Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
2.
Clin Immunol ; 229: 108765, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34089859

RESUMO

Whether and how an acute immune challenge may affect DNA Damage Response (DDR) is unknown. By studying vaccinations against Influenza and SARS-CoV-2 (mRNA-based) we found acute increases of type-I interferon-inducible gene expression, oxidative stress and DNA damage accumulation in blood mononuclear cells of 9 healthy controls, coupled with effective anti-SARS-CoV-2 neutralizing antibody production in all. Increased DNA damage after SARS-CoV-2 vaccine, partly due to increased oxidative stress, was transient, whereas the inherent DNA repair capacity was found intact. In contrast, in 26 patients with Systemic Lupus Erythematosus, who served as controls in the context of chronic immune activation, we validated increased DNA damage accumulation, increased type-I interferon-inducible gene expression and induction of oxidative stress, however aberrant DDR was associated with deficiencies in nucleotide excision repair pathways. These results indicate that acute immune challenge can indeed activate DDR pathways, whereas, contrary to chronic immune challenge, successful repair of DNA lesions occurs.


Assuntos
Anticorpos Neutralizantes/fisiologia , Vacinas contra COVID-19/imunologia , COVID-19/prevenção & controle , Dano ao DNA , Lúpus Eritematoso Sistêmico/imunologia , SARS-CoV-2/imunologia , Adolescente , Adulto , Idoso , COVID-19/patologia , Estudos de Casos e Controles , Feminino , Regulação da Expressão Gênica/imunologia , Humanos , Interferon Tipo I/metabolismo , Masculino , Pessoa de Meia-Idade , Estresse Oxidativo , Vacinas Sintéticas/imunologia , Adulto Jovem , Vacinas de mRNA
4.
N Engl J Med ; 384(20): 1885-1898, 2021 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-33725432

RESUMO

BACKGROUND: Assessment of the safety and efficacy of vaccines against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in different populations is essential, as is investigation of the efficacy of the vaccines against emerging SARS-CoV-2 variants of concern, including the B.1.351 (501Y.V2) variant first identified in South Africa. METHODS: We conducted a multicenter, double-blind, randomized, controlled trial to assess the safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) in people not infected with the human immunodeficiency virus (HIV) in South Africa. Participants 18 to less than 65 years of age were assigned in a 1:1 ratio to receive two doses of vaccine containing 5×1010 viral particles or placebo (0.9% sodium chloride solution) 21 to 35 days apart. Serum samples obtained from 25 participants after the second dose were tested by pseudovirus and live-virus neutralization assays against the original D614G virus and the B.1.351 variant. The primary end points were safety and efficacy of the vaccine against laboratory-confirmed symptomatic coronavirus 2019 illness (Covid-19) more than 14 days after the second dose. RESULTS: Between June 24 and November 9, 2020, we enrolled 2026 HIV-negative adults (median age, 30 years); 1010 and 1011 participants received at least one dose of placebo or vaccine, respectively. Both the pseudovirus and the live-virus neutralization assays showed greater resistance to the B.1.351 variant in serum samples obtained from vaccine recipients than in samples from placebo recipients. In the primary end-point analysis, mild-to-moderate Covid-19 developed in 23 of 717 placebo recipients (3.2%) and in 19 of 750 vaccine recipients (2.5%), for an efficacy of 21.9% (95% confidence interval [CI], -49.9 to 59.8). Among the 42 participants with Covid-19, 39 cases (95.1% of 41 with sequencing data) were caused by the B.1.351 variant; vaccine efficacy against this variant, analyzed as a secondary end point, was 10.4% (95% CI, -76.8 to 54.8). The incidence of serious adverse events was balanced between the vaccine and placebo groups. CONCLUSIONS: A two-dose regimen of the ChAdOx1 nCoV-19 vaccine did not show protection against mild-to-moderate Covid-19 due to the B.1.351 variant. (Funded by the Bill and Melinda Gates Foundation and others; ClinicalTrials.gov number, NCT04444674; Pan African Clinical Trials Registry number, PACTR202006922165132).


Assuntos
Anticorpos Neutralizantes/sangue , Vacinas contra COVID-19/imunologia , COVID-19/prevenção & controle , Imunogenicidade da Vacina , SARS-CoV-2 , Adenoviridae , Adolescente , Adulto , Anticorpos Neutralizantes/fisiologia , COVID-19/epidemiologia , COVID-19/imunologia , Teste Sorológico para COVID-19 , Vacinas contra COVID-19/administração & dosagem , Vacinas contra COVID-19/efeitos adversos , ChAdOx1 nCoV-19 , Método Duplo-Cego , Humanos , Pessoa de Meia-Idade , África do Sul , Linfócitos T/fisiologia , Falha de Tratamento , Potência de Vacina , Adulto Jovem
5.
JCI Insight ; 6(6)2021 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-33621211

RESUMO

Comorbid medical illnesses, such as obesity and diabetes, are associated with more severe COVID-19, hospitalization, and death. However, the role of the immune system in mediating these clinical outcomes has not been determined. We used multiparameter flow cytometry and systems serology to comprehensively profile the functions of T cells and antibodies targeting spike, nucleocapsid, and envelope proteins in a convalescent cohort of COVID-19 subjects who were either hospitalized (n = 20) or not hospitalized (n = 40). To avoid confounding, subjects were matched by age, sex, ethnicity, and date of symptom onset. Surprisingly, we found that the magnitude and functional breadth of virus-specific CD4+ T cell and antibody responses were consistently higher among hospitalized subjects, particularly those with medical comorbidities. However, an integrated analysis identified more coordination between polyfunctional CD4+ T cells and antibodies targeting the S1 domain of spike among subjects who were not hospitalized. These data reveal a functionally diverse and coordinated response between T cells and antibodies targeting SARS-CoV-2, which is reduced in the presence of comorbid illnesses that are known risk factors for severe COVID-19.


Assuntos
Anticorpos Antivirais/fisiologia , Linfócitos T CD4-Positivos/fisiologia , COVID-19/virologia , Hospitalização , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus , Vírion , Adulto , Idoso , Anticorpos Neutralizantes/metabolismo , Anticorpos Neutralizantes/fisiologia , Anticorpos Antivirais/metabolismo , Linfócitos T CD4-Positivos/metabolismo , COVID-19/epidemiologia , COVID-19/imunologia , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/imunologia , Comorbidade , Diabetes Mellitus/epidemiologia , Diabetes Mellitus/imunologia , Feminino , Humanos , Imunidade Humoral , Masculino , Pessoa de Meia-Idade , Nucleocapsídeo , Índice de Gravidade de Doença , Envelope Viral , Proteínas Virais , Adulto Jovem
6.
Immunol Cell Biol ; 99(2): 177-191, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33113212

RESUMO

Coronavirus disease 2019 (COVID-19) is a zoonosis like most of the great plagues sculpting human history, from smallpox to pandemic influenza and human immunodeficiency virus. When viruses jump into a new species the outcome of infection ranges from asymptomatic to lethal, historically ascribed to "genetic resistance to viral disease." People have exploited these differences for good and bad, for developing vaccines from cowpox and horsepox virus, controlling rabbit plagues with myxoma virus and introducing smallpox during colonization of America and Australia. Differences in resistance to viral disease are at the core of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) crisis, yet our understanding of the mechanisms in any interspecies leap falls short of the mark. Here I review how the two key parameters of viral disease are countered by fundamentally different genetic mechanisms for resistance: (1) virus transmission, countered primarily by activation of innate and adaptive immune responses; and (2) pathology, countered primarily by tolerance checkpoints to limit innate and adaptive immune responses. I discuss tolerance thresholds and the role of CD8 T cells to limit pathological immune responses, the problems posed by tolerant superspreaders and the signature coronavirus evasion strategy of eliciting only short-lived neutralizing antibody responses. Pinpointing and targeting the mechanisms responsible for varying pathology and short-lived antibody were beyond reach in previous zoonoses, but this time we are armed with genomic technologies and more knowledge of immune checkpoint genes. These known unknowns must now be tackled to solve the current COVID-19 crisis and the inevitable zoonoses to follow.


Assuntos
COVID-19 , Tolerância Imunológica/imunologia , Imunidade Inata/genética , SARS-CoV-2/imunologia , Viroses/imunologia , Imunidade Adaptativa/genética , Imunidade Adaptativa/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/fisiologia , Anticorpos Antivirais/imunologia , COVID-19/genética , COVID-19/imunologia , COVID-19/virologia , Humanos , Tolerância Imunológica/genética , Coelhos , SARS-CoV-2/genética , Zoonoses Virais/genética , Zoonoses Virais/imunologia , Viroses/genética
7.
Proc Natl Acad Sci U S A ; 117(14): 7929-7940, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32209668

RESUMO

HIV-1 vaccine development aims to elicit broadly neutralizing antibodies (bnAbs) against diverse viral strains. In some HIV-1-infected individuals, bnAbs evolved from precursor antibodies through affinity maturation. To induce bnAbs, a vaccine must mediate a similar antibody maturation process. One way to test a vaccine is to immunize mouse models that express human bnAb precursors and assess whether the vaccine can convert precursor antibodies into bnAbs. A major problem with such mouse models is that bnAb expression often hinders B cell development. Such developmental blocks may be attributed to the unusual properties of bnAb variable regions, such as poly-reactivity and long antigen-binding loops, which are usually under negative selection during primary B cell development. To address this problem, we devised a method to circumvent such B cell developmental blocks by expressing bnAbs conditionally in mature B cells. We validated this method by expressing the unmutated common ancestor (UCA) of the human VRC26 bnAb in transgenic mice. Constitutive expression of the VRC26UCA led to developmental arrest of B cell progenitors in bone marrow; poly-reactivity of the VRC26UCA and poor pairing of the VRC26UCA heavy chain with the mouse surrogate light chain may contribute to this phenotype. The conditional expression strategy bypassed the impediment to VRC26UCA B cell development, enabling the expression of VRC26UCA in mature B cells. This approach should be generally applicable for expressing other bnAbs that are under negative selection during B cell development.


Assuntos
Vacinas contra a AIDS/imunologia , Anticorpos Anti-HIV/imunologia , Infecções por HIV/imunologia , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia , Vacinas contra a AIDS/farmacologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/fisiologia , Linfócitos B/imunologia , Linfócitos B/virologia , Modelos Animais de Doenças , Anticorpos Anti-HIV/farmacologia , Infecções por HIV/prevenção & controle , Infecções por HIV/virologia , Soropositividade para HIV/genética , Soropositividade para HIV/imunologia , HIV-1/efeitos dos fármacos , HIV-1/imunologia , HIV-1/patogenicidade , Humanos , Ativação Linfocitária/imunologia , Camundongos , Produtos do Gene env do Vírus da Imunodeficiência Humana/genética
8.
Exp Cell Res ; 387(1): 111756, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31811830

RESUMO

Breast cancer (BC) is one of the most common cancers among women in both developed and developing countries with a rising incidence. Using the MMTV-PyMT transgenic mouse model and xenografted breast cancer model, we found that R5, a neutralizing antibody to Robo1, significantly inhibited BC growth and metastasis. Angiogenesis is involved in the growth and metastasis of BC. Interestingly, R5 significantly decreases microvessel density in BC tissues, and inhibits blood vessel formation and development in in vivo chick embryo chorioallantoic membrane (CAM), yolk sac membrane (YSM) and Matrigel plug models. To investigate whether its anti-breast cancer efficacy is ascribed to its direct antiangiogenic properties, xenografted breast cancer model on CAM was established. Furthermore, R5 significantly reduces the tube formation of the vascular plexus on xenografted breast tumor on CAM. R5 also suppresses the migration and the tubular structure formation of human umbilical vein endothelial cells (HUVECs) by down-regulating the expression of filamin A (FLNA). These findings show that R5 has the potential to be a promising agent for the treatment of BC by suppressing the tumor-induced angiogenesis.


Assuntos
Anticorpos Neutralizantes/fisiologia , Neoplasias da Mama/tratamento farmacológico , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Filaminas/metabolismo , Neovascularização Patológica/tratamento farmacológico , Proteínas do Tecido Nervoso/metabolismo , Receptores Imunológicos/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neovascularização Patológica/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Proteínas Roundabout
9.
PLoS Pathog ; 15(2): e1007375, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30707748

RESUMO

Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease localized to China, Japan, and Korea that is characterized by severe hemorrhage and a high fatality rate. Currently, no specific vaccine or treatment has been approved for this disease. To develop a therapeutic agent for SFTS, we isolated antibodies from a phage-displayed antibody library that was constructed from a patient who recovered from SFTS virus (SFTSV) infection. One antibody, designated as Ab10, was reactive to the Gn envelope glycoprotein of SFTSV and protected host cells and A129 mice from infection in both in vitro and in vivo experiments. Notably, Ab10 protected 80% of mice, even when injected 5 days after inoculation with a lethal dose of SFTSV. Using cross-linker assisted mass spectrometry and alanine scanning, we located the non-linear epitope of Ab10 on the Gn glycoprotein domain II and an unstructured stem region, suggesting that Ab10 may inhibit a conformational alteration that is critical for cell membrane fusion between the virus and host cell. Ab10 reacted to recombinant Gn glycoprotein in Gangwon/Korea/2012, HB28, and SD4 strains. Additionally, based on its epitope, we predict that Ab10 binds the Gn glycoprotein in 247 of 272 SFTSV isolates previously reported. Together, these data suggest that Ab10 has potential to be developed into a therapeutic agent that could protect against more than 90% of reported SFTSV isolates.


Assuntos
Anticorpos Neutralizantes/metabolismo , Phlebovirus/imunologia , Adulto , Animais , Anticorpos Neutralizantes/fisiologia , Anticorpos Antivirais/metabolismo , Infecções por Bunyaviridae/terapia , Epitopos/imunologia , Feminino , Febre , Glutamina/imunologia , Glutamina/metabolismo , Glicoproteínas/imunologia , Células HEK293 , Humanos , Leucopenia , Masculino , Camundongos , Camundongos Knockout , Testes de Neutralização , Phlebovirus/patogenicidade , República da Coreia , Trombocitopenia/imunologia , Proteínas do Envelope Viral/imunologia
10.
PLoS Biol ; 17(1): e3000114, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30650070

RESUMO

During the entry process, the human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein (Env) trimer undergoes a sequence of conformational changes triggered by both CD4 and coreceptor engagement. Resolving the conformation of these transient entry intermediates has proven challenging. Here, we fine-mapped the antigenicity of entry intermediates induced by increasing CD4 engagement of cell surface-expressed Env. Escalating CD4 triggering led to the sequential adoption of different pre-fusion conformational states of the Env trimer, up to the pre-hairpin conformation, that we assessed for antibody epitope presentation. Maximal accessibility of the coreceptor binding site was detected below Env saturation by CD4. Exposure of the fusion peptide and heptad repeat 1 (HR1) required higher CD4 occupancy. Analyzing the diverse antigenic states of the Env trimer, we obtained key insights into the transitions in epitope accessibility of broadly neutralizing antibodies (bnAbs). Several bnAbs preferentially bound CD4-triggered Env, indicating a potential capacity to neutralize both pre- and post-CD4 engagement, which needs to be explored. Assessing binding and neutralization activity of bnAbs, we confirm antibody dissociation rates as a driver of incomplete neutralization. Collectively, our findings highlight a need to resolve Env conformations that are neutralization-relevant to provide guidance for immunogen development.


Assuntos
Antígenos CD4/metabolismo , Epitopos/imunologia , Produtos do Gene env do Vírus da Imunodeficiência Humana/química , Anticorpos Neutralizantes/metabolismo , Anticorpos Neutralizantes/fisiologia , Proteína gp120 do Envelope de HIV/química , HIV-1/imunologia , Humanos , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Produtos do Gene env do Vírus da Imunodeficiência Humana/metabolismo , Produtos do Gene env do Vírus da Imunodeficiência Humana/fisiologia
11.
PLoS Pathog ; 15(1): e1007427, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30605488

RESUMO

Mucosal immunoglobulins comprise mainly secretory IgA antibodies (SIgAs), which are the major contributor to pathogen-specific immune responses in mucosal tissues. These SIgAs are highly heterogeneous in terms of their quaternary structure. A recent report shows that the polymerization status of SIgA defines their functionality in the human upper respiratory mucosa. Higher order polymerization of SIgA (i.e., tetramers) leads to a marked increase in neutralizing activity against influenza viruses. However, the precise molecular mechanisms underlying the effects of SIgA polymerization remain elusive. Here, we developed a method for generating recombinant tetrameric monoclonal SIgAs. We then compared the anti-viral activities of these tetrameric SIgAs, which possessed variable regions identical to that of a broadly neutralizing anti-influenza antibody F045-092 against influenza A viruses, with that of monomeric IgG or IgA. The tetrameric SIgA showed anti-viral inhibitory activity superior to that of other forms only when the antibody exhibits low-affinity binding to the target. By contrast, SIgA tetramerization did not substantially modify anti-viral activity against targets with high-affinity binding. Taken together, the data suggest that tetramerization of SIgA improved target breadth, but not peak potency of antiviral functions of the broadly neutralizing anti-influenza antibody. This phenomenon presumably represents one of the mechanisms by which SIgAs present in human respiratory mucosa prevent infection by antigen-drifted influenza viruses. Understanding the mechanisms involved in cross neutralization of viruses by SIgAs might facilitate the development of vaccine strategies against viral infection of mucosal tissues.


Assuntos
Anticorpos Neutralizantes/imunologia , Imunoglobulina A Secretora/imunologia , Imunoglobulina A Secretora/metabolismo , Animais , Anticorpos Neutralizantes/fisiologia , Anticorpos Antivirais/imunologia , Antivirais , Linhagem Celular , Embrião de Galinha , Cães , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Imunoglobulina A/imunologia , Imunoglobulina A/metabolismo , Imunoglobulina A Secretora/fisiologia , Vírus da Influenza A/imunologia , Vacinas contra Influenza , Influenza Humana/imunologia , Células Madin Darby de Rim Canino , Testes de Neutralização , Orthomyxoviridae/imunologia , Polimerização , Ligação Proteica , Proteínas Recombinantes/metabolismo
12.
Clin Infect Dis ; 69(1): 151-154, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-30561563

RESUMO

Human immunodeficiency virus (HIV)-exposed, uninfected infants have higher risks of respiratory syncytial virus-associated hospitalization than HIV-unexposed infants. Despite similar neutralizing antibody titers between HIV-infected and -uninfected women, maternal HIV infection and hypergammaglobulinemia were independently associated with lower titers in newborns. Maternal hypergammaglobulinemia was associated with lower cord-to-maternal antibody ratio.


Assuntos
Anticorpos Neutralizantes/fisiologia , Anticorpos Antivirais/fisiologia , Infecções por HIV/complicações , Imunidade Materno-Adquirida , Vírus Sincicial Respiratório Humano/imunologia , Feminino , Humanos , Hipergamaglobulinemia , Recém-Nascido , Gravidez
13.
Epidemiol Infect ; 146(11): 1372-1383, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29909804

RESUMO

Previously we reported on the HPIV2 genotype distribution in Croatia 2011-2014. Here we expand this period up to 2017 and confirm that G1a genotype has replaced G3 genotype from the period 2011-2014. Our hypothesis was that the G1a-to-G3 genotype replacement is an antibody-driven event. A cross-neutralisation with anti-HPIV2 sera specific for either G1a or G3 genotype revealed the presence of genotype-specific antigenic determinants. By the profound, in silico analyses three potential B cell epitopic regions were identified in the hemagglutinin neuraminidase (regions 314-361 and 474-490) and fusion protein (region 440-484). The region identified in the fusion protein does not show any unique site between the G1a and G3 isolates, five differentially glycosylated sites in the G1a and G3 genotype isolates were identified in epitopic regions of hemagglutinin neuraminidase. All positively selected codons were found to be located either in the region 314-316 or in the region 474-490 what indicates a strong positive selection in this region and reveals that these regions are susceptible to evolutionary pressure possibly caused by antibodies what gives a strong verification to our hypothesis that neutralising antibodies are a key determinant in the inherently complex adaptive evolution of HPIV2 in the region.


Assuntos
Anticorpos Neutralizantes/fisiologia , Vírus da Parainfluenza 2 Humana/genética , Infecções por Rubulavirus/virologia , Adolescente , Distribuição por Idade , Animais , Anticorpos Antivirais/fisiologia , Criança , Pré-Escolar , Chlorocebus aethiops , Croácia/epidemiologia , Epitopos de Linfócito B/química , Epitopos de Linfócito B/imunologia , Feminino , Genótipo , Cobaias , Proteína HN/imunologia , Humanos , Lactente , Funções Verossimilhança , Pessoa de Meia-Idade , Vírus da Parainfluenza 2 Humana/classificação , Vírus da Parainfluenza 2 Humana/imunologia , Filogenia , RNA Viral/química , RNA Viral/genética , Recidiva , Infecções por Rubulavirus/epidemiologia , Infecções por Rubulavirus/imunologia , Estações do Ano , Alinhamento de Sequência , Células Vero
14.
Toxins (Basel) ; 10(3)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29494518

RESUMO

BACKGROUND: Shiga toxin-producing Escherichia coli (STEC) are a subset of pathogens leading to illnesses such as diarrhea, hemolytic uremic syndrome and even death. The Shiga toxins are the main virulence factors and divided in two groups: Stx1 and Stx2, of which the latter is more frequently associated with severe pathologies in humans. RESULTS: An immune library of nanobodies (Nbs) was constructed after immunizing an alpaca with recombinant Shiga toxin-2a B subunit (rStx2aB), to retrieve multiple rStx2aB-specific Nbs. The specificity of five Nbs towards rStx2aB was confirmed in ELISA and Western blot. Nb113 had the highest affinity (9.6 nM) and its bivalent construct exhibited a 100-fold higher functional affinity. The structure of the Nb113 in complex with rStx2aB was determined via X-ray crystallography. The crystal structure of the Nb113-rStx2aB complex revealed that five copies of Nb113 bind to the rStx2aB pentamer and that the Nb113 epitope overlaps with the Gb3 binding site, thereby providing a structural basis for the neutralization of Stx2a by Nb113 that was observed on Vero cells. Finally, the tandem-repeated, bivalent Nb1132 exhibits a higher toxin neutralization capacity compared to monovalent Nb113. CONCLUSIONS: The Nb of highest affinity for rStx2aB is also the best Stx2a and Stx2c toxin neutralizing Nb, especially in a bivalent format. This lead Nb neutralizes Stx2a by competing for the Gb3 receptor. The fusion of the bivalent Nb1132 with a serum albumin specific Nb is expected to combine high toxin neutralization potential with prolonged blood circulation.


Assuntos
Anticorpos Neutralizantes , Proteínas Recombinantes , Toxina Shiga II , Anticorpos de Domínio Único , Animais , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/fisiologia , Camelídeos Americanos/imunologia , Chlorocebus aethiops , Domínios Proteicos , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Toxina Shiga II/química , Toxina Shiga II/genética , Toxina Shiga II/imunologia , Toxina Shiga II/metabolismo , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/fisiologia , Células Vero
15.
Immunity ; 47(5): 990-1003.e9, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29166592

RESUMO

Understanding how broadly neutralizing antibodies (bnAbs) to HIV envelope (Env) develop during natural infection can help guide the rational design of an HIV vaccine. Here, we described a bnAb lineage targeting the Env V2 apex and the Ab-Env co-evolution that led to development of neutralization breadth. The lineage Abs bore an anionic heavy chain complementarity-determining region 3 (CDRH3) of 25 amino acids, among the shortest known for this class of Abs, and achieved breadth with only 10% nucleotide somatic hypermutation and no insertions or deletions. The data suggested a role for Env glycoform heterogeneity in the activation of the lineage germline B cell. Finally, we showed that localized diversity at key V2 epitope residues drove bnAb maturation toward breadth, mirroring the Env evolution pattern described for another donor who developed V2-apex targeting bnAbs. Overall, these findings suggest potential strategies for vaccine approaches based on germline-targeting and serial immunogen design.


Assuntos
Anticorpos Neutralizantes/fisiologia , Linhagem da Célula , Anticorpos Anti-HIV/fisiologia , Produtos do Gene env do Vírus da Imunodeficiência Humana/imunologia , Vacinas contra a AIDS/imunologia , Anticorpos Neutralizantes/química , Regiões Determinantes de Complementaridade , Anticorpos Anti-HIV/química , Humanos
16.
Nat Commun ; 8(1): 846, 2017 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-29018261

RESUMO

The aim of candidate universal influenza vaccines is to provide broad protection against influenza A and B viruses. Studies have demonstrated that broadly reactive antibodies require Fc-Fc gamma receptor interactions for optimal protection; however, the innate effector cells responsible for mediating this protection remain largely unknown. Here, we examine the roles of alveolar macrophages, natural killer cells, and neutrophils in antibody-mediated protection. We demonstrate that alveolar macrophages play a dominant role in conferring protection provided by both broadly neutralizing and non-neutralizing antibodies in mice. Our data also reveal the potential mechanisms by which alveolar macrophages mediate protection in vivo, namely antibody-induced inflammation and antibody-dependent cellular phagocytosis. This study highlights the importance of innate effector cells in establishing a broad-spectrum antiviral state, as well as providing a better understanding of how multiple arms of the immune system cooperate to achieve an optimal antiviral response following influenza virus infection or immunization.Broadly reactive antibodies that recognize influenza A virus HA can be protective, but the mechanism is not completely understood. Here, He et al. show that the inflammatory response and phagocytosis mediated by the interaction between protective antibodies and macrophages are essential for protection.


Assuntos
Anticorpos Neutralizantes/fisiologia , Vírus da Influenza A/imunologia , Influenza Humana/imunologia , Ativação de Macrófagos , Macrófagos Alveolares/fisiologia , Células A549 , Animais , Cães , Feminino , Células HEK293 , Hemaglutininas/imunologia , Humanos , Células Matadoras Naturais/fisiologia , Células Madin Darby de Rim Canino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/fisiologia , Infecções por Orthomyxoviridae/imunologia , Fagocitose , Receptores de IgG/metabolismo
17.
Nat Commun ; 7: 10844, 2016 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-26936020

RESUMO

The Fc region of HIV-1 Env-specific broadly neutralizing antibodies (bNAbs) is required for suppressing viraemia, through mechanisms which remain poorly understood. Here, we identify bNAbs that exert antibody-dependent cellular cytotoxicity (ADCC) in cell culture and kill HIV-1-infected lymphocytes through natural killer (NK) engagement. These antibodies target the CD4-binding site, the glycans/V3 and V1/V2 loops on gp120, or the gp41 moiety. The landscape of Env epitope exposure at the surface and the sensitivity of infected cells to ADCC vary considerably between viral strains. Efficient ADCC requires sustained cell surface binding of bNAbs to Env, and combining bNAbs allows a potent killing activity. Furthermore, reactivated infected cells from HIV-positive individuals expose heterogeneous Env epitope patterns, with levels that are often but not always sufficient to trigger killing by bNAbs. Our study delineates the parameters controlling ADCC activity of bNAbs, and supports the use of the most potent antibodies to clear the viral reservoir.


Assuntos
Anticorpos Neutralizantes/fisiologia , Anticorpos Antivirais/fisiologia , Linfócitos T CD4-Positivos/fisiologia , HIV-1/fisiologia , Animais , Linhagem Celular , Humanos
18.
J Infect Dis ; 212(6): 978-89, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25748320

RESUMO

The VD4 region from the Chlamydia trachomatis major outer membrane protein contains important neutralizing B-cell epitopes of relevance for antibody-mediated protection against genital tract infection. We developed a multivalent vaccine construct based on VD4s and their surrounding constant segments from serovars D, E, and F. Adjuvanted with cationic liposomes, this construct promoted strong immune responses to serovar-specific epitopes, the conserved LNPTIAG epitope and neutralized serovars D, E, and F. Vaccinated mice were protected against challenge, with protection defined as reduced bacterial numbers in vagina and prevention of pathological changes in the upper genital tract. Adoptive transfer of serum and T-cell depletion experiments demonstrated a dominant role for antibodies and CD4(+) T cells in the protective immune response. Integrating a multivalent VD4 construct into the sequence of the major outer membrane protein resulted in a protective and broadly neutralizing vaccine. Our findings emphasize the important role of antibodies in protection against Chlamydia trachomatis.


Assuntos
Anticorpos Antibacterianos/fisiologia , Anticorpos Neutralizantes/fisiologia , Proteínas da Membrana Bacteriana Externa/imunologia , Vacinas Bacterianas/imunologia , Infecções por Chlamydia/prevenção & controle , Chlamydia trachomatis/metabolismo , Animais , Anticorpos Antibacterianos/imunologia , Proteínas da Membrana Bacteriana Externa/química , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Infecções por Chlamydia/patologia , Chlamydia trachomatis/imunologia , Feminino , Camundongos
19.
J Clin Invest ; 125(3): 1255-68, 2015 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-25689254

RESUMO

The emergence and seasonal persistence of pathogenic H7N9 influenza viruses in China have raised concerns about the pandemic potential of this strain, which, if realized, would have a substantial effect on global health and economies. H7N9 viruses are able to bind to human sialic acid receptors and are also able to develop resistance to neuraminidase inhibitors without a loss in fitness. It is not clear whether prior exposure to circulating human influenza viruses or influenza vaccination confers immunity to H7N9 strains. Here, we demonstrate that 3 of 83 H3 HA-reactive monoclonal antibodies generated by individuals that had previously undergone influenza A virus vaccination were able to neutralize H7N9 viruses and protect mice against homologous challenge. The H7N9-neutralizing antibodies bound to the HA stalk domain but exhibited a difference in their breadth of reactivity to different H7 influenza subtypes. Mapping viral escape mutations suggested that these antibodies bind at least two different epitopes on the stalk region. Together, these results indicate that these broadly neutralizing antibodies may contribute to the development of therapies against H7N9 strains and may also be effective against pathogenic H7 strains that emerge in the future.


Assuntos
Anticorpos Neutralizantes/fisiologia , Anticorpos Antivirais/fisiologia , Subtipo H7N9 do Vírus da Influenza A/imunologia , Influenza Humana/prevenção & controle , Vacinação , Animais , Anticorpos Monoclonais/fisiologia , Reações Cruzadas , Cães , Feminino , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Vírus da Influenza A Subtipo H3N2/imunologia , Subtipo H7N9 do Vírus da Influenza A/genética , Vacinas contra Influenza , Influenza Humana/imunologia , Influenza Humana/virologia , Células Madin Darby de Rim Canino , Camundongos Endogâmicos BALB C , Testes de Neutralização , Mutação Puntual
20.
PLoS One ; 10(1): e0116882, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25635901

RESUMO

The plethora of virulence factors associated with Staphylococcus aureus make this bacterium an attractive candidate for a molecularly-designed epitope-focused vaccine. This approach, which necessitates the identification of neutralizing epitopes for incorporation into a vaccine construct, is being evaluated for pathogens where conventional approaches have failed to elicit protective humoral responses, like HIV-1 and malaria, but may also hold promise for pathogens like S. aureus, where the elicitation of humoral immunity against multiple virulence factors may be required for development of an effective vaccine. Among the virulence factors employed by S. aureus, animal model and epidemiological data suggest that alpha toxin, a multimeric ß-pore forming toxin like protective antigen from Bacillus anthracis, is particularly critical, yet no candidate neutralizing epitopes have been delineated in alpha toxin to date. We have previously shown that a linear determinant in the 2ß2-2ß3 loop of the pore forming domain of B. anthracis protective antigen is a linear neutralizing epitope. Antibody against this site is highly potent for neutralizing anthrax lethal toxin in vitro and for protection of rabbits in vivo from virulent B. anthracis. We hypothesized that sequences in the ß-pore of S. aureus alpha toxin that share structural and functional homology to ß-pore sequences in protective antigen would contain a similarly critical neutralizing epitope. Using an in vivo mapping strategy employing peptide immunogens, an optimized in vitro toxin neutralization assay, and an in vivo dermonecrosis model, we have now confirmed the presence of this epitope in alpha toxin, termed the pore neutralizing determinant. Antibody specific for this determinant neutralizes alpha toxin in vitro, and is highly effective for mitigating dermonecrosis and bacterial growth in a mouse model of S. aureus USA300 skin infection. The delineation of this linear neutralizing determinant in alpha toxin could facilitate the development of an epitope-focused vaccine against S. aureus.


Assuntos
Proteínas de Bactérias/química , Toxinas Bacterianas/química , Exotoxinas/química , Staphylococcus aureus/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antibacterianos/fisiologia , Anticorpos Neutralizantes/fisiologia , Citotoxicidade Celular Dependente de Anticorpos , Proteínas de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Vacinas Bacterianas/imunologia , Epitopos/imunologia , Exotoxinas/imunologia , Feminino , Humanos , Imunização Passiva , Células Jurkat , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Coelhos , Homologia de Sequência de Aminoácidos , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/prevenção & controle , Infecções Cutâneas Estafilocócicas/imunologia , Infecções Cutâneas Estafilocócicas/microbiologia , Infecções Cutâneas Estafilocócicas/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA