Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
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
2.
Infect Immun ; 89(11): e0016521, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34310889

RESUMO

Preerythrocytic vaccines prevent malaria by targeting parasites in the clinically silent sporozoite and liver stages and preventing progression to the virulent blood stages. The leading preerythrocytic vaccine, RTS,S/AS01E (Mosquirix), entered implementation programs in 2019 and targets the major sporozoite surface antigen, circumsporozoite protein (CSP). However, in phase III clinical trials, RTS,S conferred partial protection with limited durability, indicating a need to improve CSP-based vaccination. Previously, we identified highly expressed liver-stage proteins that could potentially be used in combination with CSP; they are referred to as preerythrocytic vaccine antigens (PEVAs). Here, we developed heterologous prime-boost CSP vaccination models to confer partial sterilizing immunity against Plasmodium yoelii (protein prime-adenovirus 5 [Ad5] boost) and Plasmodium berghei (DNA prime-Ad5 boost) in mice. When combined as individual antigens with P. yoelii CSP (PyCSP), three of eight P. yoelii PEVAs significantly enhanced sterile protection against sporozoite challenge, compared to PyCSP alone. Similar results were obtained when three P. berghei PEVAs and P. berghei CSP were combined in a single vaccine regimen. In general, PyCSP antibody responses were similar after CSP alone versus CSP plus PEVA vaccinations. Both P. yoelii and P. berghei CSP plus PEVA combination vaccines induced robust CD8+ T cell responses, including signature gamma interferon (IFN-γ) increases. In the P. berghei model system, IFN-γ responses were significantly higher in hepatic versus splenic CD8+ T cells. The addition of novel antigens may enhance the degree and duration of sterile protective immunity conferred by a human vaccine such as RTS,S.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Proteínas de Protozoários/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Interferon gama/biossíntese , Ativação Linfocitária , Malária/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Vacinação
3.
J Biol Chem ; 291(38): 19913-22, 2016 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-27432885

RESUMO

Development of a Plasmodium falciparum (Pf) transmission blocking vaccine (TBV) has the potential to significantly impact malaria control. Antibodies elicited against sexual stage proteins in the human bloodstream are taken up with the blood meal of the mosquitoes and inactivate parasite development in the mosquito. In a phase 1 trial, a leading TBV identified as Pfs25-EPA/Alhydrogel® appeared safe and immunogenic, however, the level of Pfs25-specific antibodies were likely too low for an effective vaccine. Pfs230, a 230-kDa sexual stage protein expressed in gametocytes is an alternative vaccine candidate. A unique 6-cysteine-rich domain structure within Pfs230 have thwarted its recombinant expression and characterization for clinical evaluation for nearly a quarter of a century. Here, we report on the identification, biochemical, biophysical, and immunological characterization of recombinant Pfs230 domains. Rabbit antibodies generated against recombinant Pfs230 domains blocked mosquito transmission of a laboratory strain and two field isolates using an ex vivo assay. A planned clinical trial of the Pfs230 vaccine is a significant step toward the potential development of a transmission blocking vaccine to eliminate malaria.


Assuntos
Antígenos de Protozoários/química , Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/química , Vacinas Antimaláricas/imunologia , Plasmodium falciparum/química , Plasmodium falciparum/imunologia , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/genética , Antígenos de Protozoários/farmacologia , Humanos , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/farmacologia , Malária Falciparum/genética , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Plasmodium falciparum/genética , Domínios Proteicos , Proteínas de Protozoários/genética , Proteínas de Protozoários/farmacologia , Coelhos
4.
J Infect Dis ; 214(4): 577-86, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27190180

RESUMO

BACKGROUND: Placental malaria is caused by Plasmodium falciparum-infected erythrocytes (IEs) that surface-express VAR2CSA and bind chondroitin sulfate A. The inflammatory response to placenta-sequestered parasites is associated with poor pregnancy outcomes, and protection may be mediated in part by VAR2CSA antibodies that block placental IE adhesion. METHODS: In this study, we used a new approach to assess VAR2CSA domains for functional epitopes recognized by naturally acquired antibodies. Antigen-specific immunoglobulin (Ig) G targeting Duffy binding-like (DBL) domains from different alleles were sequentially purified from plasma pooled from multigravid women and then characterized using enzyme-linked immunosorbent assay, flow cytometry, and antiadhesion assays. RESULTS: Different DBL domain-specific IgGs could react to homologous as well as heterologous antigens and parasites, suggesting that conserved epitopes are shared between allelic variants. Homologous blocking of IE binding was observed with ID1-DBL2-ID2a-, DBL4-, and DBL5-specific IgG (range, 42%-75%), whereas partial cross-inhibition activity was observed with purified IgG specific to ID1-DBL2-ID2a and DBL4 antigens. Plasma retained broadly neutralizing activity after complete depletion of these VAR2CSA specificities. CONCLUSIONS: Broadly neutralizing antibodies of multigravidae are not depleted on VAR2CSA recombinant antigens, and hence development of VAR2CSA vaccines based on a single construct and variant might induce antibodies with limited broadly neutralizing activity.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/imunologia , Malária Falciparum/imunologia , Doenças Placentárias/imunologia , Plasmodium falciparum/imunologia , Adesão Celular , Ensaio de Imunoadsorção Enzimática , Mapeamento de Epitopos , Epitopos de Linfócito B , Feminino , Citometria de Fluxo , Humanos , Vacinas Antimaláricas/imunologia , Gravidez
5.
Infect Immun ; 83(10): 3771-80, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26169272

RESUMO

The extended rod-like Plasmodium falciparum circumsporozoite protein (CSP) is comprised of three primary domains: a charged N terminus that binds heparan sulfate proteoglycans, a central NANP repeat domain, and a C terminus containing a thrombospondin-like type I repeat (TSR) domain. Only the last two domains are incorporated in RTS,S, the leading malaria vaccine in phase 3 trials that, to date, protects about 50% of vaccinated children against clinical disease. A seroepidemiological study indicated that the N-terminal domain might improve the efficacy of a new CSP vaccine. Using a panel of CSP-specific monoclonal antibodies, well-characterized recombinant CSPs, label-free quantitative proteomics, and in vitro inhibition of sporozoite invasion, we show that native CSP is N-terminally processed in the mosquito host and undergoes a reversible conformational change to mask some epitopes in the N- and C-terminal domains until the sporozoite interacts with the liver hepatocyte. Our findings show the importance of understanding processing and the biophysical change in conformation, possibly due to a mechanical or molecular signal, and may aid in the development of a new CSP vaccine.


Assuntos
Malária Falciparum/parasitologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia , Esporozoítos/imunologia , Animais , Anopheles/parasitologia , Anticorpos Antiprotozoários/imunologia , Epitopos/química , Epitopos/genética , Epitopos/imunologia , Hepatócitos/imunologia , Hepatócitos/parasitologia , Humanos , Malária Falciparum/imunologia , Plasmodium falciparum/química , Plasmodium falciparum/genética , Plasmodium falciparum/crescimento & desenvolvimento , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas de Protozoários/genética , Esporozoítos/química , Esporozoítos/crescimento & desenvolvimento
6.
Proc Natl Acad Sci U S A ; 109(17): 6692-7, 2012 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-22493233

RESUMO

The s48/45 domain was first noted in Plasmodium proteins more than 15 y ago. Previously believed to be unique to Plasmodium, the s48/45 domain is present in other aconoidasidans. In Plasmodium, members of the s48/45 family of proteins are localized on the surface of the parasite in different stages, mostly by glycosylphosphatydylinositol-anchoring. Members such as P52 and P36 seem to play a role in invasion of hepatocytes, and Pfs230 and Pfs48/45 are involved in fertilization in the sexual stages and have been consistently studied as targets of transmission-blocking vaccines for years. In this report, we present the molecular structure for the s48/45 domain corresponding to the C-terminal domain of the blood-stage protein Pf12 from Plasmodium falciparum, obtained by NMR. Our results indicate that this domain is a ß-sandwich formed by two sheets with a mixture of parallel and antiparallel strands. Of the six conserved cysteines, two pairs link the ß-sheets by two disulfide bonds, and the third pair forms a bond outside the core. The structure of the s48/45 domain conforms well to the previously defined surface antigen 1 (SAG1)-related-sequence (SRS) fold observed in the SAG family of surface antigens found in Toxoplasma gondii. Despite extreme sequence divergence, remarkable spatial conservation of one of the disulfide bonds is observed, supporting the hypothesis that the domains have evolved from a common ancestor. Furthermore, a homologous domain is present in ephrins, raising the possibility that the precursor of the s48/45 and SRS domains emerged from an ancient transfer to Apicomplexa from metazoan hosts.


Assuntos
Cisteína/química , Plasmodium falciparum/química , Animais , Ressonância Magnética Nuclear Biomolecular , Proteínas de Protozoários/química
7.
Vet Comp Orthop Traumatol ; 37(2): 64-73, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37967844

RESUMO

OBJECTIVE: The aim of this study was to use computed tomography (CT) images obtained from English springer spaniels (ESS) with different sizes of humeral intracondylar fissure (HIF) to describe the typical shape, origin and a possible propagation pattern of HIF in this breed. STUDY DESIGN: It is a retrospective analysis of 32 elbow CT from 27 ESS with incomplete HIF. Measurements included HIF articular surface length, centre of HIF at articular surface relative to the caudal edge of the supratrochlear foramen (CHIF), HIF depth and sagittal area of fissure. Measurement of isthmus area and diameter was obtained for each elbow. Humeral intracondylar fissure measurements were analysed as proportions of the isthmus. For parts of analysis, elbows were grouped by HIF area as a percentage of isthmus area (%HIF) into less than 20% (n = 10), 20 to less than 40% (n = 8), 40 to less than 60% (n = 9) and 60 to less than 90% (n = 5). RESULTS: The mean isthmus diameter was 12.31 mm (range: 10.96-13.69 mm). Mean CHIF for %HIF groups less than 20%, 20 to less than 40%, 40 to less than 60% and 60 to less than 90% were 57, 74, 86 and 96 degrees, respectively. The less than 20% group was significantly lower than 20 to less than 40% group (p = 0.035) and 40 to less than 60% and 60 to less than 90% groups (p < 0.001); the 20 to less than 40% group was significantly lower than the 60 to less than 90% group (p = 0.015). Humeral intracondylar fissure articular surface length increased in a sigmoidal fashion relative to %HIF, corresponding to segmental enlargement of the fissure as %HIF increases. CONCLUSION: In ESS, HIF typically originates approximately 57 degrees caudal to the supratrochlear foramen in the sagittal plane and may propagate in a segmental fashion with lesser propagation through the proximal intracondylar region.


Assuntos
Doenças do Cão , Úmero , Cães , Animais , Estudos Retrospectivos , Tomografia Computadorizada por Raios X/veterinária
8.
Infect Immun ; 81(5): 1491-501, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23429537

RESUMO

Apical membrane antigen 1 (AMA1) is a leading vaccine candidate, but the allelic polymorphism is a stumbling block for vaccine development. We previously showed that a global set of AMA1 haplotypes could be grouped into six genetic populations. Using this information, six recombinant AMA1 proteins representing each population were produced. Rabbits were immunized with either a single recombinant AMA1 protein or mixtures of recombinant AMA1 proteins (mixtures of 4, 5, or 6 AMA1 proteins). Antibody levels were measured by enzyme-linked immunosorbent assay (ELISA), and purified IgG from each rabbit was used for growth inhibition assay (GIA) with 12 different clones of parasites (a total of 108 immunogen-parasite combinations). Levels of antibodies to all six AMA1 proteins were similar when the antibodies were tested against homologous antigens. When the percent inhibitions in GIA were plotted against the number of ELISA units measured with homologous AMA1, all data points followed a sigmoid curve, regardless of the immunogen. In homologous combinations, there were no differences in the percent inhibition between the single-allele and allele mixture groups. However, all allele mixture groups showed significantly higher percent inhibition than the single-allele groups in heterologous combinations. The 5-allele-mixture group showed significantly higher inhibition to heterologous parasites than the 4-allele-mixture group. On the other hand, there was no difference between the 5- and 6-allele-mixture groups. These data indicate that mixtures with a limited number of alleles may cover a majority of the parasite population. In addition, using the data from 72 immunogen-parasite combinations, we mathematically identified 13 amino acid polymorphic sites which significantly impact GIA activities. These results could be a foundation for the rational design of a future AMA1 vaccine.


Assuntos
Alelos , Especificidade de Anticorpos/imunologia , Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Proteínas de Membrana/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/genética , Ensaio de Imunoadsorção Enzimática , Imunoglobulina G/imunologia , Proteínas de Membrana/genética , Plasmodium falciparum/genética , Plasmodium falciparum/crescimento & desenvolvimento , Proteínas de Protozoários/genética , Coelhos , Proteínas Recombinantes/imunologia
9.
Infect Immun ; 81(6): 1984-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23509152

RESUMO

Pfs25 is a leading candidate for a malaria transmission-blocking vaccine whose potential has been demonstrated in a phase 1 trial with recombinant Pfs25 formulated with Montanide ISA51. Because of limited sequence polymorphism, the anti-Pfs25 antibodies induced by this vaccine are likely to have transmission-blocking or -reducing activity against most, if not all, field isolates. To test this hypothesis, we evaluated transmission-blocking activities by membrane feeding assay of anti-Pfs25 plasma from the Pfs25/ISA51 phase 1 trial against Plasmodium falciparum parasites from patients in two different geographical regions of the world, Thailand and Burkina Faso. In parallel, parasite isolates from these patients were sequenced for the Pfs25 gene and genotyped for seven microsatellites. The results indicate that despite different genetic backgrounds among parasite isolates, the Pfs25 sequences are highly conserved, with a single nonsynonymous nucleotide polymorphism detected in 1 of 41 patients in Thailand and Burkina Faso. The anti-Pfs25 immune plasma had significantly higher transmission-reducing activity against parasite isolates from the two geographical regions than the nonimmune controls (P < 0.0001).


Assuntos
Anticorpos Antiprotozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/transmissão , Plasmodium falciparum/genética , Proteínas de Protozoários/imunologia , Animais , Anopheles/parasitologia , Burkina Faso/epidemiologia , Variação Genética , Humanos , Soros Imunes/imunologia , Malária Falciparum/epidemiologia , Malária Falciparum/parasitologia , Repetições de Microssatélites , Plasmodium falciparum/imunologia , Tailândia/epidemiologia
10.
NPJ Vaccines ; 8(1): 56, 2023 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-37061547

RESUMO

Development of a malaria vaccine that blocks transmission of different parasite stages to humans and mosquitoes is considered critical for elimination efforts. A vaccine using Pfs25, a protein on the surface of zygotes and ookinetes, is under investigation as a transmission-blocking vaccine (TBV) that would interrupt parasite passage from mosquitoes to humans. The most extensively studied Pfs25 TBVs use Pichia pastoris-produced recombinant forms of Pfs25, chemically conjugated to a recombinant carrier protein, ExoProtein A (EPA). The recombinant form of Pfs25 first used in humans was identified as Pfs25H, which contained a total of 14 heterologous amino acid residues located at the amino- and carboxyl-termini including a His6 affinity tag. A second recombinant Pfs25, identified as Pfs25M, was produced to remove the heterologous amino acid residues and conjugated to EPA (Pfs25M-EPA). Here, monomeric Pfs25M was characterized biochemically and biophysically for identity, purity, and integrity including protein structure to assess its comparability with Pfs25H. Although the biological activities of Pfs25H and Pfs25M, whether generated by monomeric forms or conjugated nanoparticles, appeared similar, fine-mapping studies with two transmission-blocking monoclonal antibodies detected structural and immunological differences. In addition, evaluation of antisera generated against conjugated Pfs25H or Pfs25M nanoparticles in nonhuman primates identified polyclonal IgG that recognized these structural differences.

11.
Lancet Infect Dis ; 23(11): 1266-1279, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37499679

RESUMO

BACKGROUND: Malaria transmission-blocking vaccines target mosquito-stage parasites and will support elimination programmes. Gamete vaccine Pfs230D1-EPA/Alhydrogel induced superior activity to zygote vaccine Pfs25-EPA/Alhydrogel in malaria-naive US adults. Here, we compared these vaccines in malaria-experienced Malians. METHODS: We did a pilot safety study then double-blind, block-randomised, comparator-controlled main-phase trial in malaria-intense Bancoumana, Mali. 18-50-year-old healthy non-pregnant, non-breastfeeding consenting adult residents were randomly assigned (1:1:1:1) to receive four doses at months 0, 1, 4·5, and 16·5 of either 47 µg Pfs25, 40 µg Pfs230D1 or comparator (Twinrix or Menactra)-all co-administered with normal saline for blinding-or 47 µg Pfs25 plus 40 µg Pfs230D1 co-administered. We documented safety and tolerability (primary endpoint in the as-treated populations) and immunogenicity (secondary endpoint in the as-treated populations: ELISA, standard-membrane-feeding assay, and mosquito direct skin feed assay). This trial is registered at ClinicalTrials.gov, NCT02334462. FINDINGS: Between March 19, and June 2, 2015, we screened 471 individuals. Of 225 enrolled for the pilot and main cohorts, we randomly assigned 25 participants to pilot safety cohort groups of five (20%) to receive a two-dose series of Pfs25-EPA/Alhydrogel (16 µg), Pfs230D1-EPA/Alhydrogel (15 µg) or comparator, followed by Pfs25-EPA/Alhydrogel (16 µg) plus Pfs230D1-EPA/Alhydrogel (15 µg) or comparator plus saline. For the main cohort, we enrolled 200 participants between May 11 and June 2, 2015, to receive a four-dose series of 47 µg Pfs25-EPA/Alhydrogel plus saline (n=50 [25%]; Pfs25), 40 µg Pfs230D1-EPA/Alhydrogel plus saline (n=49 [25%]; Pfs230D1), 47 µg Pfs25-EPA/Alhydrogel plus 40 µg Pfs230D1-EPA/Alhydrogel (n=50 [25%]; Pfs25 plus Pfs230D1), or comparator (Twinrix or Menactra) plus saline (n=51 [25%]). Vaccinations were well tolerated in the pilot safety and main phases. Most vaccinees became seropositive after two Pfs230D1 or three Pfs25 doses; peak titres increased with each dose thereafter (Pfs230D1 geometric mean: 77·8 [95% CI 56·9-106·3], 146·4 [108·3-198·0], and 410·2 [301·6-558·0]; Pfs25 geometric mean 177·7 [130·3-242·4] and 315·7 [209·9-474·6]). Functional activity (mean peak transmission-reducing activity) appeared for Pfs230D1 (74·5% [66·6-82·5]) and Pfs25 plus Pfs230D1 (68·6% [57·3-79·8]), after the third dose and after the fourth dose (88·9% [81·7-96·2] for Pfs230D1 and 85·0% [78·4-91·5] Pfs25 plus Pfs230D1) but not for Pfs25 (58·2% [49·1-67·3] after the third dose and 58·2% [48·5-67·9] after the fourth dose). Pfs230D1 transmission-reducing activity (73·7% [64·1-83·3]) persisted 10 weeks after the fourth dose. Transmission-reducing activity of 80% was estimated at 1659 ELISA units for Pfs25, 218 for Pfs230D1, and 223 for Pfs230D1 plus Pfs25. After 3850 direct skin feed assays, 35 participants (12 Pfs25, eight Pfs230D1, five Pfs25 plus Pfs230D1, and ten comparator) had transmitted parasites at least once. The proportion of positive assays in vaccine groups (Pfs25 33 [3%] of 982 [-0·013 to 0·014], Pfs230D1 22 [2%] of 954 [-0·005 to 0·027], and combination 11 [1%] of 940 [-0·024 to 0·002]) did not differ from that of the comparator (22 [2%] of 974), nor did Pfs230D1 and combination groups differ (-0·024 to 0·001). INTERPRETATION: Pfs230D1 but not Pfs25 vaccine induces durable serum functional activity in Malian adults. Direct skin feed assays detect parasite transmission to mosquitoes but increased event rates are needed to assess vaccine effectiveness. FUNDING: Intramural Research Program of the National Institute of Allergy and Infectious Diseases and US National Institutes of Health.


Assuntos
Vacinas Antimaláricas , Malária Falciparum , Vacinas Meningocócicas , Animais , Adulto , Humanos , Adolescente , Adulto Jovem , Pessoa de Meia-Idade , Hidróxido de Alumínio , Plasmodium falciparum , Vacinas Antimaláricas/efeitos adversos , Método Duplo-Cego , Imunogenicidade da Vacina
12.
J Clin Invest ; 131(7)2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33561016

RESUMO

BACKGROUNDVaccines that block human-to-mosquito Plasmodium transmission are needed for malaria eradication, and clinical trials have targeted zygote antigen Pfs25 for decades. We reported that a Pfs25 protein-protein conjugate vaccine formulated in alum adjuvant induced serum functional activity in both US and Malian adults. However, antibody levels declined rapidly, and transmission-reducing activity required 4 vaccine doses. Functional immunogenicity and durability must be improved before advancing transmission-blocking vaccines further in clinical development. We hypothesized that the prefertilization protein Pfs230 alone or in combination with Pfs25 would improve functional activity.METHODSTransmission-blocking vaccine candidates based on gamete antigen Pfs230 or Pfs25 were conjugated with Exoprotein A, formulated in Alhydrogel, and administered to mice, rhesus macaques, and humans. Antibody levels were measured by ELISA and transmission-reducing activity was assessed by the standard membrane feeding assay.RESULTSPfs25-EPA/Alhydrogel and Pfs230D1-EPA/Alhydrogel induced similar serum functional activity in mice, but Pfs230D1-EPA induced significantly greater activity in rhesus monkeys that was enhanced by complement. In US adults, 2 vaccine doses induced complement-dependent activity in 4 of 5 Pfs230D1-EPA/Alhydrogel recipients but no significant activity in 5 Pfs25-EPA recipients, and combination with Pfs25-EPA did not increase activity over Pfs230D1-EPA alone.CONCLUSIONThe complement-dependent functional immunogenicity of Pfs230D1-EPA represents a significant improvement over Pfs25-EPA in this comparative study. The rhesus model is more predictive of the functional human immune response to Pfs230D1 than is the mouse model.TRIAL REGISTRATIONClinicalTrials.gov NCT02334462.FUNDINGIntramural Research Program of the National Institute of Allergy and Infectious Diseases, National Institutes of Health.


Assuntos
Hidróxido de Alumínio/administração & dosagem , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/administração & dosagem , Vacinas Antimaláricas/administração & dosagem , Plasmodium falciparum/imunologia , Proteínas de Protozoários/administração & dosagem , Adulto , Animais , Antígenos de Protozoários/imunologia , Feminino , Humanos , Macaca mulatta , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Protozoários/imunologia
13.
Nat Commun ; 12(1): 1750, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33741942

RESUMO

Malaria elimination requires tools that interrupt parasite transmission. Here, we characterize B cell receptor responses among Malian adults vaccinated against the first domain of the cysteine-rich 230 kDa gamete surface protein Pfs230, a key protein in sexual stage development of P. falciparum parasites. Among nine Pfs230 human monoclonal antibodies (mAbs) that we generated, one potently blocks transmission to mosquitoes in a complement-dependent manner and reacts to the gamete surface; the other eight show only low or no blocking activity. The structure of the transmission-blocking mAb in complex with vaccine antigen reveals a large discontinuous conformational epitope, specific to domain 1 of Pfs230 and comprising six structural elements in the protein. The epitope is conserved, suggesting the transmission-blocking mAb is broadly functional. This study provides a rational basis to improve malaria vaccines and develop therapeutic antibodies for malaria elimination.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Antiprotozoários/farmacologia , Epitopos/imunologia , Células Germinativas/imunologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/efeitos dos fármacos , Adulto , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/química , Antígenos de Protozoários/imunologia , Sítios de Ligação , Células Cultivadas , Epitopos/química , Interações Hospedeiro-Parasita/efeitos dos fármacos , Interações Hospedeiro-Parasita/imunologia , Humanos , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/imunologia , Malária Falciparum/parasitologia , Malária Falciparum/transmissão , Mosquitos Vetores/parasitologia , Plasmodium falciparum/imunologia , Plasmodium falciparum/fisiologia , Conformação Proteica , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia
14.
J Biol Chem ; 284(39): 26951-63, 2009 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-19633296

RESUMO

The Plasmodium falciparum circumsporozoite protein (CSP) is critical for sporozoite function and invasion of hepatocytes. Given its critical nature, a phase III human CSP malaria vaccine trial is ongoing. The CSP is composed of three regions as follows: an N terminus that binds heparin sulfate proteoglycans, a four amino acid repeat region (NANP), and a C terminus that contains a thrombospondin-like type I repeat (TSR) domain. Despite the importance of CSP, little is known about its structure. Therefore, recombinant forms of CSP were produced by expression in both Escherichia coli (Ec) and then refolded (EcCSP) or in the methylotrophic yeast Pichia pastoris (PpCSP) for structural analyses. To analyze the TSR domain of recombinant CSP, conformation-dependent monoclonal antibodies that recognized unfixed P. falciparum sporozoites and inhibited sporozoite invasion of HepG2 cells in vitro were identified. These monoclonal antibodies recognized all recombinant CSPs, indicating the recombinant CSPs contain a properly folded TSR domain structure. Characterization of both EcCSP and PpCSP by dynamic light scattering and velocity sedimentation demonstrated that both forms of CSP appeared as highly extended proteins (R(h) 4.2 and 4.58 nm, respectively). Furthermore, high resolution atomic force microscopy revealed flexible, rod-like structures with a ribbon-like appearance. Using this information, we modeled the NANP repeat and TSR domain of CSP. Consistent with the biochemical and biophysical results, the repeat region formed a rod-like structure about 21-25 nm in length and 1.5 nm in width. Thus native CSP appears as a glycosylphosphatidylinositol-anchored, flexible rod-like protein on the sporozoite surface.


Assuntos
Vacinas Antimaláricas/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Esporozoítos/imunologia , Animais , Anticorpos Monoclonais/imunologia , Antígenos de Protozoários/química , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Dicroísmo Circular , Eletroforese em Gel de Poliacrilamida , Glicosilfosfatidilinositóis/metabolismo , Heparina/análogos & derivados , Heparina/metabolismo , Hepatócitos/imunologia , Hepatócitos/parasitologia , Hepatócitos/patologia , Humanos , Immunoblotting , Modelos Moleculares , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Ligação Proteica , Dobramento de Proteína , Estrutura Terciária de Proteína , Proteoglicanas/metabolismo , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Esporozoítos/metabolismo , Temperatura , Ultracentrifugação
15.
Commun Biol ; 3(1): 395, 2020 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-32709983

RESUMO

Proteins Pfs230 and Pfs48/45 are Plasmodium falciparum transmission-blocking (TB) vaccine candidates that form a membrane-bound protein complex on gametes. The biological role of Pfs230 or the Pfs230-Pfs48/45 complex remains poorly understood. Here, we present the crystal structure of recombinant Pfs230 domain 1 (Pfs230D1M), a 6-cysteine domain, in complex with the Fab fragment of a TB monoclonal antibody (mAb) 4F12. We observed the arrangement of Pfs230 on the surface of macrogametes differed from that on microgametes, and that Pfs230, with no known membrane anchor, may exist on the membrane surface in the absence of Pfs48/45. 4F12 appears to sterically interfere with Pfs230 function. Combining mAbs against different epitopes of Pfs230D1 or of Pfs230D1 and Pfs48/45, significantly increased TB activity. These studies elucidate a mechanism of action of the Pfs230D1 vaccine, model the functional activity induced by a polyclonal antibody response and support the development of TB vaccines targeting Pfs230D1 and Pfs230D1-Pfs48/45.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/farmacologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/patogenicidade , Animais , Antígenos de Protozoários/genética , Humanos , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Malária Falciparum/transmissão , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/imunologia
16.
JCI Insight ; 5(22)2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-33048842

RESUMO

Plasma antimalarial Ab can mediate antiparasite immunity but has not previously been characterized at the molecular level. Here, we develop an innovative strategy to characterize humoral responses by integrating profiles of plasma immunoglobulins (IGs) or Abs with those expressed on B cells as part of the B cell receptor. We applied this strategy to define plasma IG and to determine variable (V) gene usage after vaccination with the Plasmodium falciparum zygote antigen Pfs25. Using proteomic tools coupled with bulk immunosequencing data, we determined human antigen-binding fragment [F(ab')2] peptide sequences from plasma IG of adults who received 4 doses of Pfs25-EPA/Alhydrogel. Specifically, Pfs25 antigen-specific F(ab')2 peptides (Pfs25-IG) were aligned to cDNA sequences of IG heavy (IGH) chain complementarity determining region 3 from a data set generated by total peripheral B cell immunosequencing of the entire vaccinated population. IGHV4 was the most commonly identified IGHV subgroup of Pfs25-IG, a pattern that was corroborated by V heavy/V light chain sequencing of Pfs25-specific single B cells from 5 vaccinees and by matching plasma Pfs25-IG peptides and V-(D)-J sequences of Pfs25-specific single B cells from the same donor. Among 13 recombinant human mAbs generated from IG sequences of Pfs25-specific single B cells, a single IGHV4 mAb displayed strong neutralizing activity, reducing the number of P. falciparum oocysts in infected mosquitoes by more than 80% at 100 µg/mL. Our approach characterizes the human plasma Ab repertoire in response to the Pfs25-EPA/Alhydrogel vaccine and will be useful for studying circulating Abs in response to other vaccines as well as those induced during infections or autoimmune disorders.


Assuntos
Anticorpos Antiprotozoários/sangue , Antimaláricos/imunologia , Linfócitos B/imunologia , Imunoglobulinas/sangue , Malária Falciparum/sangue , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Adjuvantes Imunológicos , Adolescente , Adulto , Anticorpos Monoclonais/sangue , Anticorpos Monoclonais/imunologia , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Antimaláricos/administração & dosagem , Ensaios Clínicos como Assunto , Feminino , Humanos , Imunoglobulinas/imunologia , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Malária Falciparum/prevenção & controle , Masculino , Pessoa de Meia-Idade , Vacinação , Adulto Jovem
17.
Mol Biochem Parasitol ; 164(1): 45-56, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19073223

RESUMO

Immunization with a recombinant yeast-expressed Plasmodium falciparum merozoite surface protein 3 (MSP3) protected Aotus nancymai monkeys against a virulent challenge infection. Unfortunately, the production process for this yeast-expressed material was not optimal for human trials. In an effort to produce a recombinant MSP3 protein in a scaleable manner, we expressed and purified near-full-length MSP3 in Escherichia coli (EcMSP3). Purified EcMSP3 formed non-globular dimers as determined by analytical size-exclusion HPLC with in-line multi-angle light scatter and quasi-elastic light scatter detection and velocity sedimentation (R(h) 7.6+/-0.2nm and 6.9nm, respectively). Evaluation by high-resolution atomic force microscopy revealed non-linear asymmetric structures, with beaded domains and flexible loops that were recognized predominantly as dimers, although monomers and larger multimers were observed. The beaded substructure corresponds to predicted structural domains, which explains the velocity sedimentation results and improves the conceptual model of the protein. Vaccination with EcMSP3 in Freund's adjuvant-induced antibodies that recognized native MSP3 in parasitized erythrocytes by an immunofluorescence assay and gave delayed time to treatment in a group of Aotus monkeys in a virulent challenge infection with the FVO strain of P. falciparum. Three of the seven monkeys vaccinated with EcMSP3 had low peak parasitemias. EcMSP3, which likely mimics the native MSP3 structure located on the merozoite surface, is a viable candidate for inclusion in a multi-component malaria vaccine.


Assuntos
Antígenos de Protozoários/química , Escherichia coli/genética , Modelos Moleculares , Plasmodium falciparum/química , Proteínas de Protozoários/química , Proteínas Recombinantes/química , Sequência de Aminoácidos , Animais , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Aotidae , Clonagem Molecular , Expressão Gênica , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Dados de Sequência Molecular , Plasmodium falciparum/genética , Estrutura Terciária de Proteína , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Distribuição Aleatória , Proteínas Recombinantes/genética , Vacinas Sintéticas/imunologia
18.
Vaccine ; 37(38): 5762-5769, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30262247

RESUMO

Efforts to develop a vaccine for the elimination of malaria include the use of carrier proteins to assemble monomeric antigens into nanoparticles to maximize immunogenicity. Recombinant ExoProtein A (EPA) is a detoxified form of Pseudomonas aeruginosa Exotoxin A which has been used as a carrier in the conjugate vaccine field. A pilot-scale process developed for purification of EPA yielded product that consistently approached a preset upper limit for host cell protein (HCP) content per human dose. To minimize the risk of bulk material exceeding the specification, the purification process was redeveloped using mixed-mode chromatography resins. Purified EPA derived from the primary and redeveloped processes were comparable following full biochemical and biophysical characterization. However, using a process specific immunoassay, the HCP content was shown to decrease from a range of 0.14-0.24% w/w of total protein to below the level of detection with the revised process. The improved process reproducibly yields EPA with highly similar quality characteristics as the original process but with an improved profile for the HCP content.


Assuntos
ADP Ribose Transferases/química , ADP Ribose Transferases/imunologia , Toxinas Bacterianas/química , Toxinas Bacterianas/imunologia , Fenômenos Químicos , Exotoxinas/química , Exotoxinas/imunologia , Vacinas contra Pseudomonas/química , Vacinas contra Pseudomonas/imunologia , Vacinas Sintéticas/química , Vacinas Sintéticas/imunologia , Fatores de Virulência/química , Fatores de Virulência/imunologia , ADP Ribose Transferases/isolamento & purificação , Sequência de Aminoácidos , Animais , Toxinas Bacterianas/isolamento & purificação , Epitopos/imunologia , Exotoxinas/isolamento & purificação , Humanos , Imunogenicidade da Vacina , Camundongos , Peptídeos/imunologia , Processamento de Proteína Pós-Traducional , Infecções por Pseudomonas/prevenção & controle , Vacinas contra Pseudomonas/isolamento & purificação , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Análise Espectral , Vacinas Sintéticas/isolamento & purificação , Fatores de Virulência/isolamento & purificação , Exotoxina A de Pseudomonas aeruginosa
19.
Front Immunol ; 10: 2295, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31636633

RESUMO

Plasmodium vivax malaria incidence has increased in Latin America and Asia and is responsible for nearly 74.1% of malaria cases in Latin America. Immune responses to P. vivax are less well characterized than those to P. falciparum, partly because P. vivax is more difficult to cultivate in the laboratory. While antibodies are known to play an important role in P. vivax disease control, few studies have evaluated responses to P. vivax sexual stage antigens. We collected sera or plasma samples from P. vivax-infected subjects from Brazil (n = 70) and Cambodia (n = 79) to assess antibody responses to domain 1 of the gametocyte/gamete stage protein Pvs230 (Pvs230D1M). We found that 27.1% (19/70) and 26.6% (21/79) of subjects from Brazil and Cambodia, respectively, presented with detectable antibody responses to Pvs230D1M antigen. The most frequent subclasses elicited in response to Pvs230D1M were IgG1 and IgG3. Although age did not correlate significantly with Pvs230D1M antibody levels overall, we observed significant differences between age strata. Hemoglobin concentration inversely correlated with Pvs230D1M antibody levels in Brazil, but not in Cambodia. Additionally, we analyzed the antibody response against Pfs230D1M, the P. falciparum ortholog of Pvs230D1M. We detected antibodies to Pfs230D1M in 7.2 and 16.5% of Brazilian and Cambodian P. vivax-infected subjects. Depletion of Pvs230D1M IgG did not impair the response to Pfs230D1M, suggesting pre-exposure to P. falciparum, or co-infection. We also analyzed IgG responses to sporozoite protein PvCSP (11.4 and 41.8% in Brazil and Cambodia, respectively) and to merozoite protein PvDBP-RII (67.1 and 48.1% in Brazil and Cambodia, respectively), whose titers also inversely correlated with hemoglobin concentration only in Brazil. These data establish patterns of seroreactivity to sexual stage Pvs230D1M and show similar antibody responses among P. vivax-infected subjects from regions of differing transmission intensity in Brazil and Cambodia.


Assuntos
Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Plasmodium vivax/imunologia , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Feminino , Hemoglobinas/análise , Humanos , Imunoglobulina G/sangue , Malária Vivax/prevenção & controle , Masculino , Pessoa de Meia-Idade , Plasmodium falciparum/imunologia
20.
Vet Surg ; 37(7): 696-701, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19134092

RESUMO

OBJECTIVE: To assess whether en bloc ligation of all caudal mediastinal tissue between the aorta and thoracic vertebrae will occlude all branches of the thoracic duct. STUDY DESIGN: Descriptive study. ANIMALS: Canine cadavers (n=15). METHODS: The cisterna chyli was approached through a right paralumbar abdominal incision and cannulated. A right 10th intercostal thoracotomy was performed and the tissue of the caudal mediastinum dorsal to the aorta and ventral to the thoracic vertebrae was ligated. Radio-opaque contrast material was injected into the cisterna chyli during fluoroscopic imaging of the thorax and cranial abdomen. Still images were obtained before and after contrast injection and a subtraction image produced. Gray values were measured from these images, cranial and caudal to the ligature site. RESULTS: Cannulation of the cisterna chyli in 1 cadaver was impossible and it was, therefore, excluded. In 13 (93%) cadavers contrast material did not pass cranial to the ligature site. In 1 cadaver, an intact thoracic duct was identified on lymphangiography outside the en bloc ligation. CONCLUSION: En bloc ligation of the caudal mediastinal tissue dorsal to the aorta was successful in preventing opacification with contrast material of all branches of the thoracic duct in 93% of cadavers. CLINICAL RELEVANCE: En bloc ligation is successful in 93% of cadavers and reduces the operative time compared with techniques requiring some type of mesenteric lymphangiography to aid identification of the thoracic duct.


Assuntos
Quilotórax/veterinária , Doenças do Cão/cirurgia , Ligadura/veterinária , Ducto Torácico/cirurgia , Toracoscopia/veterinária , Animais , Cadáver , Quilotórax/cirurgia , Cães , Ligadura/métodos , Linfografia/veterinária , Projetos Piloto , Toracoscopia/métodos , Fatores de Tempo , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA