Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
J Infect Dis ; 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38875153

RESUMO

A hallmark of cerebral malaria is sequestration of Plasmodium falciparum-infected erythrocytes (IEs) in the brain microcirculation. Antibodies contribute to malaria immunity, but it remains unclear whether functional antibodies targeting parasite-expressed ligand can block cytoadhesion in the brain. Here, we screened the plasma of older children and young adults in Malawi to characterize the antibody response against the P. falciparum-IE surface and used a bioengineered 3D human brain microvessel model incorporating variable flow dynamics to measure adhesion blocking responses. We found a strong correlation between surface antibody reactivity by flow cytometry and reduced P. falciparum-IE binding in 3D microvessels. Moreover, there was a threshold of surface antibody reactivity necessary to achieve robust inhibitory activity. Our findings provide evidence of the acquisition of adhesion blocking antibodies against cerebral binding variants in people exposed to stable P. falciparum transmission and suggest the quality of the inhibitory response can be influenced by flow dynamics.

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

RESUMO

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.


Assuntos
Aderência Bacteriana , Complemento C1/metabolismo , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Sequência Conservada , Humanos
3.
J Infect Dis ; 219(5): 819-828, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30365004

RESUMO

BACKGROUND: Sequestration of Plasmodium falciparum-infected erythrocytes (IEs) in the microvasculature contributes to pathogenesis of severe malaria in children. This mechanism is mediated by antigens expressed on the IE surface. However, knowledge of specific targets and functions of antibodies to IE surface antigens that protect against severe malaria is limited. METHODS: Antibodies to IE surface antigens were examined in a case-control study of young children in Papua New Guinea presenting with severe or uncomplicated malaria (n = 448), using isolates with a virulent phenotype associated with severe malaria, and functional opsonic phagocytosis assays. We used genetically modified isolates and recombinant P. falciparum erythrocyte membrane protein 1 (PfEMP1) domains to quantify PfEMP1 as a target of antibodies associated with disease severity. RESULTS: Antibodies to the IE surface and recombinant PfEMP1 domains were significantly higher in uncomplicated vs severe malaria and were boosted following infection. The use of genetically modified P. falciparum revealed that PfEMP1 was a major target of antibodies and that PfEMP1-specific antibodies were associated with reduced odds of severe malaria. Furthermore, antibodies promoting the opsonic phagocytosis of IEs by monocytes were lower in those with severe malaria. CONCLUSIONS: Findings suggest that PfEMP1 is a dominant target of antibodies associated with reduced risk of severe malaria, and function in part by promoting opsonic phagocytosis.


Assuntos
Anticorpos Antiprotozoários/sangue , Eritrócitos/parasitologia , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Anticorpos Antiprotozoários/imunologia , Estudos de Casos e Controles , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Proteínas Opsonizantes/sangue , Proteínas Opsonizantes/imunologia , Papua Nova Guiné , Fagocitose
4.
Nature ; 498(7455): 502-5, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-23739325

RESUMO

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.


Assuntos
Antígenos CD/metabolismo , Malária Falciparum/patologia , Malária Falciparum/parasitologia , Plasmodium falciparum/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Coagulação Sanguínea , Encéfalo/irrigação sanguínea , Células CHO , Adesão Celular , Linhagem Celular , Cricetinae , Células Endoteliais/metabolismo , Receptor de Proteína C Endotelial , Membrana Eritrocítica/metabolismo , Humanos , Inflamação/complicações , Inflamação/parasitologia , Inflamação/patologia , Malária Falciparum/complicações , Microcirculação , Plasmodium falciparum/química , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo
5.
Proc Natl Acad Sci U S A ; 113(23): E3270-9, 2016 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-27185931

RESUMO

The interplay between cellular and molecular determinants that lead to severe malaria in adults is unexplored. Here, we analyzed parasite virulence factors in an infected adult population in India and investigated whether severe malaria isolates impair endothelial protein C receptor (EPCR), a protein involved in coagulation and endothelial barrier permeability. Severe malaria isolates overexpressed specific members of the Plasmodium falciparum var gene/PfEMP1 (P. falciparum erythrocyte membrane protein 1) family that bind EPCR, including DC8 var genes that have previously been linked to severe pediatric malaria. Machine learning analysis revealed that DC6- and DC8-encoding var transcripts in combination with high parasite biomass were the strongest indicators of patient hospitalization and disease severity. We found that DC8 CIDRα1 domains from severe malaria isolates had substantial differences in EPCR binding affinity and blockade activity for its ligand activated protein C. Additionally, even a low level of inhibition exhibited by domains from two cerebral malaria isolates was sufficient to interfere with activated protein C-barrier protective activities in human brain endothelial cells. Our findings demonstrate an interplay between parasite biomass and specific PfEMP1 adhesion types in the development of adult severe malaria, and indicate that low impairment of EPCR function may contribute to parasite virulence.


Assuntos
Malária Falciparum/parasitologia , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/genética , Adulto , Antígenos CD/genética , Antígenos CD/metabolismo , Biomassa , Receptor de Proteína C Endotelial , Feminino , Humanos , Aprendizado de Máquina , Malária Falciparum/genética , Malária Falciparum/metabolismo , Masculino , Pessoa de Meia-Idade , Proteína C/metabolismo , Domínios Proteicos , Proteínas de Protozoários/química , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Virulência , Adulto Jovem
6.
Cell Microbiol ; 17(12): 1883-99, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26119044

RESUMO

Plasmodium falciparum-infected erythrocytes (IRBC) expressing the domain cassettes (DC) 8 and 13 of the cytoadherent ligand P. falciparum erythrocyte membrane protein 1 adhere to the endothelial protein C receptor (EPCR). By interfering with EPCR anti-coagulant and pro-endothelial barrier functions, IRBC adhesion could promote coagulation and vascular permeability that contribute to the pathogenesis of cerebral malaria. In this study, we examined the adhesion of DC8- and DC13-expressing parasite lines to endothelial cells from different microvasculature, and the consequences of EPCR engagement on endothelial cell function. We found that IRBC from IT4var19 (DC8) and IT4var07 (DC13) parasite lines adhered to human brain, lung and dermal endothelial cells under shear stress. However, the relative contribution of EPCR to parasite cytoadherence on different types of endothelial cell varied. We also observed divergent functional outcomes for DC8 cysteine-rich interdomain region (CIDR)α1.1 and DC13 CIDRα1.4 domains. IT4var07 CIDRα1.4 inhibited generation of activated protein C (APC) on lung and dermal endothelial cells and blocked the APC-EPCR binding interaction on brain endothelial cells. IT4var19 CIDRα1.1 inhibited thrombin-induced endothelial barrier dysfunction in lung endothelial cells, whereas IT4var07 CIDRα1.4 inhibited the protective effect of APC on thrombin-induced permeability. Overall, these findings reveal a much greater complexity of how CIDRα1-expressing parasites may modulate malaria pathogenesis through EPCR adhesion.


Assuntos
Antígenos CD/metabolismo , Adesão Celular , Células Endoteliais/fisiologia , Eritrócitos/parasitologia , Interações Hospedeiro-Patógeno , Plasmodium falciparum/fisiologia , Receptores de Superfície Celular/metabolismo , Células Cultivadas , Receptor de Proteína C Endotelial , Humanos , Ligadura , Resultado do Tratamento
7.
Cell Microbiol ; 17(12): 1868-82, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26118955

RESUMO

Cytoadhesion of Plasmodium falciparum-infected erythrocytes to endothelial protein C receptor (EPCR) is associated with severe malaria. It has been postulated that parasite binding could exacerbate microvascular coagulation and endothelial dysfunction in cerebral malaria by impairing the protein C-EPCR interaction, but the extent of binding inhibition has not been fully determined. Here we expressed the cysteine-rich interdomain region (CIDRα1) domain from a variety of domain cassette (DC) 8 and DC13 P. falciparum erythrocyte membrane protein 1 proteins and show they interact in a distinct manner with EPCR resulting in weak, moderate and strong inhibition of the activated protein C (APC)-EPCR interaction. Overall, there was a positive correlation between CIDRα1-EPCR binding activity and APC blockade activity. In addition, our analysis from a combination of mutagenesis and blocking antibodies finds that an Arg81 (R81) in EPCR plays a pivotal role in CIDRα1 binding, but domains with weak and strong APC blockade activity were distinguished by their sensitivity to inhibition by anti-EPCR mAb 1535, implying subtle differences in their binding footprints. These data reveal a previously unknown functional heterogeneity in the interaction between P. falciparum and EPCR and have major implications for understanding the distinct clinical pathologies of cerebral malaria and developing new treatment strategies.


Assuntos
Adesão Celular , Células Endoteliais/fisiologia , Interações Hospedeiro-Patógeno , Malária/parasitologia , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/metabolismo , Receptores de Superfície Celular/antagonistas & inibidores , Animais , Antígenos CD/genética , Células CHO , Cricetulus , Análise Mutacional de DNA , Receptor de Proteína C Endotelial , Humanos , Malária/patologia , Ligação Proteica , Estrutura Terciária de Proteína , Receptores de Superfície Celular/genética , Análise de Sequência de DNA
8.
Malar J ; 15(1): 569, 2016 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-27884146

RESUMO

BACKGROUND: Malaria remains an important cause of morbidity and mortality in India. Though many comprehensive studies have been carried out in Africa and Southeast Asia to characterize and examine determinants of Plasmodium falciparum and Plasmodium vivax malaria pathogenesis, fewer have been conducted in India. METHODS: A prospective study of malaria-positive individuals was conducted at Goa Medical College and Hospital (GMC) from 2012 to 2015 to identify demographic, diagnostic and clinical indicators associated with P. falciparum and P. vivax infection on univariate analysis. RESULTS: Between 2012 and 2015, 74,571 febrile individuals, 6287 (8.4%) of whom were malaria positive, presented to GMC. The total number of malaria cases at GMC increased more than two-fold over four years, with both P. vivax and P. falciparum cases present year-round. Some 1116 malaria-positive individuals (mean age = 27, 91% male), 88.2% of whom were born outside of Goa and 51% of whom were construction workers, were enroled in the study. Of 1088 confirmed malaria-positive patients, 77.0% had P. vivax, 21.0% had P. falciparum and 2.0% had mixed malaria. Patients over 40 years of age and with P. falciparum infection were significantly (p < 0.001) more likely to be hospitalised than younger and P. vivax patients, respectively. While approximately equal percentages of hospitalised P. falciparum (76.6%) and P. vivax (78.9%) cases presented with at least one WHO severity indicator, a greater percentage of P. falciparum inpatients presented with at least two (43.9%, p < 0.05) and at least three (29.9%, p < 0.01) severity features. There were six deaths among the 182 hospitalised malaria positive patients, all of whom had P. falciparum. CONCLUSION: During the four year study period at GMC, the number of malaria cases increased substantially and the greatest burden of severe disease was contributed by P. falciparum.


Assuntos
Malária Falciparum/patologia , Malária Vivax/patologia , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Demografia , Feminino , Humanos , Incidência , Índia/epidemiologia , Lactente , Malária Falciparum/diagnóstico , Malária Falciparum/epidemiologia , Malária Vivax/diagnóstico , Malária Vivax/epidemiologia , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Centros de Atenção Terciária , Adulto Jovem
9.
PLoS Pathog ; 9(6): e1003430, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23825944

RESUMO

During blood stage infection, Plasmodium falciparum infected erythrocytes (IE) bind to host blood vessels. This virulence determinant enables parasites to evade spleen-dependent killing mechanisms, but paradoxically in some cases may reduce parasite fitness by killing the host. Adhesion of infected erythrocytes is mediated by P. falciparum erythrocyte membrane protein 1 (PfEMP1), a family of polymorphic adhesion proteins encoded by var genes. Whereas cerebral binding and severe malaria are associated with parasites expressing DC8 and DC13 var genes, relatively little is known about the non-brain endothelial selection on severe malaria adhesive types. In this study, we selected P. falciparum-IEs on diverse endothelial cell types and demonstrate that DC8 and DC13 var genes were consistently among the major var transcripts selected on non-brain endothelial cells (lung, heart, bone marrow). To investigate the molecular basis for this avid endothelial binding activity, recombinant proteins were expressed from the predominant upregulated DC8 transcript, IT4var19. In-depth binding comparisons revealed that multiple extracellular domains from this protein bound brain and non-brain endothelial cells, and individual domains largely did not discriminate between different endothelial cell types. Additionally, we found that recombinant DC8 and DC13 CIDR1 domains exhibited a widespread endothelial binding activity and could compete for DC8-IE binding to brain endothelial cells, suggesting they may bind the same host receptor. Our findings provide new insights into the interaction of severe malaria adhesive types and host blood vessels and support the hypothesis that parasites causing severe malaria express PfEMP1 variants with a superior ability to adhere to diverse endothelial cell types, and may therefore endow these parasites with a growth and transmission advantage.


Assuntos
Células Endoteliais/metabolismo , Malária Cerebral/metabolismo , Malária Falciparum/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/biossíntese , Linhagem Celular Transformada , Células Endoteliais/patologia , Feminino , Humanos , Malária Cerebral/genética , Malária Cerebral/patologia , Malária Falciparum/genética , Malária Falciparum/patologia , Masculino , Plasmodium falciparum/genética , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas de Protozoários/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Índice de Gravidade de Doença
10.
PLoS Pathog ; 9(6): e1003420, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23853575

RESUMO

Glycan masking is an emerging vaccine design strategy to focus antibody responses to specific epitopes, but it has mostly been evaluated on the already heavily glycosylated HIV gp120 envelope glycoprotein. Here this approach was used to investigate the binding interaction of Plasmodium vivax Duffy Binding Protein (PvDBP) and the Duffy Antigen Receptor for Chemokines (DARC) and to evaluate if glycan-masked PvDBPII immunogens would focus the antibody response on key interaction surfaces. Four variants of PVDBPII were generated and probed for function and immunogenicity. Whereas two PvDBPII glycosylation variants with increased glycan surface coverage distant from predicted interaction sites had equivalent binding activity to wild-type protein, one of them elicited slightly better DARC-binding-inhibitory activity than wild-type immunogen. Conversely, the addition of an N-glycosylation site adjacent to a predicted PvDBP interaction site both abolished its interaction with DARC and resulted in weaker inhibitory antibody responses. PvDBP is composed of three subdomains and is thought to function as a dimer; a meta-analysis of published PvDBP mutants and the new DBPII glycosylation variants indicates that critical DARC binding residues are concentrated at the dimer interface and along a relatively flat surface spanning portions of two subdomains. Our findings suggest that DARC-binding-inhibitory antibody epitope(s) lie close to the predicted DARC interaction site, and that addition of N-glycan sites distant from this site may augment inhibitory antibodies. Thus, glycan resurfacing is an attractive and feasible tool to investigate protein structure-function, and glycan-masked PvDBPII immunogens might contribute to P. vivax vaccine development.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Plasmodium vivax/imunologia , Proteínas de Protozoários/imunologia , Receptores de Superfície Celular/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/genética , Células COS , Chlorocebus aethiops , Sistema do Grupo Sanguíneo Duffy/genética , Sistema do Grupo Sanguíneo Duffy/imunologia , Glicosilação , Vacinas Antimaláricas/genética , Camundongos , Plasmodium vivax/genética , Ligação Proteica , Proteínas de Protozoários/genética , Ratos , Receptores de Superfície Celular/genética
11.
Proc Natl Acad Sci U S A ; 109(26): E1782-90, 2012 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-22619321

RESUMO

Cerebral malaria (CM) is a deadly complication of Plasmodium falciparum infection, but specific interactions involved in cerebral homing of infected erythrocytes (IEs) are poorly understood. In this study, P. falciparum-IEs were characterized for binding to primary human brain microvascular endothelial cells (HBMECs). Before selection, CD36 or ICAM-1-binding parasites exhibited punctate binding to a subpopulation of HBMECs and binding was CD36 dependent. Panning of IEs on HBMECs led to a more dispersed binding phenotype and the selection of three var genes, including two that encode the tandem domain cassette 8 (DC8) and were non-CD36 binders. Multiple domains in the DC8 cassette bound to brain endothelium and the cysteine-rich interdomain region 1 inhibited binding of P. falciparum-IEs by 50%, highlighting a key role for the DC8 cassette in cerebral binding. It is mysterious how deadly binding variants are maintained in the parasite population. Clonal parasite lines expressing the two brain-adherent DC8-var genes did not bind to any of the known microvascular receptors, indicating unique receptors are involved in cerebral binding. They could also adhere to brain, lung, dermis, and heart endothelial cells, suggesting cerebral binding variants may have alternative sequestration sites. Furthermore, young African children with CM or nonsevere control cases had antibodies to HBMEC-selected parasites, indicating they had been exposed to related variants during childhood infections. This analysis shows that specific P. falciparum erythrocyte membrane protein 1 types are linked to cerebral binding and suggests a potential mechanism by which individuals may build up immunity to severe disease, in the absence of CM.


Assuntos
Encéfalo/irrigação sanguínea , Adesão Celular , Endotélio Vascular/patologia , Eritrócitos/parasitologia , Genes de Protozoários , Malária Cerebral/parasitologia , Plasmodium falciparum/fisiologia , Animais , Pré-Escolar , Eritrócitos/patologia , Humanos , Malária Cerebral/patologia , Plasmodium falciparum/genética
12.
Cell Microbiol ; 15(12): 1976-83, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23957661

RESUMO

Cytoadhesion of Plasmodium falciparum-infected erythrocytes to host microvasculature is a key virulence determinant. Parasite binding is mediated by a large family of clonally variant adhesion proteins, termed P. falciparum erythrocyte membrane protein 1 (PfEMP1), encoded by var genes and expressed at the infected erythrocyte surface. Although PfEMP1 proteins have extensively diverged under opposing selection pressure to maintain ligand binding while avoiding antibody-mediated detection, recent work has revealed they can be classified into different groups based on chromosome location and domain composition. This grouping reflects functional specialization of PfEMP1 proteins for different human host and microvascular binding niches and appears to be maintained by gene recombination hierarchies. Inone extreme, a specific PfEMP1 variant is associated with placental binding and malaria during pregnancy, while other PfEMP1 subtypes appear to be specialized for infection of malaria naïve hosts. Here, we discuss recent findings on the origins and evolution of the var gene family, the structure-function of PfEMP1 proteins, and a distinct subset of PfEMP1 variants that have been associated with severe childhood malaria.


Assuntos
Adesão Celular/imunologia , Membrana Eritrocítica/parasitologia , Malária Falciparum/sangue , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/imunologia , Variação Antigênica/genética , Variação Antigênica/imunologia , Células Endoteliais/metabolismo , Membrana Eritrocítica/imunologia , Eritrócitos/imunologia , Eritrócitos/metabolismo , Feminino , Humanos , Plasmodium falciparum/genética , Plasmodium falciparum/imunologia , Gravidez , Proteínas de Protozoários/classificação , Proteínas de Protozoários/genética , Formação de Roseta
13.
Cell Microbiol ; 15(8): 1401-18, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23421990

RESUMO

The human malaria parasite, Plasmodium falciparum, modifies the red blood cells (RBCs) that it infects by exporting proteins to the host cell. One key virulence protein, P. falciparum Erythrocyte Membrane Protein-1 (PfEMP1), is trafficked to the surface of the infected RBC, where it mediates adhesion to the vascular endothelium. We have investigated the organization and development of the exomembrane system that is used for PfEMP1 trafficking. Maurer's cleft cisternae are formed early after invasion and proteins are delivered to these (initially mobile) structures in a temporally staggered and spatially segregated manner. Membrane-Associated Histidine-Rich Protein-2 (MAHRP2)-containing tether-like structures are generated as early as 4 h post invasion and become attached to Maurer's clefts. The tether/Maurer's cleft complex docks onto the RBC membrane at ~20 h post invasion via a process that is not affected by cytochalasin D treatment. We have examined the trafficking of a GFP chimera of PfEMP1 expressed in transfected parasites. PfEMP1B-GFP accumulates near the parasite surface, within membranous structures exhibiting a defined ultrastructure, before being transferred to pre-formed mobile Maurer's clefts. Endogenous PfEMP1 and PfEMP1B-GFP are associated with Electron-Dense Vesicles that may be responsible for trafficking PfEMP1 from the Maurer's clefts to the RBC membrane.


Assuntos
Eritrócitos/parasitologia , Plasmodium falciparum/patogenicidade , Transporte Proteico/fisiologia , Proteínas de Protozoários/fisiologia , Células Cultivadas , Membrana Eritrocítica/parasitologia , Membrana Eritrocítica/fisiologia , Eritrócitos/patologia , Proteínas de Fluorescência Verde , Interações Hospedeiro-Parasita/fisiologia , Humanos , Técnicas In Vitro , Plasmodium falciparum/fisiologia
14.
Eukaryot Cell ; 12(11): 1490-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24014767

RESUMO

Cytoadhesion of Plasmodium falciparum parasitized red blood cells (pRBCs) has been implicated in the virulence of malaria infection. Cytoadhesive interactions are mediated by the protein family of Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1). The PfEMP1 family is under strong antibody and binding selection, resulting in extensive sequence and size variation of the extracellular domains. Here, we investigated cytoadhesion of pRBCs to CD36, a common receptor of P. falciparum field isolates, under dynamic flow conditions. Isogeneic parasites, predominantly expressing single PfEMP1 variants, were evaluated for binding to recombinant CD36 under dynamic flow conditions using microfluidic devices. We tested if PfEMP1 size (number of extracellular domains) or sequence variation affected the pRBC-CD36 interaction. Our analysis showed that clonal parasite variants varied ∼5-fold in CD36 rolling velocity despite extensive PfEMP1 sequence polymorphism. In addition, adherent pRBCs exhibited a characteristic hysteresis in rolling velocity at microvascular flow rates, which was accompanied by changes in pRBC shape and may represent important adaptations that favor stable binding.


Assuntos
Antígenos CD36/metabolismo , Microfluídica , Plasmodium falciparum/metabolismo , Polimorfismo Genético , Proteínas de Protozoários/metabolismo , Adesão Celular , Humanos , Plasmodium falciparum/genética , Plasmodium falciparum/fisiologia , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas de Protozoários/química , Proteínas de Protozoários/genética
15.
Life Sci Alliance ; 7(5)2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38467420

RESUMO

Kinases are key players in endothelial barrier regulation, yet their temporal function and regulatory phosphosignaling networks are incompletely understood. We developed a novel methodology, Temporally REsolved KInase Network Generation (TREKING), which combines a 28-kinase inhibitor screen with machine learning and network reconstruction to build time-resolved, functional phosphosignaling networks. We demonstrated the utility of TREKING for identifying pathways mediating barrier integrity after activation by thrombin with or without TNF preconditioning in brain endothelial cells. TREKING predicted over 100 kinases involved in barrier regulation and discerned complex condition-specific pathways. For instance, the MAPK-activated protein kinase 2 (MAPKAPK2/MK2) had early barrier-weakening activity in both inflammatory conditions but late barrier-strengthening activity exclusively with thrombin alone. Using temporal Western blotting, we confirmed that MAPKAPK2/MK2 was differentially phosphorylated under the two inflammatory conditions. We further showed with lentivirus-mediated knockdown of MAPK14/p38α and drug targeting the MAPK14/p38α-MAPKAPK2/MK2 complex that a MAP3K20/ZAK-MAPK14/p38α axis controlled the late activation of MAPKAPK2/MK2 in the thrombin-alone condition. Beyond the MAPKAPK2/MK2 switch, TREKING predicts extensive interconnected networks that control endothelial barrier dynamics.


Assuntos
Células Endoteliais , Proteína Quinase 14 Ativada por Mitógeno , Células Endoteliais/metabolismo , Trombina/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Inibidores de Proteínas Quinases
16.
Infect Immun ; 81(2): 487-95, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23208604

RESUMO

Pregnancy malaria is caused by Plasmodium falciparum-infected erythrocytes that adhere to the placental receptor chondroitin sulfate A (CSA) and sequester in the placenta; women become resistant to pregnancy malaria as they acquire antiadhesion antibodies that target surface proteins of placental parasites. VAR2CSA, a member of the P. falciparum EMP1 variant surface antigen family, is the leading candidate for a pregnancy malaria vaccine. Because VAR2CSA is a high-molecular-weight protein, a vaccine based on the full-length protein may not be feasible. An alternative approach has been to develop a vaccine targeting individual Duffy binding-like (DBL) domains. In this study, a consortium of laboratories under the Pregnancy Malaria Initiative compared the functional activity of antiadhesion antibodies elicited by different VAR2CSA domains and variants produced in prokaryotic and eukaryotic expression systems. Antisera were initially tested against laboratory lines of maternal parasites, and the most promising reagents were evaluated in the field against fresh placental parasite samples. Recombinant proteins expressed in Escherichia coli elicited antibody levels similar to those expressed in eukaryotic systems, as did the two allelic forms of the DBL4 and DBL5 domains. The procedures developed for this head-to-head comparison will be useful for future evaluation and down-selection of malaria vaccine immunogens.


Assuntos
Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Placenta/imunologia , Plasmodium falciparum/imunologia , Complicações Parasitárias na Gravidez/imunologia , Animais , Sulfatos de Condroitina/imunologia , Estudos de Coortes , Feminino , Humanos , Soros Imunes/imunologia , Imunoglobulina G/imunologia , Estudos Longitudinais , Vacinas Antimaláricas/farmacologia , Malária Falciparum/prevenção & controle , Gravidez , Complicações Parasitárias na Gravidez/prevenção & controle , Ratos , Proteínas Recombinantes/imunologia
17.
PLoS Pathog ; 7(5): e1002032, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21573138

RESUMO

The Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) family plays a central role in antigenic variation and cytoadhesion of P. falciparum infected erythrocytes. PfEMP1 proteins/var genes are classified into three main subfamilies (UpsA, UpsB, and UpsC) that are hypothesized to have different roles in binding and disease. To investigate whether these subfamilies have diverged in binding specificity and test if binding could be predicted by adhesion domain classification, we generated a panel of 19 parasite lines that primarily expressed a single dominant var transcript and assayed binding against 12 known host receptors. By limited dilution cloning, only UpsB and UpsC var genes were isolated, indicating that UpsA var gene expression is rare under in vitro culture conditions. Consequently, three UpsA variants were obtained by rosette purification and selection with specific monoclonal antibodies to create a more representative panel. Binding assays showed that CD36 was the most common adhesion partner of the parasite panel, followed by ICAM-1 and TSP-1, and that CD36 and ICAM-1 binding variants were highly predicted by adhesion domain sequence classification. Binding to other host receptors, including CSA, VCAM-1, HABP1, CD31/PECAM, E-selectin, Endoglin, CHO receptor "X", and Fractalkine, was rare or absent. Our findings identify a category of larger PfEMP1 proteins that are under dual selection for ICAM-1 and CD36 binding. They also support that the UpsA group, in contrast to UpsB and UpsC var genes, has diverged from binding to the major microvasculature receptor CD36 and likely uses other mechanisms to sequester in the microvasculature. These results demonstrate that CD36 and ICAM-1 have left strong signatures of selection on the PfEMP1 family that can be detected by adhesion domain sequence classification and have implications for how this family of proteins is specializing to exploit hosts with varying levels of anti-malaria immunity.


Assuntos
Eritrócitos/parasitologia , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Variação Antigênica , Antígenos CD36/metabolismo , Células CHO , Adesão Celular , Células Cultivadas , Clonagem Molecular , Cricetinae , Cricetulus , Eritrócitos/metabolismo , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Malária Falciparum/genética , Malária Falciparum/metabolismo , Malária Falciparum/parasitologia , Fenótipo , Plasmodium falciparum/patogenicidade , Ligação Proteica/genética , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trombospondina 1/metabolismo , Transcrição Gênica
18.
Cell Rep ; 42(10): 113253, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37819760

RESUMO

Sequestration of Plasmodium falciparum-infected erythrocytes (IEs) in the brain microcirculation is a hallmark of cerebral malaria (CM), which leads to endothelial activation, brain swelling, and death. Here, we probed CM inflammation in a perfusable 3D human brain microvessel model. 3D brain microvessels supported in vivo-like capacities for parasite binding and maturation in situ, leading to a distinct inflammatory response from the pro-inflammatory cytokine tumor necrosis factor α (TNF-α). By combining transcriptional analysis, imaging, and leukocyte perfusion, we showed that whereas TNF-α promotes a reversible inflammatory phenotype with widespread leukocyte recruitment, parasites induce unique stress response pathways and cause localized cell adhesivity changes, focal endothelial disruptions, and apoptosis. Furthermore, parasites modified the temporal kinetics of the TNF transcriptional response, suggesting augmented inflammatory damage with the two sequential stimuli. Our findings offer mechanistic insights into CM biology in a 3D brain microvessel mimetic platform and suggest that multiple events intersect to promote brain barrier inflammation in CM.


Assuntos
Malária Cerebral , Malária Falciparum , Humanos , Fator de Necrose Tumoral alfa , Encéfalo/patologia , Plasmodium falciparum/genética , Inflamação/patologia , Microvasos/patologia , Eritrócitos/parasitologia , Malária Falciparum/parasitologia
19.
Heliyon ; 9(8): e18581, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37593608

RESUMO

This paper aims at reviewing and analyzing the operation and design of a utility flare in an oilfield in the Iraq/Kurdistan region. The flare supports a gas separation unit that separates 100 MMSCFD of natural gas from other liquid compounds in petroleum refining. The actual flare dimensions are 50 m high and 0.6 m diameter and works in summer where the crosswind speed is 9 m/s and a flow of 1.2 MMSCFD of treated natural gas is flaring through it. At the beginning, the flare design was performed using the API-521 recommended approach based on full operating capacity of the unit and composition of the gas to be flared. The API-521 based design resulted in a flare with a 0.76 m diameter and 48.19 m height. The effects of stack height on heat radiation in case of full capacity firing showed that as the flare height increases from 42.34 m to 133.05 m, the heat radiation decreases from 15.8 kW/m^2 to 1.6 kW/m^2 within 45.7 m dimeter. Furthermore, the relation between stack height and heat radiation was studied for the actual firing rate 1.2MMSCFD using simulation, where the results showed that as the stack height increasing from 10 m to 50 m the heat radiation decreasing from over 1000 w/m^2 to around 150 W/m^2. In fact, CFD code C3d was used to analyze flare performance at normal firing condition during summer operation of 1.2 MMSCFD with a flare diameter and height of 50 m and 0.6 m, respectively. The code was able to predict the flame shape and size during actual flare operation. The results of the simulation demonstrated by defining four locations in the domain to measure the average temperatures and emissions, and to calculate the Combustion Efficiency (CE) and Destruction and Removal Efficiency (DRE). These points were 6 m, 8 m, 10 m, 12 m far from the tip on x-axis and at height of 52 m. The results showed that the average temperature at 6 m far from the flare is 658 K and it decreasing to 490 K at 12 m away from the tip. The CO and CO2 also decreased from 7.27E-5 and 0.033 mass% to 4.53E-6 and 0.027 mass%, respectively. Generally, soot formation was low but at points 8 m and 10 m from the tip the soot formation was considerably lower, respectively at 6.16E-5 and 8.71E-5 mass%. The emissions of C1, C2, C3 and C6+ were measured at 7.46E-9, 5.39E-9, 5.13E-9 and 4.35E-9 mass% at 6 m away from the tip. The emissions increased slightly at 8 m and 10 m from the tip but at 12 m they were observed to decrease. The flare CE and DRE were estimated to be 98% and 100%, respectively. Analysis results confirmed that the flare design was safe and the flare operation was highly efficient with very little smoke produced as indicated by the predicted CE and DRE.

20.
Infect Immun ; 80(4): 1479-90, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22331427

RESUMO

Placental malaria, caused by sequestration of Plasmodium falciparum-infected erythrocytes in the placenta, is associated with increased risk of maternal morbidity and poor birth outcomes. The parasite antigen VAR2CSA (variant surface antigen 2-chondroitin sulfate A) is expressed on infected erythrocytes and mediates binding to chondroitin sulfate A, initiating inflammation and disrupting homeostasis at the maternal-fetal interface. Although antibodies can prevent sequestration, it is unclear whether parasite clearance is due to antibodies to a single Duffy binding-like (DBL) domain or to an extensive repertoire of antibodies to multiple DBL domains and allelic variants. Accordingly, plasma samples collected longitudinally from pregnant women were screened for naturally acquired antibodies against an extensive panel of VAR2CSA proteins, including 2 to 3 allelic variants for each of 5 different DBL domains. Analyses were performed on plasma samples collected from 3 to 9 months of pregnancy from women living in areas in Cameroon with high and low malaria transmission. The results demonstrate that high antibody levels to multiple VAR2CSA domains, rather than a single domain, were associated with the absence of placental malaria when antibodies were present from early in the second trimester until term. Absence of placental malaria was associated with increasing antibody breadth to different DBL domains and allelic variants in multigravid women. Furthermore, the antibody responses of women in the lower-transmission site had both lower magnitude and lesser breadth than those in the high-transmission site. These data suggest that immunity to placental malaria results from high antibody levels to multiple VAR2CSA domains and allelic variants and that antibody breadth is influenced by malaria transmission intensity.


Assuntos
Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/imunologia , Malária Falciparum/imunologia , Placenta/parasitologia , Plasmodium falciparum/imunologia , Complicações Parasitárias na Gravidez/imunologia , Adulto , Anticorpos Antiprotozoários/imunologia , Camarões , Feminino , Humanos , Malária Falciparum/parasitologia , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Doenças Placentárias/imunologia , Doenças Placentárias/parasitologia , Gravidez , Complicações Parasitárias na Gravidez/parasitologia , Proteínas de Protozoários/imunologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa