Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Infect Dis ; 219(1): 59-67, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30107412

RESUMEN

Background: Transplacental respiratory syncytial virus (RSV) antibody transfer has been characterized, but little is known about the protective effect of breast milk RSV-specific antibodies. Serum antibodies against the prefusion RSV fusion protein (pre-F) exhibit high neutralizing activity. We investigate protection of breast milk pre-F antibodies against RSV acute respiratory infection (ARI). Methods: Breast milk at 1, 3, and 6 months postpartum and midnasal swabs during infant illness episodes were collected in mother-infant pairs in Nepal. One hundred seventy-four infants with and without RSV ARI were matched 1:1 by risk factors for RSV ARI. Pre-F immunoglobulin A (IgA) and immunoglobulin G (IgG) antibody levels were measured in breast milk. Results: The median breast milk pre-F IgG antibody concentration before illness was lower in mothers of infants with RSV ARI (1.4 [interquartile range {IQR}, 1.1-1.6] log10 ng/mL) than without RSV ARI (1.5 [IQR, 1.3-1.8] log10 ng/mL) (P = .001). There was no difference in median maternal pre-F IgA antibody concentrations in cases vs controls (1.7 [IQR, 0.0-2.2] log10 ng/mL vs 1.7 [IQR, 1.2-2.2] log10 ng/mL, respectively; P = .58). Conclusions: Low breast milk pre-F IgG antibodies before RSV ARI support a potential role for pre-F IgG as a correlate of protection against RSV ARI. Induction of breast milk pre-F IgG may be a mechanism of protection for maternal RSV vaccines.


Asunto(s)
Inmunoglobulina G/análisis , Leche Humana/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Adulto , Anticuerpos Antivirales/análisis , Estudios de Cohortes , Femenino , Humanos , Inmunoglobulina A/análisis , Lactante , Masculino , Nepal , Infecciones por Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales de Fusión/inmunología , Adulto Joven
2.
Immunol Rev ; 268(1): 74-87, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26497514

RESUMEN

Fc receptors (FcR) are expressed on immune cells and bind to the Fc tail of antibodies. This interaction is essential for FcR-mediated signaling and triggering of cellular effector functions. FcR activation is tightly regulated to prevent immune responses by non-antigen bound antibodies or in the absence of 'danger signals'. FcR activity may be modulated at the plasma membrane via cross-talk with integrins. In addition, cytokines at the site of infection/inflammation can increase FcR avidity, a process referred to as inside-out signaling. This regulatory mechanism has been described for FcγRI (CD64), FcγRIIa (CD32a), and FcαRI (CD89) and is also well-known for integrins. Key cellular events during inside-out signaling are (de)phosphorylation, clustering, cytoskeleton rearrangements, and conformational changes. The latter can be studied with antibodies that specifically recognize epitopes exposed by the active (high affinity) or inactive (low affinity) state of the FcR. These antibodies are important tools to investigate the role of FcR activation in disease settings. Research on FcR has gained momentum with the rise of monoclonal antibodies (mAb) entering the clinic for the treatment of cancer and other diseases. The clinical outcome of mAb therapy may be improved by increasing FcR avidity by cytokine stimulation.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inmunomodulación , Receptores Fc/metabolismo , Transducción de Señal , Animales , Anticuerpos Monoclonales/farmacología , Antígenos CD/química , Antígenos CD/genética , Antígenos CD/metabolismo , Humanos , Inmunoglobulinas/inmunología , Inmunoglobulinas/metabolismo , Inmunomodulación/efectos de los fármacos , Integrinas/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores Fc/química , Receptores Fc/genética , Receptores de IgG/química , Receptores de IgG/genética , Receptores de IgG/metabolismo , Transducción de Señal/efectos de los fármacos
3.
Cell Microbiol ; 15(10): 1753-65, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23601501

RESUMEN

Neutrophil recruitment is essential in clearing pneumococcal infections. The first step in neutrophil extravasation involves the interaction between P-selectin on activated endothelium and P-Selectin Glycoprotein 1 (PSGL-1) on neutrophils. Here, we identify pneumococcal Zinc metalloproteinase C as a potent inhibitor of PSGL-1. ZmpC degrades the N-terminal domain of PSGL-1, thereby disrupting the initial rolling of neutrophils on activated human umbilical vein endothelial cells. Furthermore, mice infected with wild-type strain in the model of pneumococcal pneumonia showed lower lungs neutrophil infiltration compare to animals infected with ZmpC mutant. In addition, we confirmed the association of zmpC with serotype 8 and 11A and found it to be associated with serotype 33F as well. In conclusion, wereport PSGL-1 as a novel target for ZmpC and show that ZmpC inhibits neutrophil extravasation during pneumococcal pneumonia.


Asunto(s)
Interacciones Huésped-Patógeno , Evasión Inmune , Glicoproteínas de Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Neutrófilos/inmunología , Streptococcus pneumoniae/fisiología , Animales , Adhesión Celular , Modelos Animales de Enfermedad , Células Endoteliales/fisiología , Eliminación de Gen , Humanos , Pulmón/inmunología , Pulmón/patología , Metaloendopeptidasas/genética , Ratones , Neumonía Neumocócica/patología , Proteolisis , Streptococcus pneumoniae/genética
5.
Front Immunol ; 11: 1701, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32849597

RESUMEN

Respiratory syncytial virus (RSV) infections represent a major burden of disease in infants and are the second most prevalent cause of death worldwide. Human milk immunoglobulins provide protection against RSV. However, many infants depend on processed bovine milk-based nutrition, which lacks intact immunoglobulins. We investigated the potential of bovine antibodies to neutralize human RSV and facilitate-cell immune activation. We show cow's milk IgG (bIgG) and Intravenous Immunoglobulin (IVIG) have a similar RSV neutralization capacity, even though bIgG has a lower pre-F to post-F binding ratio compared to human IVIG, with the majority of bIgG binding to pre-F. RSV is better neutralized with human IVIG. Consequently, we enriched RSV specific T cells by culturing human PBMC with a mixture of RSV peptides, and used these T cells to study the effect of bIgG and IVIG on the activation of pre-F-pecific T cells. bIgG facilitated in vitro T cell activation in a similar manner as IVIG. Moreover, bIgG was able to mediate T cell activation and internalization of pathogens, which are prerequisites for inducing an adaptive viral response. Using in vivo mouse experiments, we showed that bIgG is able to bind the murine activating IgG Fc Receptors (FcγR), but not the inhibiting FcγRII. Intranasal administration of the monoclonal antibody palivizumab, but also of bIgG and IVIG prevented RSV infection in mice. The concentration of bIgG needed to prevent infection was ~5-fold higher compared to IVIG. In conclusion, the data presented here indicate that functionally active bIgG facilitates adaptive antiviral T cell responses and prevents RSV infection in vitro and in vivo.


Asunto(s)
Antivirales/farmacología , Inmunoglobulina G/farmacología , Activación de Linfocitos/efectos de los fármacos , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Animales , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Antivirales/aislamiento & purificación , Bovinos , Línea Celular , Calostro/inmunología , Modelos Animales de Enfermedad , Epítopos , Femenino , Interacciones Huésped-Patógeno , Humanos , Inmunoglobulina G/aislamiento & purificación , Inmunoglobulinas Intravenosas/farmacología , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis/efectos de los fármacos , Embarazo , Receptores de IgG/genética , Receptores de IgG/metabolismo , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/inmunología , Virus Sincitial Respiratorio Humano/patogenicidad , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/virología
6.
MAbs ; 10(3): 453-462, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29553863

RESUMEN

Respiratory syncytial virus (RSV) infection is a leading cause of hospitalization and mortality in young children. Protective therapy options are limited. Currently, palivizumab, a monoclonal IgG1 antibody, is the only licensed drug for RSV prophylaxis, although other IgG antibody candidates are being evaluated. However, at the respiratory mucosa, IgA antibodies are most abundant and act as the first line of defense against invading pathogens. Therefore, it would be logical to explore the potential of recombinant human IgA antibodies to protect against viral respiratory infection, but very little research on the topic has been published. Moreover, it is unknown whether human antibodies of the IgA isotype are better suited than those of the IgG isotype as antiviral drugs to combat respiratory infections. To address this, we generated various human IgA antibody formats of palivizumab and motavizumab, two well-characterized human IgG1 anti-RSV antibodies. We evaluated their efficacy to prevent RSV infection in vitro and in vivo and found similar, but somewhat decreased efficacy for different IgA subclasses and formats. Thus, reformatting palivizumab or motavizumab into IgA reduces the antiviral potency of either antibody. Moreover, our results indicate that the efficacy of intranasal IgA prophylaxis against RSV infection in human FcαRI transgenic mice is independent of Fc receptor expression.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Anticuerpos Antivirales , Inmunoglobulina A , Inmunoglobulina G , Palivizumab , Infecciones por Virus Sincitial Respiratorio , Virus Sincitiales Respiratorios/inmunología , Animales , Anticuerpos Monoclonales Humanizados/genética , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Antivirales/genética , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/farmacología , Línea Celular , Humanos , Inmunoglobulina A/genética , Inmunoglobulina A/inmunología , Inmunoglobulina A/farmacología , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Inmunoglobulina G/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Palivizumab/genética , Palivizumab/inmunología , Palivizumab/farmacología , Ingeniería de Proteínas , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/genética , Infecciones por Virus Sincitial Respiratorio/inmunología
7.
MAbs ; 8(1): 87-98, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26466856

RESUMEN

Antibody therapy is a validated treatment approach for several malignancies. All currently clinically applied therapeutic antibodies (Abs) are of the IgG isotype. However, not all patients respond to this therapy and relapses can occur. IgA represents an alternative isotype for antibody therapy that engages FcαRI expressing myeloid effector cells, such as neutrophils and monocytes. IgA Abs have been shown to effectively kill tumor cells both in vitro and in vivo. However, due to the short half-life of IgA Abs in mice, daily injections are required to reach an effect comparable to IgG Abs. The relatively long half-life of IgG Abs and serum albumin arises from their capability of interacting with the neonatal Fc receptor (FcRn). As IgA Abs lack a binding site for FcRn, we generated IgA Abs with the variable regions of the Her2-specific Ab trastuzumab and attached an albumin-binding domain (ABD) to the heavy or light chain (HCABD/LCABD) to extend their serum half-life. These modified Abs were able to bind albumin from different species in vitro. Furthermore, tumor cell lysis of IgA-Her2-LCABD Abs in vitro was similar to unmodified IgA-Her2 Abs. Pharmacokinetic studies in mice revealed that the serum exposure and half-life of the modified IgA-Her2 Abs was extended. In a xenograft mouse model, the modified IgA1 Abs exhibited a slightly, but significantly, improved anti-tumor response compared to the unmodified Ab. In conclusion, empowering IgA Abs with albumin-binding capacity results in in vitro and in vivo functional Abs with an enhanced exposure and prolonged half-life.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunoglobulina A , Neoplasias Experimentales/tratamiento farmacológico , Receptor ErbB-2/antagonistas & inhibidores , Receptores Fc/metabolismo , Trastuzumab , Animales , Femenino , Semivida , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Inmunoglobulina A/química , Inmunoglobulina A/genética , Inmunoglobulina A/farmacología , Ratones , Ratones Transgénicos , Neoplasias Experimentales/sangre , Estructura Terciaria de Proteína , Receptores Fc/genética , Trastuzumab/química , Trastuzumab/genética , Trastuzumab/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA