Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell Chem Biol ; 30(11): 1377-1389.e8, 2023 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-37586370

RESUMO

TruAB Discovery is an approach that integrates cellular immunology, high-throughput immunosequencing, bioinformatics, and computational biology in order to discover naturally occurring human antibodies for prophylactic or therapeutic use. We adapted our previously described pairSEQ technology to pair B cell receptor heavy and light chains of SARS-CoV-2 spike protein-binding antibodies derived from enriched antigen-specific memory B cells and bulk antibody-secreting cells. We identified approximately 60,000 productive, in-frame, paired antibody sequences, from which 2,093 antibodies were selected for functional evaluation based on abundance, isotype and patterns of somatic hypermutation. The exceptionally diverse antibodies included RBD-binders with broad neutralizing activity against SARS-CoV-2 variants, and S2-binders with broad specificity against betacoronaviruses and the ability to block membrane fusion. A subset of these RBD- and S2-binding antibodies demonstrated robust protection against challenge in hamster and mouse models. This high-throughput approach can accelerate discovery of diverse, multifunctional antibodies against any target of interest.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , Camundongos , Humanos , Anticorpos Neutralizantes , Anticorpos Amplamente Neutralizantes , Anticorpos Antivirais
2.
Cell Rep ; 42(1): 112014, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36681898

RESUMO

The SARS-CoV-2 Omicron variant of concern (VoC) and its sublineages contain 31-36 mutations in spike and escape neutralization by most therapeutic antibodies. In a pseudovirus neutralization assay, 66 of the nearly 400 candidate therapeutics in the Coronavirus Immunotherapeutic Consortium (CoVIC) panel neutralize Omicron and multiple Omicron sublineages. Among natural immunoglobulin Gs (IgGs), especially those in the receptor-binding domain (RBD)-2 epitope community, nearly all Omicron neutralizers recognize spike bivalently, with both antigen-binding fragments (Fabs) simultaneously engaging adjacent RBDs on the same spike. Most IgGs that do not neutralize Omicron bind either entirely monovalently or have some (22%-50%) monovalent occupancy. Cleavage of bivalent-binding IgGs to Fabs abolishes neutralization and binding affinity, with disproportionate loss of activity against Omicron pseudovirus and spike. These results suggest that VoC-resistant antibodies overcome mutagenic substitution via avidity. Hence, vaccine strategies targeting future SARS-CoV-2 variants should consider epitope display with spacing and organization identical to trimeric spike.


Assuntos
COVID-19 , Humanos , SARS-CoV-2 , Etnicidade , Epitopos , Anticorpos Antivirais , Anticorpos Neutralizantes , Testes de Neutralização
3.
J Infect Dis ; 225(11): 2011-2022, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34718641

RESUMO

BACKGROUND: Plasmodium falciparum-infected red blood cells (iRBCs) bind and sequester in deep vascular beds, causing malaria-related disease and death. In pregnant women, VAR2CSA binds to chondroitin sulfate A (CSA) and mediates placental sequestration, making it the major placental malaria (PM) vaccine target. METHODS: In this study, we characterize an invariant protein associated with PM called P falciparum chondroitin sulfate A ligand (PfCSA-L). RESULTS: Recombinant PfCSA-L binds both placental CSA and VAR2CSA with nanomolar affinity, and it is coexpressed on the iRBC surface with VAR2CSA. Unlike VAR2CSA, which is anchored by a transmembrane domain, PfCSA-L is peripherally associated with the outer surface of knobs through high-affinity protein-protein interactions with VAR2CSA. This suggests that iRBC sequestration involves complexes of invariant and variant surface proteins, allowing parasites to maintain both diversity and function at the iRBC surface. CONCLUSIONS: The PfCSA-L is a promising target for intervention because it is well conserved, exposed on infected cells, and expressed and localized with VAR2CSA.


Assuntos
Vacinas Antimaláricas , Malária Falciparum , Malária , Anticorpos Antiprotozoários , Antígenos de Protozoários , Sulfatos de Condroitina , Eritrócitos/parasitologia , Feminino , Humanos , Malária/prevenção & controle , Malária Falciparum/parasitologia , Placenta/parasitologia , Plasmodium falciparum , Gravidez
4.
J Exp Med ; 218(4)2021 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-33661302

RESUMO

Multimeric immunoglobulin-like molecules arose early in vertebrate evolution, yet the unique contributions of multimeric IgM antibodies to infection control are not well understood. This is partially due to the difficulty of distinguishing low-affinity IgM, secreted rapidly by plasmablasts, from high-affinity antibodies derived from later-arising memory cells. We developed a pipeline to express B cell receptors (BCRs) from Plasmodium falciparum-specific IgM+ and IgG+ human memory B cells (MBCs) as both IgM and IgG molecules. BCRs from both subsets were somatically hypermutated and exhibited comparable monomeric affinity. Crystallization of one IgM+ MBC-derived antibody complexed with antigen defined a linear epitope within a conserved Plasmodium protein. In its physiological multimeric state, this antibody displayed exponentially higher antigen binding than a clonally identical IgG monomer, and more effectively inhibited P. falciparum invasion. Forced multimerization of this IgG significantly improved both antigen binding and parasite restriction, underscoring how avidity can alter antibody function. This work demonstrates the potential of high-avidity IgM in both therapeutics and vaccines.


Assuntos
Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Linfócitos B/imunologia , Imunoglobulina M/química , Imunoglobulina M/imunologia , Memória Imunológica , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Multimerização Proteica/imunologia , Adolescente , Afinidade de Anticorpos , Células Cultivadas , Criança , Estudos de Coortes , Epitopos de Linfócito B/imunologia , Feminino , Humanos , Imunoglobulina G/química , Imunoglobulina G/imunologia , Malária Falciparum/sangue , Malária Falciparum/parasitologia , Masculino , Mali , Receptores de Antígenos de Linfócitos B/genética , Receptores de Antígenos de Linfócitos B/imunologia
5.
medRxiv ; 2020 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-32793919

RESUMO

T cells are involved in the early identification and clearance of viral infections and also support the development of antibodies by B cells. This central role for T cells makes them a desirable target for assessing the immune response to SARS-CoV-2 infection. Here, we combined two high-throughput immune profiling methods to create a quantitative picture of the T-cell response to SARS-CoV-2. First, at the individual level, we deeply characterized 3 acutely infected and 58 recovered COVID-19 subjects by experimentally mapping their CD8 T-cell response through antigen stimulation to 545 Human Leukocyte Antigen (HLA) class I presented viral peptides (class II data in a forthcoming study). Then, at the population level, we performed T-cell repertoire sequencing on 1,815 samples (from 1,521 COVID-19 subjects) as well as 3,500 controls to identify shared "public" T-cell receptors (TCRs) associated with SARS-CoV-2 infection from both CD8 and CD4 T cells. Collectively, our data reveal that CD8 T-cell responses are often driven by a few immunodominant, HLA-restricted epitopes. As expected, the T-cell response to SARS-CoV-2 peaks about one to two weeks after infection and is detectable for at least several months after recovery. As an application of these data, we trained a classifier to diagnose SARS-CoV-2 infection based solely on TCR sequencing from blood samples, and observed, at 99.8% specificity, high early sensitivity soon after diagnosis (Day 3-7 = 85.1% [95% CI = 79.9-89.7]; Day 8-14 = 94.8% [90.7-98.4]) as well as lasting sensitivity after recovery (Day 29+/convalescent = 95.4% [92.1-98.3]). These results demonstrate an approach to reliably assess the adaptive immune response both soon after viral antigenic exposure (before antibodies are typically detectable) as well as at later time points. This blood-based molecular approach to characterizing the cellular immune response has applications in clinical diagnostics as well as in vaccine development and monitoring.

6.
Eur J Immunol ; 48(1): 80-86, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28948612

RESUMO

CD4+ tissue resident cells are an important first line of defense against viral infections in the lungs and are critical for promoting the localization of lung resident CD8+ T cells. However, relatively little is known about the signaling programs required for the development of viral-specific CD4+ tissue resident cells in the lungs. Recently, it was shown that signaling through the high affinity IL-2 receptor is required for the differentiation of lung-resident Th2 memory (Trm) cells in a murine model of airway inflammation. We therefore tested if IL-2 signaling is also required for the development of viral antigen-specific CD4+ Th1 cells in the lung after i.n. infection with lymphocytic choriomeningitis virus. These studies demonstrate that Th1 CD4+ T cells also require IL-2 for lung Trm development. Additionally, they show that B cells potently inhibit early Th1 cell lung residency, but are required for the maintenance of a long-lived population of CD4+ Th1 Trm.


Assuntos
Linfócitos B/imunologia , Memória Imunológica/imunologia , Interleucina-2/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Células Th1/imunologia , Células Th2/imunologia , Animais , Antígenos Virais/imunologia , Feminino , Memória Imunológica/genética , Interleucina-2/genética , Pulmão/citologia , Pulmão/imunologia , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/patologia , Coriomeningite Linfocítica/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
7.
Cell Rep ; 17(12): 3193-3205, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-28009289

RESUMO

Many current malaria vaccines target the pre-erythrocytic stage of infection in the liver. However, in malaria-endemic regions, increased blood stage exposure is associated with decreased vaccine efficacy, thereby challenging current vaccine efforts. We hypothesized that pre-erythrocytic humoral immunity is directly disrupted by blood stage infection. To investigate this possibility, we used Plasmodium-antigen tetramers to analyze B cells after infection with either late liver stage arresting parasites or wild-type parasites that progress to the blood stage. Our data demonstrate that immunoglobulin G (IgG) antibodies against the pre-erythrocytic antigen, circumsporozoite protein (CSP), are generated only in response to the attenuated, but not the wild-type, infection. Further analyses revealed that blood stage malaria inhibits CSP-specific germinal center B cell differentiation and modulates chemokine expression. This results in aberrant memory formation and the loss of a rapid secondary B cell response. These data highlight how immunization with attenuated parasites may drive optimal immunity to malaria.


Assuntos
Imunidade Humoral , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Animais , Antígenos de Protozoários/imunologia , Linfócitos B/imunologia , Linfócitos B/parasitologia , Eritrócitos/imunologia , Eritrócitos/parasitologia , Humanos , Imunoglobulina G/imunologia , Fígado/imunologia , Fígado/parasitologia , Vacinas Antimaláricas/uso terapêutico , Malária Falciparum/genética , Malária Falciparum/parasitologia , Plasmodium falciparum/patogenicidade , Vacinação
8.
Immunity ; 45(2): 402-14, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27473412

RESUMO

Humoral immunity consists of pre-existing antibodies expressed by long-lived plasma cells and rapidly reactive memory B cells (MBC). Recent studies of MBC development and function after protein immunization have uncovered significant MBC heterogeneity. To clarify functional roles for distinct MBC subsets during malaria infection, we generated tetramers that identify Plasmodium-specific MBCs in both humans and mice. Long-lived murine Plasmodium-specific MBCs consisted of three populations: somatically hypermutated immunoglobulin M(+) (IgM(+)) and IgG(+) MBC subsets and an unmutated IgD(+) MBC population. Rechallenge experiments revealed that high affinity, somatically hypermutated Plasmodium-specific IgM(+) MBCs proliferated and gave rise to antibody-secreting cells that dominated the early secondary response to parasite rechallenge. IgM(+) MBCs also gave rise to T cell-dependent IgM(+) and IgG(+)B220(+)CD138(+) plasmablasts or T cell-independent B220(-)CD138(+) IgM(+) plasma cells. Thus, even in competition with IgG(+) MBCs, IgM(+) MBCs are rapid, plastic, early responders to a secondary Plasmodium rechallenge and should be targeted by vaccine strategies.


Assuntos
Subpopulações de Linfócitos B/imunologia , Linfócitos B/imunologia , Vacinas Antimaláricas/imunologia , Malária/imunologia , Plasmodium/imunologia , Animais , Afinidade de Anticorpos , Antígenos de Protozoários/imunologia , Células Cultivadas , Epitopos , Feminino , Humanos , Imunidade Humoral , Imunização Secundária , Imunoglobulina M/metabolismo , Memória Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Hipermutação Somática de Imunoglobulina , Linfócitos T/imunologia
9.
Immunity ; 44(1): 155-166, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26750312

RESUMO

Exposure to inhaled allergens generates T helper 2 (Th2) CD4(+) T cells that contribute to episodes of inflammation associated with asthma. Little is known about allergen-specific Th2 memory cells and their contribution to airway inflammation. We generated reagents to understand how endogenous CD4(+) T cells specific for a house dust mite (HDM) allergen form and function. After allergen exposure, HDM-specific memory cells persisted as central memory cells in the lymphoid organs and tissue-resident memory cells in the lung. Experimental blockade of lymphocyte migration demonstrated that lung-resident cells were sufficient to induce airway hyper-responsiveness, which depended upon CD4(+) T cells. Investigation into the differentiation of pathogenic Trm cells revealed that interleukin-2 (IL-2) signaling was required for residency and directed a program of tissue homing migrational cues. These studies thus identify IL-2-dependent resident Th2 memory cells as drivers of lung allergic responses.


Assuntos
Asma/imunologia , Memória Imunológica/imunologia , Interleucina-2/imunologia , Pulmão/imunologia , Células Th2/imunologia , Alérgenos/imunologia , Animais , Antígenos de Dermatophagoides/imunologia , Diferenciação Celular/imunologia , Separação Celular , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pyroglyphidae/imunologia
10.
PLoS Pathog ; 11(5): e1004855, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25974076

RESUMO

Malaria, caused by Plasmodium parasite infection, continues to be one of the leading causes of worldwide morbidity and mortality. Development of an effective vaccine has been encumbered by the complex life cycle of the parasite that has distinct pre-erythrocytic and erythrocytic stages of infection in the mammalian host. Historically, malaria vaccine development efforts have targeted each stage in isolation. An ideal vaccine, however, would target multiple life cycle stages with multiple arms of the immune system and be capable of eliminating initial infection in the liver, the subsequent blood stage infection, and would prevent further parasite transmission. We have previously shown that immunization of mice with Plasmodium yoelii genetically attenuated parasites (GAP) that arrest late in liver stage development elicits stage-transcending protection against both a sporozoite challenge and a direct blood stage challenge. Here, we show that this immunization strategy engenders both T- and B-cell responses that are essential for stage-transcending protection, but the relative importance of each is determined by the host genetic background. Furthermore, potent anti-blood stage antibodies elicited after GAP immunization rely heavily on FC-mediated functions including complement fixation and FC receptor binding. These protective antibodies recognize the merozoite surface but do not appear to recognize the immunodominant merozoite surface protein-1. The antigen(s) targeted by stage-transcending immunity are present in both the late liver stages and blood stage parasites. The data clearly show that GAP-engendered protective immune responses can target shared antigens of pre-erythrocytic and erythrocytic parasite life cycle stages. As such, this model constitutes a powerful tool to identify novel, protective and stage-transcending T and B cell targets for incorporation into a multi-stage subunit vaccine.


Assuntos
Imunização , Fígado/imunologia , Vacinas Antimaláricas/imunologia , Malária/imunologia , Animais , Linfócitos B/imunologia , Feminino , Imunização/métodos , Malária/parasitologia , Vacinas Antimaláricas/genética , Camundongos , Plasmodium yoelii/crescimento & desenvolvimento , Esporozoítos/imunologia , Linfócitos T/imunologia , Vacinação
11.
Hum Vaccin Immunother ; 10(10): 2903-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25483648

RESUMO

Although recent control measures have significantly reduced malaria cases and deaths in many endemic areas, an effective vaccine will be essential to eradicate this parasitic disease. Malaria vaccine strategies developed to date focus on different phases of the parasite's complex life cycle in the human host and mosquito vector, and include both subunit-based and whole-parasite vaccines. This review focuses on the 3 live-attenuated malaria vaccination strategies that have been tested in humans to date, and discusses their progress, challenges and the immune correlates of protection that have been identified.


Assuntos
Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Malária Vivax/prevenção & controle , Plasmodium falciparum/imunologia , Plasmodium vivax/imunologia , Anticorpos Antiprotozoários/imunologia , Humanos , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Malária Vivax/imunologia , Malária Vivax/parasitologia , Proteínas de Protozoários/imunologia , Esporozoítos/imunologia , Vacinação , Vacinas Atenuadas/imunologia
12.
Infect Immun ; 82(12): 5143-53, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25267837

RESUMO

Understanding protective immunity to malaria is essential for the design of an effective vaccine to prevent the large number of infections and deaths caused by this parasitic disease. To date, whole-parasite immunization with attenuated parasites is the most effective method to confer sterile protection against malaria infection in clinical trials. Mouse model studies have highlighted the essential role that CD8(+) T cells play in protection against preerythrocytic stages of malaria; however, there is mounting evidence that antibodies are also important in these stages. Here, we show that experimental immunization of mice with Plasmodium yoelii fabb/f(-) (Pyfabb/f(-)), a genetically attenuated rodent malaria parasite that arrests late in the liver stage, induced functional antibodies that inhibited hepatocyte invasion in vitro and reduced liver-stage burden in vivo. These antibodies were sufficient to induce sterile protection from challenge by P. yoelii sporozoites in the absence of T cells in 50% of mice when sporozoites were administered by mosquito bite but not when they were administered by intravenous injection. Moreover, among mice challenged by mosquito bite, a higher proportion of BALB/c mice than C57BL/6 mice developed sterile protection (62.5% and 37.5%, respectively). Analysis of the antibody isotypes induced by immunization with Pyfabb/f(-) showed that, overall, BALB/c mice developed an IgG1-biased response, whereas C57BL/6 mice developed an IgG2b/c-biased response. Our data demonstrate for the first time that antibodies induced by experimental immunization of mice with a genetically attenuated rodent parasite play a protective role during the preerythrocytic stages of malaria. Furthermore, they highlight the importance of considering both the route of challenge and the genetic background of the mouse strains used when interpreting vaccine efficacy studies in animal models of malaria infection.


Assuntos
Anticorpos Antiprotozoários/sangue , Imunização/métodos , Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Plasmodium yoelii/imunologia , Experimentação Animal , Animais , Feminino , Imunoglobulina G/sangue , Malária/imunologia , Vacinas Antimaláricas/administração & dosagem , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia
13.
PLoS One ; 9(7): e102225, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25025375

RESUMO

Pre-erythrocytic malaria vaccines, including those based on whole-parasite approaches, have shown protective efficacy in animal and human studies. However few pre-erythocytic antigens other than the immunodominant circumsporozoite protein (CSP) have been studied in depth with the goal of developing potent subunit malaria vaccines that are suited for use in endemic areas. Here we describe a novel technique to identify pre-erythrocytic malaria antigens that contribute to protection elicited by whole-parasite vaccination in the mouse model. Our approach combines immunization with genetically attenuated parasites and challenge with DNA plasmids encoding for potential protective pre-erythrocytic malaria antigens as luciferase fusions by hydrodynamic tail vein injection. After optimizing the technique, we first showed that immunization with Pyfabb/f-, a P. yoelii genetically attenuated parasite, induces killing of CSP-presenting hepatocytes. Depletion of CD8+ but not CD4+ T cells diminished the killing of CSP-expressing hepatocytes, indicating that killing is CD8+ T cell-dependent. Finally we showed that the use of heterologous prime/boost immunization strategies that use genetically attenuated parasites and DNA vaccines enabled the characterization of a novel pre-erythrocytic antigen, Tmp21, as a contributor to Pyfabb/f- induced protection. This technique will be valuable for identification of potentially protective liver stage antigens and has the potential to contribute to the understanding of immunity elicited by whole parasite vaccination, as well as the development of effective subunit malaria vaccines.


Assuntos
Antígenos de Protozoários/imunologia , Hepatócitos/imunologia , Vacinas Antimaláricas/imunologia , Plasmodium/imunologia , Animais , Feminino , Humanos , Vacinas Antimaláricas/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos
14.
J Immunol ; 193(3): 1268-77, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24958899

RESUMO

Sterile protection against malaria infection can be achieved through vaccination of mice and humans with whole Plasmodium spp. parasites. One such method, known as infection-treatment-vaccination (ITV), involves immunization with wild type sporozoites (spz) under drug coverage. In this work, we used the different effects of antimalarial drugs chloroquine (CQ) and artesunate (AS) on blood stage (BS) parasites to dissect the stage-specific immune responses in mice immunized with Plasmodium yoelii spz under either drug, as well as their ability to protect mice against challenge with spz or infected RBCs (iRBCs). Whereas CQ-ITV induced sterile protection against challenge with both spz and iRBCs, AS-ITV only induced sterile protection against spz challenge. Importantly, AS-ITV delayed the onset of BS infection, indicating that both regimens induced cross-stage immunity. Moreover, both CQ- and AS-ITV induced CD8(+) T cells in the liver that eliminated malaria-infected hepatocytes in vitro, as well as Abs that recognized pre-erythrocytic parasites. Sera from both groups of mice inhibited spz invasion of hepatocytes in vitro, but only CQ-ITV induced high levels of anti-BS Abs. Finally, passive transfer of sera from CQ-ITV-treated mice delayed the onset of erythrocytic infection in the majority of mice challenged with P. yoelii iRBCs. Besides constituting the first characterization, to our knowledge, of AS-ITV as a vaccination strategy, our data show that ITV strategies that lead to subtle differences in the persistence of parasites in the blood enable the characterization of the resulting immune responses, which will contribute to future research in vaccine design and malaria interventions.


Assuntos
Artemisininas/administração & dosagem , Cloroquina/administração & dosagem , Eritrócitos/imunologia , Eritrócitos/parasitologia , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/imunologia , Malária/imunologia , Plasmodium yoelii/imunologia , Animais , Anopheles/imunologia , Anticorpos Antiprotozoários/biossíntese , Anticorpos Antiprotozoários/sangue , Antimaláricos/administração & dosagem , Antimaláricos/uso terapêutico , Artemisia/imunologia , Artemisininas/uso terapêutico , Artesunato , Cloroquina/uso terapêutico , Eritrócitos/efeitos dos fármacos , Feminino , Malária/sangue , Malária/prevenção & controle , Vacinas Antimaláricas/sangue , Camundongos , Camundongos Endogâmicos BALB C , Fitoterapia/métodos , Plasmodium yoelii/efeitos dos fármacos , Esporozoítos/efeitos dos fármacos , Esporozoítos/imunologia , Esporozoítos/transplante , Fatores de Tempo , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/uso terapêutico
15.
Vaccine ; 32(19): 2135-8, 2014 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-24582635

RESUMO

Whole-parasite malaria vaccines have shown promise in clinical trials. We recently reported the first human trial of a malaria vaccine based on Plasmodium falciparum genetically attenuated parasites (PfGAP). Herein we report for the first time that PfGAP induces prolonged functional humoral responses in humans. Six volunteers were exposed to 5 bites of PfGAP-infected mosquitoes followed by approximately 200 bites. Plasma collected from all volunteers 3 months after the last exposure efficiently inhibits invasion of hepatocytes by P. falciparum sporozoites. The level of inhibition observed is comparable to that attained using plasma collected after 4-5 intravenously administrations of high numbers of irradiated sporozoites, validating the potential of PfGAP malaria vaccines. Our data highlight the role of antibody responses in pre-erythrocytic stages of human malaria, and suggests that to be protective, malaria vaccines might need to elicit long-lasting functional antibodies in addition to cellular responses.


Assuntos
Anticorpos Antiprotozoários/sangue , Hepatócitos/parasitologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Linhagem Celular , Humanos , Imunidade Humoral , Plasmodium falciparum/genética , Esporozoítos/imunologia , Vacinas Atenuadas/imunologia
16.
Infect Immun ; 81(4): 1031-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23319559

RESUMO

Placental malaria (PM) is characterized by infected erythrocytes (IEs) that selectively bind to chondroitin sulfate A (CSA) and sequester in placental tissue. Variant surface antigen 2-CSA (VAR2CSA), a Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) protein family member, is expressed on the surface of placental IEs and mediates adherence to CSA on the surface of syncytiotrophoblasts. This transmembrane protein contains 6 Duffy binding-like (DBL) domains which might contribute to the specific adhesive properties of IEs. Here, we use laboratory isolate 3D7 VAR2CSA DBL domains expressed in Escherichia coli to generate antibodies specific for this protein. Flow cytometry results showed that antibodies generated against DBL4ε, DBL5ε, DBL6ε, and tandem double domains of DBL4-DBL5 and DBL5-DBL6 all bind to placental parasite isolates and to lab strains selected for CSA binding but do not bind to children's parasites. Antisera to DBL4ε and to DBL5ε inhibit maternal IE binding to placental tissue in a manner comparable to that for plasma collected from multigravid women. These antibodies also inhibit binding to CSA of several field isolates derived from pregnant women, while antibodies to double domains do not enhance the functional immune response. These data support DBL4ε and DBL5ε as vaccine candidates for pregnancy malaria and demonstrate that E. coli is a feasible tool for the large-scale manufacture of a vaccine based on these VAR2CSA domains.


Assuntos
Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/imunologia , Sulfatos de Condroitina/imunologia , Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Placenta/parasitologia , Adulto , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/genética , Adesão Celular , Sulfatos de Condroitina/genética , Sulfatos de Condroitina/metabolismo , Escherichia coli/genética , Feminino , Expressão Gênica , Humanos , Recém-Nascido , Vacinas Antimaláricas/administração & dosagem , Plasmodium falciparum/genética , Plasmodium falciparum/imunologia , Gravidez , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
17.
PLoS One ; 3(1): e1375, 2008 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-18167536

RESUMO

MAPK signaling cascades are evolutionally conserved. The bacterial effector, YopJ, uses the unique activity of Ser/Thr acetylation to inhibit the activation of the MAPK kinase (MKK) and prevent activation by phosphorylation. YopJ is also able to block yeast MAPK signaling pathways using this mechanism. Based on these observations, we performed a genetic screen to isolate mutants in the yeast MKK, Pbs2, that suppress YopJ inhibition. One suppressor contains a mutation in a conserved tyrosine residue and bypasses YopJ inhibition by increasing the basal activity of Pbs2. Mutations on the hydrophobic face of the conserved G alpha-helix in the kinase domain prevent both binding and acetylation by YopJ. Corresponding mutants in human MKKs showed that they are conserved not only structurally, but also functionally. These studies reveal a conserved binding site found on the superfamily of MAPK kinases while providing insight into the molecular interactions required for YopJ inhibition.


Assuntos
Proteínas de Bactérias/fisiologia , Sistema de Sinalização das MAP Quinases , Acetilação , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mutação , Ligação Proteica , Conformação Proteica , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Yersinia pestis/fisiologia
18.
J Biol Chem ; 283(22): 15127-33, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18223255

RESUMO

Human T-cell leukemia virus type-I expresses Tax, a 40-kDa oncoprotein that activates IkappaB kinase (IKK), resulting in constitutive activation of NFkappaB. Herein, we have developed an in vitro signaling assay to analyze IKK complex activation by recombinant Tax. Using this assay in combination with reporter assays, we demonstrate that Tax-mediated activation of IKK is independent of phosphatases. We show that sustained activation of the Tax-mediated activation of the NFkappaB pathway is dependent on an intact Hsp90-IKK complex. By acetylating and thereby preventing activation of the IKK complex by the Yersinia effector YopJ, we demonstrate that Tax-mediated activation of the IKK complex requires a phosphorylation step. Our characterization of an in vitro signaling assay system for the mechanism of Tax-mediated activation of the IKK complex with a variety of mutants and inhibitors results in a working model for the biochemical mechanism of Tax-induced activation.


Assuntos
Proteínas de Choque Térmico HSP90/metabolismo , Vírus Linfotrópico T Tipo 1 Humano/metabolismo , Quinase I-kappa B/metabolismo , Complexos Multiproteicos/metabolismo , NF-kappa B/metabolismo , Acetilação/efeitos dos fármacos , Proteínas de Bactérias/farmacologia , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Produtos do Gene tax , Proteínas de Choque Térmico HSP90/genética , Vírus Linfotrópico T Tipo 1 Humano/genética , Humanos , Quinase I-kappa B/genética , Complexos Multiproteicos/genética , NF-kappa B/genética , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
J Biol Chem ; 282(47): 34299-305, 2007 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-17881352

RESUMO

The bacterial pathogen Vibrio parahemeolyticus manipulates host signaling pathways during infections by injecting type III effectors. One of these effectors, Vibrio outer protein A (VopA), inhibits MAPK signaling via a novel mechanism, distinct from those described for other bacterial toxins, that disrupts this signaling pathway. VopA is an acetyltransferase that potently inhibits MAPK signaling pathways not only by preventing the activation of MAPK kinases (MKKs) but also by inhibiting the activity of activated MKKs. VopA acetylates a conserved lysine found in the catalytic loop of all kinases and blocks the binding of ATP, but not ADP, on the MKKs, resulting in an inactive phosphorylated kinase. Acetylation of this conserved lysine inhibits kinase activity by a new mechanism of regulation that has not been observed previously. Identifying the target of VopA reveals a way that the reversible post-translational modification of lysine acetylation can be used to regulate the activity of an enzyme.


Assuntos
Aminoácido N-Acetiltransferase/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Toxinas Bacterianas/metabolismo , Sistema de Sinalização das MAP Quinases , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Vibrioses/enzimologia , Vibrio parahaemolyticus/enzimologia , Acetilação/efeitos dos fármacos , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Aminoácido N-Acetiltransferase/farmacologia , Animais , Proteínas da Membrana Bacteriana Externa/farmacologia , Toxinas Bacterianas/farmacologia , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , Lisina/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Vibrio parahaemolyticus/patogenicidade
20.
Science ; 312(5777): 1211-4, 2006 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-16728640

RESUMO

Yersinia species use a variety of type III effector proteins to target eukaryotic signaling systems. The effector YopJ inhibits mitogen-activated protein kinase (MAPK) and the nuclear factor kappaB (NFkappaB) signaling pathways used in innate immune response by preventing activation of the family of MAPK kinases (MAPKK). We show that YopJ acted as an acetyltransferase, using acetyl-coenzyme A (CoA) to modify the critical serine and threonine residues in the activation loop of MAPKK6 and thereby blocking phosphorylation. The acetylation on MAPKK6 directly competed with phosphorylation, preventing activation of the modified protein. This covalent modification may be used as a general regulatory mechanism in biological signaling.


Assuntos
Proteínas de Bactérias/metabolismo , Quinase I-kappa B/metabolismo , MAP Quinase Quinase 6/metabolismo , Yersinia/metabolismo , Acetilcoenzima A/metabolismo , Acetilação , Acetiltransferases/metabolismo , Domínio Catalítico , Linhagem Celular , Sistema Livre de Células , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , MAP Quinase Quinase 6/química , Sistema de Sinalização das MAP Quinases , NF-kappa B/metabolismo , Fosforilação , Proteínas Recombinantes/metabolismo , Serina/metabolismo , Transdução de Sinais , Treonina/metabolismo , Yersinia/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA