Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
Eur J Appl Physiol ; 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38801445

RESUMEN

Independently, both prolonged uninterrupted sitting and the onset of menopause negatively impact markers of cardiovascular risk. Whether their combination augment these responses additively remains unknown. This study assessed whether prolonged uninterrupted sitting causes greater central and peripheral cardiovascular dysfunction in post-menopausal women compared to pre-menopausal women. To address this, 23 healthy women (13 pre-menopausal [43.77 ± 4.30 years] and 10 post-menopausal [57.20 ± 8.55 years]) sat uninterrupted for 2-h. Carotid-femoral pulse wave velocity (cf-PWV), pulse wave analysis (PWA), lower limb venous pooling (HHb), and calf circumference were assessed pre-and post-sitting using general linear mixed models, with age as a covariate. Changes in MAP over time (both between and within groups) was assessed using a two-way repeated-measures-ANOVA. There were no significant interactions for any outcome measures. However, for cf-PWV, there was a significant main effect of group (Δ = 0.854 ± 0.354 m s-1; p = 0.026, ηp2 = 0.707). For PWA, only heart rate (HR) and pressure forwards (Pf) showed significant main effects 13 of time [Δ = 6 ± 1 bts-min-1, p < 0.001, ηp2 = 0.861] and group [Δ = 3.893 ± 1.450 mmHg, p = 0.016, ηp2 = 0.271], respectively. Both HHb (Δ = 2.737 ± 0.952, p = 0.009, ηp2 = 0.742) and calf circumference (Δ = 0.812 ± 0.128 cm, p < 0.001, ηp2 = 0.863) significantly increased over time. Whilst post-menopausal women demonstrated greater overall arterial stiffness (increased cf-PWV at baseline), there was no difference in cardiovascular response (central or peripheral) to 2-h of prolonged sitting between the pre- and post-menopausal women.

2.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34035177

RESUMEN

Cytoadhesion of Plasmodium falciparum-infected erythrocytes (IEs) to the endothelial lining of blood vessels protects parasites from splenic destruction, but also leads to detrimental inflammation and vessel occlusion. Surface display of the P. falciparum erythrocyte membrane protein 1 (PfEMP1) adhesion ligands exposes them to host antibodies and serum proteins. PfEMP1 are important targets of acquired immunity to malaria, and through evolution, the protein family has expanded and diversified to bind a select set of host receptors through antigenically diversified receptor-binding domains. Here, we show that complement component 1s (C1s) in serum cleaves PfEMP1 at semiconserved arginine motifs located at interdomain regions between the receptor-binding domains, rendering the IE incapable of binding the two main PfEMP1 receptors, CD36 and endothelial protein C receptor (EPCR). Bioinformatic analyses of PfEMP1 protein sequences from 15 P. falciparum genomes found the C1s motif was present in most PfEMP1 variants. Prediction of C1s cleavage and loss of binding to endothelial receptors was further corroborated by testing of several different parasite lines. These observations suggest that the parasites have maintained susceptibility for cleavage by the serine protease, C1s, and provides evidence for a complex relationship between the complement system and the P. falciparum cytoadhesion virulence determinant.


Asunto(s)
Adhesión Bacteriana , Complemento C1/metabolismo , Plasmodium falciparum/fisiología , Proteínas Protozoarias/metabolismo , Secuencia de Aminoácidos , Línea Celular , Secuencia Conservada , Humanos
3.
PLoS Pathog ; 17(6): e1009659, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34115805

RESUMEN

Sickle-trait hemoglobin protects against severe Plasmodium falciparum malaria. Severe malaria is governed in part by the expression of the Plasmodium falciparum Erythrocyte Membrane Protein 1 (PfEMP1) that are encoded by var genes, specifically those variants that bind Endothelial Protein C Receptor (EPCR). In this study, we investigate the effect of sickle-trait on parasite var gene expression and function in vitro and in field-collected parasites. We mapped var gene reads generated from RNA sequencing in parasite cultures in normal and sickle-cell trait blood throughout the asexual lifecycle. We investigated sickle-trait effect on PfEMP1 interactions with host receptors CD36 and EPCR using static adhesion assays and flow cytometry. Var expression in vivo was compared by assembling var domains sequenced from total RNA in parasites infecting Malian children with HbAA and HbAS. Sickle-trait did not alter the abundance or type of var gene transcripts in vitro, nor the abundance of overall transcripts or of var functional domains in vivo. In adhesion assays using recombinant host receptors, sickle-trait reduced adhesion by 73-86% to CD36 and 83% to EPCR. Similarly, sickle-trait reduced the surface expression of EPCR-binding PfEMP1. In conclusion, Sickle-cell trait does not directly affect var gene transcription but does reduce the surface expression and function of PfEMP1. This provides a direct mechanism for protection against severe malaria conferred by sickle-trait hemoglobin. Trial Registration: ClinicalTrials.gov Identifier: NCT02645604.


Asunto(s)
Hemoglobina Falciforme/metabolismo , Malaria Falciparum/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Antígenos CD36/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Eritrocitos/parasitología , Hemoglobina Falciforme/genética , Humanos , Malaria Falciparum/metabolismo , Rasgo Drepanocítico/genética , Rasgo Drepanocítico/metabolismo
4.
Int J Mol Sci ; 24(12)2023 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-37373123

RESUMEN

Expression of human endogenous retrovirus type W (HERV-W) has been linked to cancer, making HERV-W antigens potential targets for therapeutic cancer vaccines. In a previous study, we effectively treated established tumours in mice by using adenoviral-vectored vaccines targeting the murine endogenous retrovirus envelope and group-specific antigen (Gag) of melanoma-associated retrovirus (MelARV) in combination with anti-PD-1. To break the immunological tolerance to MelARV, we mutated the immunosuppressive domain (ISD) of the MelARV envelope. However, reports on the immunogenicity of the HERV-W envelope, Syncytin-1, and its ISD are conflicting. To identify the most effective HERV-W cancer vaccine candidate, we evaluated the immunogenicity of vaccines encoding either the wild-type or mutated HERV-W envelope ISD in vitro and in vivo. Here, we show that the wild-type HERV-W vaccine generated higher activation of murine antigen-presenting cells and higher specific T-cell responses than the ISD-mutated counterpart. We also found that the wild-type HERV-W vaccine was sufficient to increase the probability of survival in mice subjected to HERV-W envelope-expressing tumours compared to a control vaccine. These findings provide the foundation for developing a therapeutic cancer vaccine targeting HERV-W-positive cancers in humans.


Asunto(s)
Vacunas contra el Cáncer , Retrovirus Endógenos , Neoplasias , Humanos , Animales , Ratones , Retrovirus Endógenos/genética , Linfocitos T , Terapia de Inmunosupresión
5.
BMC Infect Dis ; 22(1): 846, 2022 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-36371172

RESUMEN

BACKGROUND: African countries stand out globally as the region seemingly least affected by the COVID-19 pandemic, caused by the virus SARS-CoV-2. Besides a younger population and potential pre-existing immunity to a SARS-CoV-2-like virus, it has been hypothesized that co-infection or recent history of Plasmodium falciparum malaria may be protective of COVID-19 severity and mortality. The number of COVID-19 cases and deaths, however, may be vastly undercounted. Very little is known about the extent to which the Tanzanian population has been exposed to SARS-CoV-2. Here, we investigated the seroprevalence of IgG to SARS-CoV-2 spike protein in two Tanzanian rural communities 1½ years into the pandemic and the association of coinciding malaria infection and exposure. METHODS: During a malariometric survey in July 2021 in two villages in north-eastern Tanzania, blood samples were taken from 501 participants (0-19 years old). Malaria was detected by mRDT and microscopy. Levels of IgG against the spike protein of SARS-CoV-2 were measured by ELISA as well as IgG against five different antigens of P. falciparum; CIDRα1.1, CIDRα1.4 and CIDRα1.5 of PfEMP1 and GLURP and MSP3. RESULTS: The seroprevalence of SARS-CoV-2 IgG was 39.7% (106/267) in Kwamasimba and 32.5% (76/234) in Mkokola. In both villages the odds of being seropositive increased significantly with age (AOR = 1.12, 95% CI 1.07-1.17, p < 0.001). P. falciparum malaria prevalence by blood smear microscopy was 7.9% in Kwamasimba and 2.1% in Mkokola. 81.3% and 70.5% in Kwamasimba and Mkokola, respectively, showed recognition of minimum one malaria antigen. Residing in Kwamasimba was associated with a broader recognition (AOR = 1.91, 95% CI 1.34-2.71, p < 0.001). The recognition of malaria antigens increased significantly with age in both villages (AOR = 1.12; 95% CI 1.08-1.16, p < 0.001). Being SARS-CoV-2 seropositive did not associate with the breadth of malaria antigen recognition when adjusting for age (AOR = 0.99; 95% CI 0.83-1.18; p = 0.91). CONCLUSION: More than a third of the children and adolescents in two rural communities in Tanzania had antibodies to SARS-CoV-2. In particular, the adolescents were seropositive but being seropositive did not associate with the status of coinciding malaria infections or previous exposure. In Tanzania, natural immunity may have developed fast, potentially protecting a substantial part of the population from later variants.


Asunto(s)
Anticuerpos Antivirales , COVID-19 , Malaria Falciparum , Adolescente , Niño , Preescolar , Humanos , Lactante , Recién Nacido , Adulto Joven , Anticuerpos Antivirales/sangre , Antígenos de Protozoos , COVID-19/epidemiología , Inmunoglobulina G , Malaria Falciparum/epidemiología , Pandemias , SARS-CoV-2 , Estudios Seroepidemiológicos , Tanzanía/epidemiología
6.
Dermatol Surg ; 48(10): 1076-1082, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35985005

RESUMEN

BACKGROUND: Aesthetic muscle stimulation (AMS) using high-intensity electromagnetic field (HIFEM) targets skeletal muscle neurons, causing muscle hypertrophy and loss of adipose tissue, thereby cultivating a sculpted physique. Many studies have evaluated AMS for noninvasive body contouring; however, the efficacy, safety, and long-term data remain unclear. OBJECTIVE: To critically evaluate the current literature on the use of electromagnetic muscle stimulation for body contouring and provide a consensus on patient selection and long-term efficacy of AMS. MATERIALS AND METHODS: PubMed and Embase were searched using the terms: "HIFEM," "Electromagnetic therapy," and "muscle" or "Electrical stimulation muscle treatments" and "aesthetics." Studies involving the use of muscle stimulation for nonaesthetic/dermatologic, in vitro studies or studies involving animals were excluded. RESULTS: Twenty studies in total were included [9 moderate-quality, 8 low-quality, and 3 very low‒quality studies] based on the Grading of Recommendations, Assessment, Development, and Evaluation scale, representing 521 patients. Body sites evaluated included the abdomen (378 patients), buttock (156 patients), arms (22 patients), and calves (15 patients). CONCLUSION: Electromagnetic muscle stimulation represents an effective therapeutic intervention for abdominal contouring that yields increased muscle thickness, and reduced abdominal fat thickness, for up to 1 year after treatment. Larger, controlled studies are needed to determine the efficacy of electromagnetic muscle stimulation alone for contouring of buttocks, thighs, arms, and calves.


Asunto(s)
Contorneado Corporal , Magnetoterapia , Animales , Nalgas/cirugía , Campos Electromagnéticos , Estética
7.
PLoS Pathog ; 15(7): e1007906, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31295334

RESUMEN

The pathogenesis of Plasmodium falciparum malaria is linked to the variant surface antigen PfEMP1, which mediates tethering of infected erythrocytes to the host endothelium and is encoded by approximately 60 var genes per parasite genome. Repeated episodes of malaria infection result in the gradual acquisition of protective antibodies against PfEMP1 variants. The antibody repertoire is believed to provide a selective pressure driving the clonal expansion of parasites expressing unrecognized PfEMP1 variants, however, due to the lack of experimental in vivo models there is only limited experimental evidence in support of this concept. To get insight into the impact of naturally acquired immunity on the expressed var gene repertoire early during infection we performed controlled human malaria infections of 20 adult African volunteers with life-long malaria exposure using aseptic, purified, cryopreserved P. falciparum sporozoites (Sanaria PfSPZ Challenge) and correlated serological data with var gene expression patterns from ex vivo parasites. Among the 10 African volunteers who developed patent infections, individuals with low antibody levels showed a steep rise in parasitemia accompanied by broad activation of multiple, predominantly subtelomeric var genes, similar to what we previously observed in naïve volunteers. In contrast, individuals with intermediate antibody levels developed asymptomatic infections and the ex vivo parasite populations expressed only few var gene variants, indicative of clonal selection. Importantly, in contrast to parasites from naïve volunteers, expression of var genes coding for endothelial protein C receptor (EPCR)-binding PfEMP1 that are associated with severe childhood malaria was rarely detected in semi-immune adult African volunteers. Moreover, we followed var gene expression for up to six parasite replication cycles and demonstrated for the first time in vivo a shift in the dominant var gene variant. In conclusion, our data suggest that P. falciparum activates multiple subtelomeric var genes at the onset of blood stage infection facilitating rapid expansion of parasite clones which express PfEMP1 variants unrecognized by the host's immune system, thus promoting overall parasite survival in the face of host immunity.


Asunto(s)
Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Plasmodium falciparum/patogenicidad , Adolescente , Adulto , Animales , Anticuerpos Antiprotozoarios/sangre , Femenino , Regulación de la Expresión Génica , Genes Protozoarios , Humanos , Inmunidad Innata , Masculino , Plasmodium falciparum/genética , Plasmodium falciparum/inmunología , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Virulencia/genética , Virulencia/inmunología , Adulto Joven
8.
J Immunol ; 202(8): 2320-2331, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30833346

RESUMEN

Adenoviral vectors can induce T and B cell immune responses to Ags encoded in the recombinant vector. The MHC class II invariant chain (Ii) has been used as an adjuvant to enhance T cell responses to tethered Ag encoded in adenoviral vectors. In this study, we modified the Ii adjuvant by insertion of a furin recognition site (Ii-fur) to obtain a secreted version of the Ii. To test the capacity of this adjuvant to enhance immune responses, we recombined vectors to encode Plasmodium falciparum virulence factors: two cysteine-rich interdomain regions (CIDR) α1 (IT4var19 and PFCLINvar30 var genes), expressed as a dimeric Ag. These domains are members of a highly polymorphic protein family involved in the vascular sequestration and immune evasion of parasites in malaria. The Ii-fur molecule directed secretion of both Ags in African green monkey cells and functioned as an adjuvant for MHC class I and II presentation in T cell hybridomas. In mice, the Ii-fur adjuvant induced a similar T cell response, as previously demonstrated with Ii, accelerated and enhanced the specific Ab response against both CIDR Ags, with an increased binding capacity to the cognate endothelial protein C receptor, and enhanced the breadth of the response toward different CIDRs. We also demonstrate that the endosomal sorting signal, secretion, and the C-terminal part of Ii were needed for the full adjuvant effect for Ab responses. We conclude that engineered secretion of Ii adjuvant-tethered Ags establishes a single adjuvant and delivery vehicle platform for potent T and B cell-dependent immunity.


Asunto(s)
Adenoviridae , Anticuerpos Antiprotozoarios/inmunología , Formación de Anticuerpos , Antígenos de Histocompatibilidad Clase II/inmunología , Vacunas contra la Malaria/inmunología , Plasmodium falciparum/inmunología , Vacunación , Animales , Células COS , Chlorocebus aethiops , Femenino , Humanos , Vacunas contra la Malaria/genética , Ratones , Ratones Endogámicos BALB C , Plasmodium falciparum/genética
9.
Malar J ; 19(1): 132, 2020 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-32228596

RESUMEN

BACKGROUND: During the erythrocytic cycle, Plasmodium falciparum malaria parasites express P. falciparum Erythrocyte Membrane Protein 1 (PfEMP1) that anchor the infected erythrocytes (IE) to the vascular lining of the host. The CIDRα1 domain of PfEMP1 is responsible for binding host endothelial protein C receptor (EPCR), and increasing evidence support that this interaction triggers severe malaria, accounting for the majority of malaria-related deaths. In high transmission regions, children develop immunity to severe malaria after the first few infections. This immunity is believed to be mediated by antibodies targeting and inhibiting PfEMP1, causing infected erythrocytes to circulate and be cleared in the spleen. The development of immunity to malaria coincides with acquisition of broad antibody reactivity across the CIDRα1 protein family. Altogether, this identifies CIDRα1 as an important vaccine target. However, the antigenic diversity of the CIDRα1 domain family is a challenge for vaccine development. METHODS: Immune responses in mice vaccinated with Virus-Like Particles (VLP) presenting CIDRα1 antigens were investigated. Antibody reactivity was tested to a panel of recombinant CIDRα1 domains, and the antibodies ability to inhibit EPCR binding by the recombinant CIDRα1 domains was tested in Luminex-based multiplex assays. RESULTS: VLP-presented CIDRα1.4 antigens induced a rapid and strong IgG response capable of inhibiting EPCR-binding of multiple CIDRα1 domains mainly within the group A CIDRα1.4-7 subgroups. CONCLUSIONS: The study observations mirror those from previous CIDRα1 vaccine studies using other vaccine constructs and platforms. This suggests that broad CIDRα1 antibody reactivity may be achieved through vaccination with a limited number of CIDRα1 variants. In addition, this study suggest that this may be achieved through vaccination with a human compatible VLP vaccine platform.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Inmunización , Proteínas Protozoarias/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Animales , Femenino , Ratones , Ratones Endogámicos BALB C , Dominios Proteicos
10.
Malar J ; 19(1): 194, 2020 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-32471507

RESUMEN

BACKGROUND: The multicopy var gene family of Plasmodium falciparum is of crucial importance for pathogenesis and antigenic variation. So far only var2csa, the var gene responsible for placental malaria, was found to be highly conserved among all P. falciparum strains. Here, a new conserved 3D7 var gene (PF3D7_0617400) is identified in several field isolates. METHODS: DNA sequencing, transcriptional analysis, Cluster of Differentiation (CD) 36-receptor binding, indirect immunofluorescence with PF3D7_0617400-antibodies and quantification of surface reactivity against semi-immune sera were used to characterize an NF54 clone and a Gabonese field isolate clone (MOA C3) transcribing the gene. A population of 714 whole genome sequenced parasites was analysed to characterize the conservation of the locus in African and Asian isolates. The genetic diversity of two var2csa fragments was compared with the genetic diversity of 57 microsatellites fragments in field isolates. RESULTS: PFGA01_060022400 was identified in a Gabonese parasite isolate (MOA) from a chronic infection and found to be 99% identical with PF3D7_0617400 of the 3D7 genome strain. Transcriptional analysis and immunofluorescence showed expression of the gene in an NF54 and a MOA clone but CD36 binding assays and surface reactivity to semi-immune sera differed markedly in the two clones. Long-read Pacific bioscience whole genome sequencing showed that PFGA01_060022400 is located in the internal cluster of chromosome 6. The full length PFGA01_060022400 was detected in 36 of 714 P. falciparum isolates and 500 bp fragments were identified in more than 100 isolates. var2csa was in parts highly conserved (He = 0) but in other parts as variable (He = 0.86) as the 57 microsatellites markers (He = 0.8). CONCLUSIONS: Individual var gene sequences exhibit conservation in the global parasite population suggesting that purifying selection may limit overall genetic diversity of some var genes. Notably, field and laboratory isolates expressing the same var gene exhibit markedly different phenotypes.


Asunto(s)
Plasmodium falciparum/genética , Proteínas Protozoarias/análisis , Gabón , Análisis de Secuencia de ADN
11.
Nature ; 513(7518): 431-5, 2014 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-25043062

RESUMEN

Antigenic variation of the Plasmodium falciparum multicopy var gene family enables parasite evasion of immune destruction by host antibodies. Expression of a particular var subgroup, termed upsA, is linked to the obstruction of blood vessels in the brain and to the pathogenesis of human cerebral malaria. The mechanism determining upsA activation remains unknown. Here we show that an entirely new type of gene silencing mechanism involving an exonuclease-mediated degradation of nascent RNA controls the silencing of genes linked to severe malaria. We identify a novel chromatin-associated exoribonuclease, termed PfRNase II, that controls the silencing of upsA var genes by marking their transcription start site and intron-promoter regions leading to short-lived cryptic RNA. Parasites carrying a deficient PfRNase II gene produce full-length upsA var transcripts and intron-derived antisense long non-coding RNA. The presence of stable upsA var transcripts overcomes monoallelic expression, resulting in the simultaneous expression of both upsA and upsC type PfEMP1 proteins on the surface of individual infected red blood cells. In addition, we observe an inverse relationship between transcript levels of PfRNase II and upsA-type var genes in parasites from severe malaria patients, implying a crucial role of PfRNase II in severe malaria. Our results uncover a previously unknown type of post-transcriptional gene silencing mechanism in malaria parasites with repercussions for other organisms. Additionally, the identification of RNase II as a parasite protein controlling the expression of virulence genes involved in pathogenesis in patients with severe malaria may provide new strategies for reducing malaria mortality.


Asunto(s)
Exorribonucleasas/metabolismo , Silenciador del Gen , Genes Protozoarios/genética , Malaria Cerebral/parasitología , Plasmodium falciparum/enzimología , Plasmodium falciparum/genética , ARN Protozoario/metabolismo , Alelos , Variación Antigénica/genética , Cromatina/enzimología , Regulación hacia Abajo/genética , Eritrocitos/parasitología , Exorribonucleasas/deficiencia , Exorribonucleasas/genética , Humanos , Intrones/genética , Malaria Falciparum/parasitología , Plasmodium falciparum/patogenicidad , Regiones Promotoras Genéticas/genética , Proteínas Protozoarias/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Protozoario/genética , ARN no Traducido/genética , ARN no Traducido/metabolismo , Sitio de Iniciación de la Transcripción , Virulencia/genética , Factores de Virulencia/genética
12.
J Infect Dis ; 219(6): 945-954, 2019 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-30335152

RESUMEN

BACKGROUND: The interaction of Plasmodium falciparum-infected erythrocytes (IEs) with the host receptor CD36 is among the most studied host-parasite interfaces. CD36 is a scavenger receptor that binds numerous ligands including the cysteine-rich interdomain region (CIDR)α domains of the erythrocyte membrane protein 1 family (PfEMP1) expressed on the surface of IEs. CD36 is conserved across species, but orthologs display differential binding of IEs. METHODS: In this study, we exploited these differences, combined with the recent crystal structure and 3-dimensional modeling of CD36, to investigate malaria-CD36 structure-function relationships and further define IE-CD36 binding interactions. RESULTS: We show that a charged surface in the membrane-distal region of CD36 is necessary for IE binding. Moreover, IE interaction with this binding surface is influenced by additional CD36 domains, both proximal to and at a distance from this site. CONCLUSIONS: Our data indicate that subtle sequence and spatial differences in these domains modify receptor conformation and regulate the ability of CD36 to selectively interact with its diverse ligands.


Asunto(s)
Antígenos CD36/metabolismo , Eritrocitos/parasitología , Plasmodium falciparum/inmunología , Animales , Antígenos de Protozoos/metabolismo , Sitios de Unión , Antígenos CD36/química , Antígenos CD36/genética , Células CHO , Células COS , Chlorocebus aethiops , Cricetulus , Eritrocitos/fisiología , Interacciones Huésped-Parásitos/genética , Humanos , Malaria Falciparum/inmunología , Mutagénesis , Plasmodium falciparum/fisiología , Relación Estructura-Actividad
13.
J Infect Dis ; 219(5): 808-818, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30365003

RESUMEN

BACKGROUND: Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) mediates parasite sequestration in postcapillary venules in P. falciparum malaria. PfEMP1 types can be classified based on their cysteine-rich interdomain region (CIDR) domains. Antibodies to different PfEMP1 types develop gradually after repeated infections as children age, and antibodies to specific CIDR types may confer protection. METHODS: Levels of immunoglobulin G to 35 recombinant CIDR domains were measured by means of Luminex assay in acute-stage (baseline) and convalescent-stage plasma samples from Papua New Guinean children with severe or uncomplicated malaria and in healthy age-matched community controls. RESULTS: At baseline, antibody levels were similar across the 3 groups. After infection, children with severe malaria had higher antibody levels than those with uncomplicated malaria against the endothelial protein C receptor (EPCR) binding CIDRα1 domains, and this difference was largely confined to older children. Antibodies to EPCR-binding domains increased from presentation to follow-up in severe malaria, but not in uncomplicated malaria. CONCLUSIONS: The acquisition of antibodies against EPCR-binding CIDRα1 domains of PfEMP1 after a severe malaria episode suggest that EPCR-binding PfEMP1 may have a role in the pathogenesis of severe malaria in Papua New Guinea.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Receptor de Proteína C Endotelial/metabolismo , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Niño , Preescolar , Femenino , Humanos , Inmunoglobulina G/sangre , Lactante , Recién Nacido , Masculino , Papúa Nueva Guinea
14.
Nature ; 498(7455): 502-5, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23739325

RESUMEN

Sequestration of Plasmodium falciparum-infected erythrocytes in host blood vessels is a key triggering event in the pathogenesis of severe childhood malaria, which is responsible for about one million deaths every year. Sequestration is mediated by specific interactions between members of the P. falciparum erythrocyte membrane protein 1 (PfEMP1) family and receptors on the endothelial lining. Severe childhood malaria is associated with expression of specific PfEMP1 subtypes containing domain cassettes (DCs) 8 and 13 (ref. 3), but the endothelial receptor for parasites expressing these proteins was unknown. Here we identify endothelial protein C receptor (EPCR), which mediates the cytoprotective effects of activated protein C, as the endothelial receptor for DC8 and DC13 PfEMP1. We show that EPCR binding is mediated through the amino-terminal cysteine-rich interdomain region (CIDRα1) of DC8 and group A PfEMP1 subfamilies, and that CIDRα1 interferes with protein C binding to EPCR. This PfEMP1 adhesive property links P. falciparum cytoadhesion to a host receptor involved in anticoagulation and endothelial cytoprotective pathways, and has implications for understanding malaria pathology and the development of new malaria interventions.


Asunto(s)
Antígenos CD/metabolismo , Malaria Falciparum/patología , Malaria Falciparum/parasitología , Plasmodium falciparum/metabolismo , Receptores de Superficie Celular/metabolismo , Animales , Coagulación Sanguínea , Encéfalo/irrigación sanguínea , Células CHO , Adhesión Celular , Línea Celular , Cricetinae , Células Endoteliales/metabolismo , Receptor de Proteína C Endotelial , Membrana Eritrocítica/metabolismo , Humanos , Inflamación/complicaciones , Inflamación/parasitología , Inflamación/patología , Malaria Falciparum/complicaciones , Microcirculación , Plasmodium falciparum/química , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo
15.
Nature ; 499(7457): 223-7, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23823717

RESUMEN

The variant antigen Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1), which is expressed on the surface of P. falciparum-infected red blood cells, is a critical virulence factor for malaria. Each parasite has 60 antigenically distinct var genes that each code for a different PfEMP1 protein. During infection the clonal parasite population expresses only one gene at a time before switching to the expression of a new variant antigen as an immune-evasion mechanism to avoid the host antibody response. The mechanism by which 59 of the 60 var genes are silenced remains largely unknown. Here we show that knocking out the P. falciparum variant-silencing SET gene (here termed PfSETvs), which encodes an orthologue of Drosophila melanogaster ASH1 and controls histone H3 lysine 36 trimethylation (H3K36me3) on var genes, results in the transcription of virtually all var genes in the single parasite nuclei and their expression as proteins on the surface of individual infected red blood cells. PfSETvs-dependent H3K36me3 is present along the entire gene body, including the transcription start site, to silence var genes. With low occupancy of PfSETvs at both the transcription start site of var genes and the intronic promoter, expression of var genes coincides with transcription of their corresponding antisense long noncoding RNA. These results uncover a previously unknown role of PfSETvs-dependent H3K36me3 in silencing var genes in P. falciparum that might provide a general mechanism by which orthologues of PfSETvs repress gene expression in other eukaryotes. PfSETvs knockout parasites expressing all PfEMP1 proteins may also be applied to the development of a malaria vaccine.


Asunto(s)
Silenciador del Gen , Histonas/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/metabolismo , Factores de Virulencia/genética , Proteínas de Unión al ADN , Proteínas de Drosophila , Eritrocitos/citología , Eritrocitos/metabolismo , Eritrocitos/parasitología , Genes Protozoarios/genética , Histonas/química , Intrones/genética , Lisina/metabolismo , Vacunas contra la Malaria/genética , Metilación , Plasmodium falciparum/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Protozoarias/genética , ARN Largo no Codificante/genética , Factores de Transcripción , Sitio de Iniciación de la Transcripción , Virulencia/genética
16.
Eur J Appl Physiol ; 119(2): 509-518, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30467593

RESUMEN

PURPOSE: Marathon and ultramarathon provoke respiratory muscle fatigue and pulmonary dysfunction; nevertheless, it is unknown how the respiratory system responds to multiple, consecutive days of endurance exercise. METHODS: Nine trained individuals (six male) contested 10 marathons in 10 consecutive days. Respiratory muscle strength (maximum static inspiratory and expiratory mouth-pressures), pulmonary function (spirometry), perceptual ratings of respiratory muscle soreness (Visual Analogue Scale), breathlessness (dyspnea, modified Borg CR10 scale), and symptoms of Upper Respiratory Tract Infection (URTI), were assessed before and after marathons on days 1, 4, 7, and 10. RESULTS: Group mean time for 10 marathons was 276 ± 35 min. Relative to pre-challenge baseline (159 ± 32 cmH2O), MEP was reduced after day 1 (136 ± 31 cmH2O, p = 0.017), day 7 (138 ± 42 cmH2O, p = 0.035), and day 10 (130 ± 41 cmH2O, p = 0.008). There was no change in pre-marathon MEP across days 1, 4, 7, or 10 (p > 0.05). Pre-marathon forced vital capacity was significantly diminished at day 4 (4.74 ± 1.09 versus 4.56 ± 1.09 L, p = 0.035), remaining below baseline at day 7 (p = 0.045) and day 10 (p = 0.015). There were no changes in FEV1, FEV1/FVC, PEF, MIP, or respiratory perceptions during the course of the challenge (p > 0.05). In the 15-day post-challenge period, 5/9 (56%) runners reported symptoms of URTI, relative to 1/9 (11%) pre-challenge. CONCLUSIONS: Single-stage marathon provokes acute expiratory muscle fatigue which may have implications for health and/or performance, but 10 consecutive days of marathon running does not elicit cumulative (chronic) changes in respiratory function or perceptions of dyspnea. These data allude to the robustness of the healthy respiratory system.


Asunto(s)
Pulmón/fisiología , Fatiga Muscular/fisiología , Resistencia Física/fisiología , Músculos Respiratorios/fisiología , Carrera/fisiología , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Respiración , Pruebas de Función Respiratoria , Capacidad Vital/fisiología
17.
Infect Immun ; 86(11)2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30150256

RESUMEN

Plasmodium falciparum malaria pathogenesis is tied to the sequestration of parasites in the microvasculature. Parasite sequestration leading to severe malaria is mediated by P. falciparum erythrocyte membrane protein 1 (PfEMP1) binding to endothelial protein C receptor (EPCR) via its CIDRα1 domains. CIDRα1 domains are targets of naturally acquired immunity, and a vaccine eliciting antibodies inhibiting the EPCR binding of CIDRα1 could potentially prevent disease and death from malaria. CIDRα1 domains have diversified in sequence to escape immune recognition but preserved structure to maintain EPCR binding. The EPCR-binding CIDRα1 domains separate into six major sequence types predicted to form a conserved structure in which only the amino acids essential for EPCR binding are highly conserved. Here, we investigated whether antibodies elicited by vaccination with single or multiple recombinant CIDRα1 domains are able to bind and inhibit diverse CIDRα1 domains. We found that EPCR binding-inhibitory antibodies to CIDRα1 variants closely related to those used for vaccination are readily elicited, whereas antibodies binding distant CIDRα1 variants are sporadically generated and are rarely inhibitory. Despite this, sequence similarity correlated poorly with the ability of induced antibodies to inhibit across diverse variants, and no continuous sequence regions of importance for cross-inhibitory antibodies could be identified. This suggested that epitopes of cross-variant inhibitory antibodies were predominantly conformational. Vaccination with immunogens engineered to focus immune responses to specific epitopes or an optimal choice of multiple CIDRα1 variants may improve elicitation of broadly reactive and inhibitory antibody responses.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antiprotozoarios/sangre , Reacciones Cruzadas , Vacunas contra la Malaria/inmunología , Proteínas Protozoarias/inmunología , Animales , Epítopos/inmunología , Variación Genética , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/genética , Unión Proteica , Proteínas Protozoarias/administración & dosificación , Ratas , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
18.
PLoS Pathog ; 12(11): e1006011, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27835682

RESUMEN

Cytoadhesion of Plasmodium falciparum infected erythrocytes to gC1qR has been associated with severe malaria, but the parasite ligand involved is currently unknown. To assess if binding to gC1qR is mediated through the P. falciparum erythrocyte membrane protein 1 (PfEMP1) family, we analyzed by static binding assays and qPCR the cytoadhesion and var gene transcriptional profile of 86 P. falciparum isolates from Mozambican children with severe and uncomplicated malaria, as well as of a P. falciparum 3D7 line selected for binding to gC1qR (Pf3D7gC1qR). Transcript levels of DC8 correlated positively with cytoadhesion to gC1qR (rho = 0.287, P = 0.007), were higher in isolates from children with severe anemia than with uncomplicated malaria, as well as in isolates from Europeans presenting a first episode of malaria (n = 21) than Mozambican adults (n = 25), and were associated with an increased IgG recognition of infected erythrocytes by flow cytometry. Pf3D7gC1qR overexpressed the DC8 type PFD0020c (5.3-fold transcript levels relative to Seryl-tRNA-synthetase gene) compared to the unselected line (0.001-fold). DBLß12 from PFD0020c bound to gC1qR in ELISA-based binding assays and polyclonal antibodies against this domain were able to inhibit binding to gC1qR of Pf3D7gC1qR and four Mozambican P. falciparum isolates by 50%. Our results show that DC8-type PfEMP1s mediate binding to gC1qR through conserved surface epitopes in DBLß12 domain which can be inhibited by strain-transcending functional antibodies. This study supports a key role for gC1qR in malaria-associated endovascular pathogenesis and suggests the feasibility of designing interventions against severe malaria targeting this specific interaction.


Asunto(s)
Proteínas Portadoras/metabolismo , Malaria Falciparum/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Protozoarias/metabolismo , Adulto , Preescolar , Ensayo de Inmunoadsorción Enzimática , Eritrocitos/parasitología , Femenino , Citometría de Flujo , Humanos , Lactante , Masculino , Plasmodium falciparum
19.
Malar J ; 15(1): 258, 2016 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-27149991

RESUMEN

BACKGROUND: Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) variants are encoded by var genes and mediate pathogenic cytoadhesion and antigenic variation in malaria. PfEMP1s can be broadly divided into three principal groups (A, B and C) and they contain conserved arrangements of functional domains called domain cassettes. Despite their tremendous diversity there is compelling evidence that a restricted subset of PfEMP1s is expressed in severe disease. In this study antibodies from patients with severe and uncomplicated malaria were compared for differences in reactivity with a range of PfEMP1s to determine whether antibodies to particular PfEMP1 domains were associated with severe or uncomplicated malaria. METHODS: Parts of expressed var genes in a severe malaria patient were identified by RNAseq and several of these partial PfEMP1 domains were expressed together with others from laboratory isolates. Antibodies from Papuan patients to these parts of multiple PfEMP1 proteins were measured. RESULTS: Patients with uncomplicated malaria were more likely to have antibodies that recognized PfEMP1 of Group C type and recognized a broader repertoire of group A and B PfEMP1s than patients with severe malaria. CONCLUSION: These data suggest that exposure to a broad range of group A and B PfEMP1s is associated with protection from severe disease in Papua, Indonesia.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Malaria Falciparum/inmunología , Proteínas Protozoarias/inmunología , Adolescente , Adulto , Preescolar , Femenino , Humanos , Indonesia , Masculino , Adulto Joven
20.
Infect Immun ; 83(10): 3972-81, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26216422

RESUMEN

The Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) adhesive proteins expressed on the surfaces of infected erythrocytes (IEs) are of key importance in the pathogenesis of P. falciparum malaria. Several structurally and functionally defined PfEMP1 types have been associated with severe clinical manifestations, such as cerebral malaria in children and placental malaria in pregnant women. PfEMP1 that can bind the Fc part of IgM (Fcµ) characterizes one such type, although the functional significance of this IgM binding to PfEMP1 remains unclear. In this study, we report the identification and functional analysis of five IgM-binding PfEMP1 proteins encoded by P. falciparum NF54. In addition to the VAR2CSA-type PFL0030c protein, already known to bind Fcµ and to mediate chondroitin sulfate A (CSA)-specific adhesion of IEs in the placenta, we found four PfEMP1 proteins not previously known to bind IgM this way. Although they all contained Duffy binding-like ε (DBLε) domains similar to those in VAR2CSA-type PfEMP1, they did not mediate IE adhesion to CSA, and IgM binding did not shield IEs from phagocytosis of IgG-opsonized IEs. In this way, these new IgM-binding PfEMP1 proteins resemble the rosette-mediating and IgM-binding PfEMP1 HB3VAR06, but none of them mediated formation of rosettes. We could map the capacity for Fc-specific IgM binding to DBLε domains near the C terminus for three of the four PfEMP1 proteins tested. Our study provides new evidence regarding Fc-dependent binding of IgM to PfEMP1, which appears to be a common and multifunctional phenotype.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina M/inmunología , Malaria Falciparum/parasitología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Anticuerpos Antiprotozoarios/genética , Genoma de Protozoos , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina M/genética , Malaria Falciparum/genética , Malaria Falciparum/inmunología , Plasmodium falciparum/genética , Unión Proteica , Proteínas Protozoarias/genética , Formación de Roseta
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda