Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.875
Filtrar
Mais filtros

Eixos temáticos
Intervalo de ano de publicação
1.
Annu Rev Immunol ; 41: 255-275, 2023 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-36737596

RESUMO

The evolution of IgE in mammals added an extra layer of immune protection at body surfaces to provide a rapid and local response against antigens from the environment. The IgE immune response employs potent expulsive and inflammatory forces against local antigen provocation, at the risk of damaging host tissues and causing allergic disease. Two well-known IgE receptors, the high-affinity FcεRI and low-affinity CD23, mediate the activities of IgE. Unlike other known antibody receptors, CD23 also regulates IgE expression, maintaining IgE homeostasis. This mechanism evolved by adapting the function of the complement receptor CD21. Recent insights into the dynamic character of IgE structure, its resultant capacity for allosteric modulation, and the potential for ligand-induced dissociation have revealed previously unappreciated mechanisms for regulation of IgE and IgE complexes. We describe recent research, highlighting structural studies of the IgE network of proteins to analyze the uniquely versatile activities of IgE and anti-IgE biologics.


Assuntos
Produtos Biológicos , Receptores de IgE , Humanos , Animais , Receptores de IgE/química , Receptores de IgE/metabolismo , Imunoglobulina E/metabolismo , Receptores Fc , Mamíferos/metabolismo
2.
Annu Rev Immunol ; 35: 285-311, 2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-28446061

RESUMO

IgG antibodies mediate a diversity of immune functions by coupling of antigen specificity through the Fab domain to signal transduction via Fc-Fc receptor interactions. Indeed, balanced IgG signaling through type I and type II Fc receptors is required for the control of proinflammatory, anti-inflammatory, and immunomodulatory processes. In this review, we discuss the mechanisms that govern IgG-Fc receptor interactions, highlighting the diversity of Fc receptor-mediated effector functions that regulate immunity and inflammation as well as determine susceptibility to infection and autoimmunity and responsiveness to antibody-based therapeutics and vaccines.


Assuntos
Anticorpos/uso terapêutico , Doenças Autoimunes/imunologia , Imunoglobulina G/metabolismo , Imunoterapia/métodos , Infecções/imunologia , Receptores Fc/metabolismo , Animais , Doenças Autoimunes/terapia , Suscetibilidade a Doenças , Humanos , Imunidade Humoral , Infecções/terapia , Inflamação , Transdução de Sinais
3.
Cell ; 185(26): 4873-4886.e10, 2022 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-36513064

RESUMO

Respiratory syncytial virus (RSV) infection is a major cause of severe lower respiratory tract infection and death in young infants and the elderly. With no effective prophylactic treatment available, current vaccine candidates aim to elicit neutralizing antibodies. However, binding and neutralization have poorly predicted protection in the past, and accumulating data across epidemiologic cohorts and animal models collectively point to a role for additional antibody Fc-effector functions. To begin to define the humoral correlates of immunity against RSV, here we profiled an adenovirus 26 RSV-preF vaccine-induced humoral immune response in a group of healthy adults that were ultimately challenged with RSV. Protection from infection was linked to opsonophagocytic functions, driven by IgA and differentially glycosylated RSV-specific IgG profiles, marking a functional humoral immune signature of protection against RSV. Furthermore, Fc-modified monoclonal antibodies able to selectively recruit effector functions demonstrated significant antiviral control in a murine model of RSV.


Assuntos
Infecções por Vírus Respiratório Sincicial , Vírus Sincicial Respiratório Humano , Camundongos , Animais , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Anticorpos Neutralizantes , Anticorpos Antivirais , Imunoglobulina G , Fragmentos Fc das Imunoglobulinas , Proteínas Virais de Fusão
4.
Cell ; 184(17): 4414-4429.e19, 2021 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-34416146

RESUMO

Alphaviruses are emerging, mosquito-transmitted pathogens that cause musculoskeletal and neurological disease in humans. Although neutralizing antibodies that inhibit individual alphaviruses have been described, broadly reactive antibodies that protect against both arthritogenic and encephalitic alphaviruses have not been reported. Here, we identify DC2.112 and DC2.315, two pan-protective yet poorly neutralizing human monoclonal antibodies (mAbs) that avidly bind to viral antigen on the surface of cells infected with arthritogenic and encephalitic alphaviruses. These mAbs engage a conserved epitope in domain II of the E1 protein proximal to and within the fusion peptide. Treatment with DC2.112 or DC2.315 protects mice against infection by both arthritogenic (chikungunya and Mayaro) and encephalitic (Venezuelan, Eastern, and Western equine encephalitis) alphaviruses through multiple mechanisms, including inhibition of viral egress and monocyte-dependent Fc effector functions. These findings define a conserved epitope recognized by weakly neutralizing yet protective antibodies that could be targeted for pan-alphavirus immunotherapy and vaccine design.


Assuntos
Alphavirus/imunologia , Anticorpos Antivirais/imunologia , Sequência Conservada/imunologia , Epitopos/imunologia , Proteínas Virais/imunologia , Infecções por Alphavirus/imunologia , Infecções por Alphavirus/virologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Febre de Chikungunya/imunologia , Febre de Chikungunya/virologia , Vírus Chikungunya/imunologia , Chlorocebus aethiops , Mapeamento de Epitopos , Epitopos/química , Humanos , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Monócitos/metabolismo , Células Vero , Proteínas Virais/química , Liberação de Vírus
5.
Cell ; 184(3): 628-642.e10, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33476549

RESUMO

SARS-CoV-2 infection causes more severe disease in pregnant women compared to age-matched non-pregnant women. Whether maternal infection causes changes in the transfer of immunity to infants remains unclear. Maternal infections have previously been associated with compromised placental antibody transfer, but the mechanism underlying this compromised transfer is not established. Here, we used systems serology to characterize the Fc profile of influenza-, pertussis-, and SARS-CoV-2-specific antibodies transferred across the placenta. Influenza- and pertussis-specific antibodies were actively transferred. However, SARS-CoV-2-specific antibody transfer was significantly reduced compared to influenza- and pertussis-specific antibodies, and cord titers and functional activity were lower than in maternal plasma. This effect was only observed in third-trimester infection. SARS-CoV-2-specific transfer was linked to altered SARS-CoV-2-antibody glycosylation profiles and was partially rescued by infection-induced increases in IgG and increased FCGR3A placental expression. These results point to unexpected compensatory mechanisms to boost immunity in neonates, providing insights for maternal vaccine design.


Assuntos
Anticorpos Antivirais/imunologia , COVID-19/imunologia , Imunoglobulina G/imunologia , Troca Materno-Fetal/imunologia , Placenta/imunologia , Complicações Infecciosas na Gravidez/imunologia , SARS-CoV-2/imunologia , Adulto , Feminino , Humanos , Recém-Nascido , Gravidez , Terceiro Trimestre da Gravidez/imunologia , Receptores de IgG/imunologia , Células THP-1
6.
Cell ; 184(7): 1821-1835.e16, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33667349

RESUMO

Human monoclonal antibodies are safe, preventive, and therapeutic tools that can be rapidly developed to help restore the massive health and economic disruption caused by the coronavirus disease 2019 (COVID-19) pandemic. By single-cell sorting 4,277 SARS-CoV-2 spike protein-specific memory B cells from 14 COVID-19 survivors, 453 neutralizing antibodies were identified. The most potent neutralizing antibodies recognized the spike protein receptor-binding domain, followed in potency by antibodies that recognize the S1 domain, the spike protein trimer, and the S2 subunit. Only 1.4% of them neutralized the authentic virus with a potency of 1-10 ng/mL. The most potent monoclonal antibody, engineered to reduce the risk of antibody-dependent enhancement and prolong half-life, neutralized the authentic wild-type virus and emerging variants containing D614G, E484K, and N501Y substitutions. Prophylactic and therapeutic efficacy in the hamster model was observed at 0.25 and 4 mg/kg respectively in absence of Fc functions.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Antivirais/administração & dosagem , Linfócitos B/imunologia , COVID-19 , Convalescença , Células 3T3 , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Neutralizantes/isolamento & purificação , Anticorpos Antivirais/isolamento & purificação , Linfócitos B/citologia , COVID-19/imunologia , COVID-19/prevenção & controle , COVID-19/terapia , Chlorocebus aethiops , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Masculino , Camundongos , Glicoproteína da Espícula de Coronavírus/imunologia , Células Vero
7.
Cell ; 183(6): 1508-1519.e12, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-33207184

RESUMO

The urgent need for an effective SARS-CoV-2 vaccine has forced development to progress in the absence of well-defined correlates of immunity. While neutralization has been linked to protection against other pathogens, whether neutralization alone will be sufficient to drive protection against SARS-CoV-2 in the broader population remains unclear. Therefore, to fully define protective humoral immunity, we dissected the early evolution of the humoral response in 193 hospitalized individuals ranging from moderate to severe. Although robust IgM and IgA responses evolved in both survivors and non-survivors with severe disease, non-survivors showed attenuated IgG responses, accompanied by compromised Fcɣ receptor binding and Fc effector activity, pointing to deficient humoral development rather than disease-enhancing humoral immunity. In contrast, individuals with moderate disease exhibited delayed responses that ultimately matured. These data highlight distinct humoral trajectories associated with resolution of SARS-CoV-2 infection and the need for early functional humoral immunity.


Assuntos
COVID-19 , Imunidade Humoral , Imunoglobulina A/imunologia , Imunoglobulina M/imunologia , Receptores de IgG/imunologia , SARS-CoV-2/imunologia , COVID-19/imunologia , COVID-19/mortalidade , Feminino , Células HL-60 , Humanos , Masculino
8.
Cell ; 177(6): 1553-1565.e16, 2019 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-31104841

RESUMO

Enterovirus B (EV-B), a major proportion of the genus Enterovirus in the family Picornaviridae, is the causative agent of severe human infectious diseases. Although cellular receptors for coxsackievirus B in EV-B have been identified, receptors mediating virus entry, especially the uncoating process of echovirus and other EV-B remain obscure. Here, we found that human neonatal Fc receptor (FcRn) is the uncoating receptor for major EV-B. FcRn binds to the virus particles in the "canyon" through its FCGRT subunit. By obtaining multiple cryo-electron microscopy structures at different stages of virus entry at atomic or near-atomic resolution, we deciphered the underlying mechanisms of enterovirus attachment and uncoating. These structures revealed that different from the attachment receptor CD55, binding of FcRn to the virions induces efficient release of "pocket factor" under acidic conditions and initiates the conformational changes in viral particle, providing a structural basis for understanding the mechanisms of enterovirus entry.


Assuntos
Enterovirus Humano B/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe I/ultraestrutura , Receptores Fc/metabolismo , Receptores Fc/ultraestrutura , Capsídeo/metabolismo , Microscopia Crioeletrônica , Enterovirus , Enterovirus Humano B/patogenicidade , Infecções por Enterovirus/metabolismo , Antígenos de Histocompatibilidade Classe I/fisiologia , Humanos , Modelos Moleculares , Filogenia , Receptores Fc/fisiologia , Vírion , Internalização do Vírus
9.
Cell ; 178(1): 190-201.e11, 2019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-31204101

RESUMO

The placental transfer of maternal IgG is critical for infant protection against infectious pathogens. However, factors that modulate the placental transfer of IgG remain largely undefined. HIV-infected women have impaired placental IgG transfer, presenting a unique "disruption model" to define factors that modulate placental IgG transfer. We measured the placental transfer efficiency of maternal HIV and pathogen-specific IgG in US and Malawian HIV-infected mothers and their HIV-exposed uninfected and infected infants. We examined the role of maternal HIV disease progression, infant factors, placental Fc receptor expression, IgG subclass, and glycan signatures and their association with placental IgG transfer efficiency. Maternal IgG characteristics, such as binding to placentally expressed Fc receptors FcγRIIa and FcγRIIIa, and Fc region glycan profiles were associated with placental IgG transfer efficiency. Our findings suggest that Fc region characteristics modulate the selective placental transfer of IgG, with implications for maternal vaccine design and infant health.


Assuntos
Infecções por HIV/transmissão , HIV/genética , Imunoglobulina G/sangue , Transmissão Vertical de Doenças Infecciosas , Placenta/metabolismo , Complicações Infecciosas na Gravidez/virologia , Receptores de IgG/metabolismo , Estudos de Coortes , Progressão da Doença , Feminino , Glicosilação , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Fragmentos Fc das Imunoglobulinas/metabolismo , Lactente , Recém-Nascido , Malaui , Gravidez , Complicações Infecciosas na Gravidez/imunologia , Estados Unidos , Carga Viral/genética
10.
Immunity ; 57(6): 1215-1224.e6, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38788711

RESUMO

Malaria is a life-threatening disease of global health importance, particularly in sub-Saharan Africa. The growth inhibition assay (GIA) is routinely used to evaluate, prioritize, and quantify the efficacy of malaria blood-stage vaccine candidates but does not reliably predict either naturally acquired or vaccine-induced protection. Controlled human malaria challenge studies in semi-immune volunteers provide an unparalleled opportunity to robustly identify mechanistic correlates of protection. We leveraged this platform to undertake a head-to-head comparison of seven functional antibody assays that are relevant to immunity against the erythrocytic merozoite stage of Plasmodium falciparum. Fc-mediated effector functions were strongly associated with protection from clinical symptoms of malaria and exponential parasite multiplication, while the gold standard GIA was not. The breadth of Fc-mediated effector function discriminated clinical immunity following the challenge. These findings present a shift in the understanding of the mechanisms that underpin immunity to malaria and have important implications for vaccine development.


Assuntos
Anticorpos Antiprotozoários , Vacinas Antimaláricas , Malária Falciparum , Plasmodium falciparum , Humanos , Plasmodium falciparum/imunologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Anticorpos Antiprotozoários/imunologia , Vacinas Antimaláricas/imunologia , Adulto , Fragmentos Fc das Imunoglobulinas/imunologia , Merozoítos/imunologia , Eritrócitos/parasitologia , Eritrócitos/imunologia , Feminino , Masculino , Adulto Jovem
11.
Cell ; 172(1-2): 305-317.e10, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-29328918

RESUMO

Phagocytic receptors must diffuse laterally to become activated upon clustering by multivalent targets. Receptor diffusion, however, can be obstructed by transmembrane proteins ("pickets") that are immobilized by interacting with the cortical cytoskeleton. The molecular identity of these pickets and their role in phagocytosis have not been defined. We used single-molecule tracking to study the interaction between Fcγ receptors and CD44, an abundant transmembrane protein capable of indirect association with F-actin, hence likely to serve as a picket. CD44 tethers reversibly to formin-induced actin filaments, curtailing receptor diffusion. Such linear filaments predominate in the trailing end of polarized macrophages, where receptor mobility was minimal. Conversely, receptors were most mobile at the leading edge, where Arp2/3-driven actin branching predominates. CD44 binds hyaluronan, anchoring a pericellular coat that also limits receptor displacement and obstructs access to phagocytic targets. Force must be applied to traverse the pericellular barrier, enabling receptors to engage their targets.


Assuntos
Citoesqueleto de Actina/metabolismo , Membrana Celular/metabolismo , Receptores de Hialuronatos/metabolismo , Receptores Imunológicos/metabolismo , Adulto , Animais , Sítios de Ligação , Células COS , Células Cultivadas , Chlorocebus aethiops , Feminino , Humanos , Receptores de Hialuronatos/química , Receptores de Hialuronatos/genética , Ácido Hialurônico/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica
12.
Cell ; 174(1): 131-142.e13, 2018 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-29958103

RESUMO

Macrophages protect the body from damage and disease by targeting antibody-opsonized cells for phagocytosis. Though antibodies can be raised against antigens with diverse structures, shapes, and sizes, it is unclear why some are more effective at triggering immune responses than others. Here, we define an antigen height threshold that regulates phagocytosis of both engineered and cancer-specific antigens by macrophages. Using a reconstituted model of antibody-opsonized target cells, we find that phagocytosis is dramatically impaired for antigens that position antibodies >10 nm from the target surface. Decreasing antigen height drives segregation of antibody-bound Fc receptors from the inhibitory phosphatase CD45 in an integrin-independent manner, triggering Fc receptor phosphorylation and promoting phagocytosis. Our work shows that close contact between macrophage and target is a requirement for efficient phagocytosis, suggesting that therapeutic antibodies should target short antigens in order to trigger Fc receptor activation through size-dependent physical segregation.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos/química , Macrófagos/imunologia , Proteínas Opsonizantes/metabolismo , Fagocitose , Animais , Anticorpos Monoclonais/química , Antígenos/genética , Antígenos/imunologia , Antígeno Carcinoembrionário/química , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/imunologia , Edição de Genes , Integrinas/metabolismo , Antígenos Comuns de Leucócito/química , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/imunologia , Macrófagos/citologia , Camundongos , Proteínas Opsonizantes/química , Fosforilação , Células RAW 264.7 , Receptores Fc/imunologia , Receptores Fc/metabolismo , Lipossomas Unilamelares/química
13.
Immunity ; 56(5): 1046-1063.e7, 2023 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-36948194

RESUMO

Immunoglobulin G (IgG) antibodies are major drivers of inflammation during infectious and autoimmune diseases. In pooled serum IgG (IVIg), however, antibodies have a potent immunomodulatory and anti-inflammatory activity, but how this is mediated is unclear. We studied IgG-dependent initiation of resolution of inflammation in cytokine- and autoantibody-driven models of rheumatoid arthritis and found IVIg sialylation inhibited joint inflammation, whereas inhibition of osteoclastogenesis was sialic acid independent. Instead, IVIg-dependent inhibition of osteoclastogenesis was abrogated in mice lacking receptors Dectin-1 or FcγRIIb. Atomistic molecular dynamics simulations and super-resolution microscopy revealed that Dectin-1 promoted FcγRIIb membrane conformations that allowed productive IgG binding and enhanced interactions with mouse and human IgG subclasses. IVIg reprogrammed monocytes via FcγRIIb-dependent signaling that required Dectin-1. Our data identify a pathogen-independent function of Dectin-1 as a co-inhibitory checkpoint for IgG-dependent inhibition of mouse and human osteoclastogenesis. These findings may have implications for therapeutic targeting of autoantibody and cytokine-driven inflammation.


Assuntos
Artrite Reumatoide , Imunoglobulinas Intravenosas , Lectinas Tipo C , Receptores de IgG , Animais , Humanos , Camundongos , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/imunologia , Membrana Celular/metabolismo , Imunoglobulinas Intravenosas/administração & dosagem , Lectinas Tipo C/metabolismo , Camundongos Endogâmicos C57BL , Osteoclastos/metabolismo , Processamento de Proteína Pós-Traducional , Receptores de IgG/metabolismo
14.
Immunity ; 55(2): 355-365.e4, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35090580

RESUMO

SARS-CoV-2 mRNA vaccines confer robust protection against COVID-19, but the emergence of variants has generated concerns regarding the protective efficacy of the currently approved vaccines, which lose neutralizing potency against some variants. Emerging data suggest that antibody functions beyond neutralization may contribute to protection from the disease, but little is known about SARS-CoV-2 antibody effector functions. Here, we profiled the binding and functional capacity of convalescent antibodies and Moderna mRNA-1273 COVID-19 vaccine-induced antibodies across SARS-CoV-2 variants of concern (VOCs). Although the neutralizing responses to VOCs decreased in both groups, the Fc-mediated responses were distinct. In convalescent individuals, although antibodies exhibited robust binding to VOCs, they showed compromised interactions with Fc-receptors. Conversely, vaccine-induced antibodies also bound robustly to VOCs but continued to interact with Fc-receptors and mediate antibody effector functions. These data point to a resilience in the mRNA-vaccine-induced humoral immune response that may continue to offer protection from SARS-CoV-2 VOCs independent of neutralization.


Assuntos
Vacina de mRNA-1273 contra 2019-nCoV/imunologia , Anticorpos Antivirais/imunologia , COVID-19/metabolismo , COVID-19/prevenção & controle , Receptores Fc/metabolismo , SARS-CoV-2/imunologia , Vacina de mRNA-1273 contra 2019-nCoV/administração & dosagem , Adulto , Anticorpos Neutralizantes/imunologia , Reações Cruzadas/imunologia , Feminino , Interações Hospedeiro-Patógeno , Humanos , Masculino , Pessoa de Meia-Idade , Testes de Neutralização , Ligação Proteica , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinação , Adulto Jovem
15.
Cell ; 167(2): 433-443.e14, 2016 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-27667685

RESUMO

While a third of the world carries the burden of tuberculosis, disease control has been hindered by a lack of tools, including a rapid, point-of-care diagnostic and a protective vaccine. In many infectious diseases, antibodies (Abs) are powerful biomarkers and important immune mediators. However, in Mycobacterium tuberculosis (Mtb) infection, a discriminatory or protective role for humoral immunity remains unclear. Using an unbiased antibody profiling approach, we show that individuals with latent tuberculosis infection (Ltb) and active tuberculosis disease (Atb) have distinct Mtb-specific humoral responses, such that Ltb infection is associated with unique Ab Fc functional profiles, selective binding to FcγRIII, and distinct Ab glycosylation patterns. Moreover, compared to Abs from Atb, Abs from Ltb drove enhanced phagolysosomal maturation, inflammasome activation, and, most importantly, macrophage killing of intracellular Mtb. Combined, these data point to a potential role for Fc-mediated Ab effector functions, tuned via differential glycosylation, in Mtb control.


Assuntos
Anticorpos Antibacterianos/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade Humoral , Tuberculose Latente/imunologia , Mycobacterium tuberculosis/imunologia , Adulto , Feminino , Glicosilação , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Ativação de Macrófagos , Masculino , Pessoa de Meia-Idade , Polissacarídeos/imunologia , Análise Serial de Proteínas , Receptores de IgG/imunologia , Adulto Jovem
16.
Immunity ; 54(4): 815-828.e5, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33852832

RESUMO

Protective Ebola virus (EBOV) antibodies have neutralizing activity and induction of antibody constant domain (Fc)-mediated innate immune effector functions. Efforts to enhance Fc effector functionality often focus on maximizing antibody-dependent cellular cytotoxicity, yet distinct combinations of functions could be critical for antibody-mediated protection. As neutralizing antibodies have been cloned from EBOV disease survivors, we sought to identify survivor Fc effector profiles to help guide Fc optimization strategies. Survivors developed a range of functional antibody responses, and we therefore applied a rapid, high-throughput Fc engineering platform to define the most protective profiles. We generated a library of Fc variants with identical antigen-binding fragments (Fabs) from an EBOV neutralizing antibody. Fc variants with antibody-mediated complement deposition and moderate natural killer (NK) cell activity demonstrated complete protective activity in a stringent in vivo mouse model. Our findings highlight the importance of specific effector functions in antibody-mediated protection, and the experimental platform presents a generalizable resource for identifying correlates of immunity to guide therapeutic antibody design.


Assuntos
Ebolavirus/imunologia , Doença pelo Vírus Ebola/imunologia , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos Fc das Imunoglobulinas/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Formação de Anticorpos/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Feminino , Células HEK293 , Doença pelo Vírus Ebola/virologia , Humanos , Imunoglobulina G/imunologia , Camundongos Endogâmicos BALB C , Receptores Fc/imunologia
17.
Immunity ; 54(9): 2143-2158.e15, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34453881

RESUMO

Neutralizing antibodies (NAbs) are effective in treating COVID-19, but the mechanism of immune protection is not fully understood. Here, we applied live bioluminescence imaging (BLI) to monitor the real-time effects of NAb treatment during prophylaxis and therapy of K18-hACE2 mice intranasally infected with SARS-CoV-2-nanoluciferase. Real-time imaging revealed that the virus spread sequentially from the nasal cavity to the lungs in mice and thereafter systemically to various organs including the brain, culminating in death. Highly potent NAbs from a COVID-19 convalescent subject prevented, and also effectively resolved, established infection when administered within three days. In addition to direct neutralization, depletion studies indicated that Fc effector interactions of NAbs with monocytes, neutrophils, and natural killer cells were required to effectively dampen inflammatory responses and limit immunopathology. Our study highlights that both Fab and Fc effector functions of NAbs are essential for optimal in vivo efficacy against SARS-CoV-2.


Assuntos
Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/metabolismo , Encéfalo/patologia , COVID-19/imunologia , Pulmão/patologia , SARS-CoV-2/fisiologia , Testículo/patologia , Enzima de Conversão de Angiotensina 2/genética , Animais , Anticorpos Neutralizantes/genética , Anticorpos Antivirais/genética , Encéfalo/virologia , COVID-19/terapia , Células Cultivadas , Modelos Animais de Doenças , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Luciferases/genética , Medições Luminescentes , Pulmão/virologia , Masculino , Camundongos , Camundongos Transgênicos , Testículo/virologia
18.
Immunity ; 53(2): 290-302.e6, 2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32768386

RESUMO

CD47 acts as a "don't eat me" signal that protects cells from phagocytosis by binding and activating its receptor SIPRA on macrophages. CD47 suppresses multiple different pro-engulfment "eat me" signals, including immunoglobulin G (IgG), complement, and calreticulin, on distinct target cells. This complexity has limited understanding of how the "don't eat me" signal is transduced biochemically. Here, we utilized a reconstituted system with a defined set of signals to interrogate the mechanism of SIRPA activation and its downstream targets. CD47 ligation altered SIRPA localization, positioning SIRPA for activation at the phagocytic synapse. At the phagocytic synapse, SIRPA inhibited integrin activation to limit macrophage spreading across the surface of the engulfment target. Chemical reactivation of integrin bypassed CD47-mediated inhibition and rescued engulfment, similar to the effect of a CD47 function-blocking antibody. Thus, the CD47-SIRPA axis suppresses phagocytosis by inhibiting inside-out activation of integrin signaling in the macrophage, with implications to cancer immunotherapy applications.


Assuntos
Antígeno CD47/metabolismo , Integrinas/metabolismo , Macrófagos/imunologia , Fagocitose/imunologia , Receptores Imunológicos/metabolismo , Animais , Calreticulina/imunologia , Linhagem Celular , Proteínas do Sistema Complemento/imunologia , Células HEK293 , Humanos , Imunoglobulina G/imunologia , Ativação de Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilserinas/imunologia , Células RAW 264.7 , Transdução de Sinais/imunologia
19.
Immunity ; 52(6): 1039-1056.e9, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32392463

RESUMO

The phenotypic and functional dichotomy between IRF8+ type 1 and IRF4+ type 2 conventional dendritic cells (cDC1s and cDC2s, respectively) is well accepted; it is unknown how robust this dichotomy is under inflammatory conditions, when additionally monocyte-derived cells (MCs) become competent antigen-presenting cells (APCs). Using single-cell technologies in models of respiratory viral infection, we found that lung cDC2s acquired expression of the Fc receptor CD64 shared with MCs and of IRF8 shared with cDC1s. These inflammatory cDC2s (inf-cDC2s) were superior in inducing CD4+ T helper (Th) cell polarization while simultaneously presenting antigen to CD8+ T cells. When carefully separated from inf-cDC2s, MCs lacked APC function. Inf-cDC2s matured in response to cell-intrinsic Toll-like receptor and type 1 interferon receptor signaling, upregulated an IRF8-dependent maturation module, and acquired antigens via convalescent serum and Fc receptors. Because hybrid inf-cDC2s are easily confused with monocyte-derived cells, their existence could explain why APC functions have been attributed to MCs.


Assuntos
Plasticidade Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Imunidade , Macrófagos/imunologia , Macrófagos/metabolismo , Infecções por Respirovirus/etiologia , Apresentação de Antígeno , Biomarcadores , Suscetibilidade a Doenças , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Imunofenotipagem , Interferon Tipo I/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Especificidade de Órgãos/imunologia , Receptores Fc/metabolismo , Infecções por Respirovirus/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Fatores de Transcrição , Viroses/genética , Viroses/imunologia , Viroses/metabolismo , Viroses/virologia
20.
Mol Cell ; 81(4): 859-869.e8, 2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-33352108

RESUMO

Active DNA demethylation via ten-eleven translocation (TET) family enzymes is essential for epigenetic reprogramming in cell state transitions. TET enzymes catalyze up to three successive oxidations of 5-methylcytosine (5mC), generating 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), or 5-carboxycytosine (5caC). Although these bases are known to contribute to distinct demethylation pathways, the lack of tools to uncouple these sequential oxidative events has constrained our mechanistic understanding of the role of TETs in chromatin reprogramming. Here, we describe the first application of biochemically engineered TET mutants that unlink 5mC oxidation steps, examining their effects on somatic cell reprogramming. We show that only TET enzymes proficient for oxidation to 5fC/5caC can rescue the reprogramming potential of Tet2-deficient mouse embryonic fibroblasts. This effect correlated with rapid DNA demethylation at reprogramming enhancers and increased chromatin accessibility later in reprogramming. These experiments demonstrate that DNA demethylation through 5fC/5caC has roles distinct from 5hmC in somatic reprogramming to pluripotency.


Assuntos
5-Metilcitosina/metabolismo , Reprogramação Celular , Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos/metabolismo , Elementos Facilitadores Genéticos , Epigênese Genética , Fibroblastos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Dioxigenases , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Mutação , Células NIH 3T3 , Proteínas Proto-Oncogênicas/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa