogen-pathogen communication and an ability to manipulate host responses for their own benefits. This review highlights intriguing findings, which show the important role of EVs in the social activity of pathogens, within and in between their communities. We further present examples of how pathogens use EVs to alter host immune and non-immune responses. Advancing our understanding of cell-cell communication in infectious diseases will be particularly useful to decipher the complexity of the cross-talk between pathogens themselves and their hosts, leading to the development of therapeutic strategies for fighting infectious agents.


Asunto(s)
Bacterias/metabolismo , Enfermedades Transmisibles/inmunología , Vesículas Extracelulares/metabolismo , Hongos/metabolismo , Interacciones Huésped-Patógeno/inmunología , Interacciones Microbianas/fisiología , Virus/metabolismo , Bacterias/genética , Bacterias/crecimiento & desarrollo , Bacterias/patogenicidad , Biopelículas/crecimiento & desarrollo , Enfermedades Transmisibles/microbiología , Enfermedades Transmisibles/virología , Hongos/genética , Hongos/crecimiento & desarrollo , Hongos/patogenicidad , Transferencia de Gen Horizontal , Humanos , Inmunidad Innata , Activación de Linfocitos , Linfocitos/inmunología , Linfocitos/microbiología , Linfocitos/virología , Percepción de Quorum/fisiología , Conducta Social , Virulencia , Virus/genética , Virus/crecimiento & desarrollo , Virus/patogenicidad
14.
PLoS Pathog ; 13(8): e1006524, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28837697

RESUMEN

Metabolic changes within the cell and its niche affect cell fate and are involved in many diseases and disorders including cancer and viral infections. Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of Kaposi's sarcoma (KS). KSHV latently infected cells express only a subset of viral genes, mainly located within the latency-associated region, among them 12 microRNAs. Notably, these miRNAs are responsible for inducing the Warburg effect in infected cells. Here we identify a novel mechanism enabling KSHV to manipulate the metabolic nature of the tumour microenvironment. We demonstrate that KSHV infected cells specifically transfer the virus-encoded microRNAs to surrounding cells via exosomes. This flow of genetic information results in a metabolic shift toward aerobic glycolysis in the surrounding non-infected cells. Importantly, this exosome-mediated metabolic reprogramming of neighbouring cells supports the growth of infected cells, thereby contributing to viral fitness. Finally, our data show that this miRNA transfer-based regulation of cell metabolism is a general mechanism used by other herpesviruses, such as EBV, as well as for the transfer of non-viral onco-miRs. This exosome-based crosstalk provides viruses with a mechanism for non-infectious transfer of genetic material without production of new viral particles, which might expose them to the immune system. We suggest that viruses and cancer cells use this mechanism to shape a specific metabolic niche that will contribute to their fitness.


Asunto(s)
Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/patogenicidad , Sarcoma de Kaposi/virología , Microambiente Tumoral/fisiología , Virulencia/fisiología , Western Blotting , Línea Celular , Exoma/fisiología , Infecciones por Herpesviridae/metabolismo , Herpesvirus Humano 8/fisiología , Humanos , MicroARNs/genética , Microscopía Electrónica de Transmisión , Reacción en Cadena de la Polimerasa , ARN Viral/genética , Sarcoma de Kaposi/metabolismo
15.
Cell Microbiol ; 20(5): e12822, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29349926

RESUMEN

Pathogens can release extracellular vesicles (EVs) for cell-cell communication and host modulation. EVs from Plasmodium falciparum, the deadliest malaria parasite species, can transfer drug resistance genes between parasites. EVs from late-stage parasite-infected RBC (iRBC-EVs) are immunostimulatory and affect endothelial cell permeability, but little is known about EVs from early stage iRBC. We detected the parasite virulence factor PfEMP1, which is responsible for iRBC adherence and a major contributor to disease severity, in EVs, only up to 12-hr post-RBC invasion. Furthermore, using PfEMP1 transport knockout parasites, we determined that EVs originated from inside the iRBC rather than the iRBC surface. Proteomic analysis detected 101 parasite and 178 human proteins in iRBC-EVs. Primary human monocytes stimulated with iRBC-EVs released low levels of inflammatory cytokines and showed transcriptomic changes. Stimulation with iRBC-EVs from PfEMP1 knockout parasites induced more gene expression changes and affected pathways involved in defence response, stress response, and response to cytokines, suggesting a novel function of PfEMP1 when present in EVs. We show for the first time the presence of PfEMP1 in early stage P. falciparum iRBC-EVs and the effects of these EVs on primary human monocytes, uncovering a new mechanism of potential parasite pathogenesis and host interaction.


Asunto(s)
Malaria Falciparum/genética , Plasmodium falciparum/genética , Proteómica , Proteínas Protozoarias/genética , Animales , Adhesión Celular/genética , Comunicación Celular/genética , Células Endoteliales/metabolismo , Células Endoteliales/patología , Eritrocitos/parasitología , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Interacciones Huésped-Parásitos/genética , Humanos , Malaria Falciparum/sangre , Malaria Falciparum/parasitología , Monocitos/metabolismo , Monocitos/parasitología , Plasmodium falciparum/patogenicidad
16.
Small ; 14(39): e1801650, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30160371

RESUMEN

Extracellular vesicles (EVs) are emerging as important mediators of cell-cell communication as well as potential disease biomarkers and drug delivery vehicles. However, the mechanical properties of these vesicles are largely unknown, and processes leading to microvesicle-shedding from the plasma membrane are not well understood. Here an in depth atomic force microscopy force spectroscopy study of the mechanical properties of natural EVs is presented. It is found that several natural vesicles of different origin have a different composition of lipids and proteins, but similar mechanical properties. However, vesicles generated by red blood cells (RBC) at different temperatures/incubation times are different mechanically. Quantifying the lipid content of EVs reveals that their stiffness decreases with the increase in their protein/lipid ratio. Further, by maintaining RBC at "extreme" nonphysiological conditions, the cells are pushed to utilize different vesicle generation pathways. It is found that RBCs can generate protein-rich soft vesicles, possibly driven by protein aggregation, and low membrane-protein content stiff vesicles, likely driven by cytoskeleton-induced buckling. Since similar cortical cytoskeleton to that of the RBC exists on the membranes of most mammalian cells, our findings help advancing the understanding of the fundamental process of vesicle generation.


Asunto(s)
Vesículas Extracelulares/metabolismo , Animales , Biofisica , Membrana Eritrocítica/metabolismo , Eritrocitos/metabolismo , Humanos , Microscopía de Fuerza Atómica
17.
Methods ; 112: 157-166, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27350362

RESUMEN

Malaria is the most devastating parasitic disease of humans, caused by the unicellular protozoa of the Plasmodium genus, such as Plasmodium falciparum (Pf) and is responsible for up to a million deaths each year. Pf life cycle is complex, with transmission of the parasite between humans via mosquitos involving a remarkable series of morphological transformations. In the bloodstream, the parasites undergo asexual multiplications inside the red blood cell (RBC), where they mature through the ring (R), trophozoite (T) and schizont (S) stages, and sexual development, resulting in gametocytes (G). All symptoms of malaria pathology are caused by the asexual blood stage parasites. Flow cytometry methods were previously used to detect malaria infected (i) RBCs, in live or fixed cells, using DNA (Hoechst) and RNA (Thiazole Orange) stains. Here, by using imaging flow cytometry, we developed improved methods of identifying and quantifying each of the four parasite blood stages (R, T, S and G). This technique allows multi-channel, high resolution imaging of individual parasites, as well as detailed morphological quantification of Pf-iRBCs cultures. Moreover, by measuring iRBC morphological properties, we can eliminate corrupted and extracellular (dying) parasites from the analysis, providing accurate quantification and robust measurement of the parasitemia profile. This new method is a valuable tool in malaria molecular biology research and drug screen assays.


Asunto(s)
Eritrocitos/parasitología , Citometría de Flujo/métodos , Citometría de Imagen/métodos , Estadios del Ciclo de Vida/fisiología , Plasmodium falciparum/crecimiento & desarrollo , Coloración y Etiquetado/métodos , Colorantes Azulados/química , Bencimidazoles/química , Benzotiazoles/química , Células Cultivadas , Eritrocitos/ultraestructura , Colorantes Fluorescentes/química , Humanos , Plasmodium falciparum/ultraestructura , Quinolinas/química
18.
J Infect Dis ; 215(3): 378-386, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-28362903

RESUMEN

BACKGROUND: Schistosomiasis traditionally has been diagnosed by detecting eggs in stool or urine. However, the sensitivity of these examinations is limited, especially in travelers with a low worm burden. Serologic tests have a greater sensitivity, but their results remain positive regardless of treatment and thus cannot be used for follow-up of patients. We hypothesized that detection of worm microRNAs (miRNAs) in serum can overcome the drawbacks of the existing diagnostic methods. METHODS AND RESULTS: Twenty-six returning travelers with schistosomiasis (based on positive results of serologic tests or detection of ova) and 17 healthy controls were included in the study. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) amplification of miRNA extracted directly from 500 µL of serum had limited sensitivity and specificity. However, qRT-PCR analysis of RNA extracted from 200 µL of serum extracellular vesicles detected 4 schistosomal miRNAs; the sensitivity and specificity of the 2 highest expressed miRNAs (bantam and miR-2c-3p) were 86% and 84%, respectively. In 7 patients with posttreatment serum available for analysis, we observed outcomes ranging from a reduction in the schistosomal miRNA level to full recovery from disease. CONCLUSIONS: qRT-PCR of pathogen miRNAs isolated from extracellular vesicles in sera from infected individuals may provide a new tool for diagnosing schistosomiasis in patients with a low parasite burden. This assay could also be used for evaluating the outcome of therapy, as well as disease-control programs.


Asunto(s)
Vesículas Extracelulares/parasitología , MicroARNs/sangre , ARN de Helminto/sangre , Schistosoma mansoni/genética , Esquistosomiasis/diagnóstico , Adulto , Animales , Femenino , Estudios de Seguimiento , Humanos , Masculino , MicroARNs/aislamiento & purificación , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa , Schistosoma mansoni/aislamiento & purificación , Esquistosomiasis/sangre , Esquistosomiasis/parasitología , Adulto Joven
20.
Subcell Biochem ; 78: 201-17, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25487023

RESUMEN

Mitochondrial chaperones mediate and affect critical organellar processes, essential for cellular function. These chaperone systems have both prokaryotic and eukaryotic features. While some of the mitochondrial co-chaperones have clear homologues in prokaryotes, some are unique to eukaryotes and have no homologues in the chaperone machinery of other cellular compartments. The mitochondrial co-chaperones are required for protein import into the organelle and in enforcing the structure of the main chaperones. In addition to novel types of interaction with their senior partners, unexpected and essential interactions between the co-chaperones themselves have recently been described.


Asunto(s)
Evolución Molecular , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Chaperonas Moleculares/metabolismo , Animales , Humanos , Proteínas Mitocondriales/química , Chaperonas Moleculares/química , Conformación Proteica , Transporte de Proteínas , Transducción de Señal , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA