Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Cancer Ther ; 23(2): 187-198, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-37828725

RESUMEN

Head and neck squamous cell carcinoma (HNSCC) is a solid tumor type that arises in the squamous epithelial cells lining the mucosal surfaces of the upper aerodigestive tract. Long-term survival of patients with advanced disease stage remains disappointing with current treatment options. We show that tissue factor is abundantly expressed on patient-derived HNSCC cell lines, xenograft tumor material, and tumor biopsies from patients with HNSCC. Tisotumab vedotin (TV) is an antibody-drug conjugate (ADC) directed to tissue factor, a protein expressed in many solid tumors. HNSCC cells and xenograft tumors were efficiently eliminated in vitro and in vivo with TV-monotherapy compared with treatment with a control antibody conjugated to monomethyl auristatin E (MMAE). Antitumor activity of TV was also tested in vivo in combination with chemoradiotherapy, standard of care for patients with advanced stage HNSCC tumors outside the oral cavity. Preclinical studies showed that by adding TV to chemoradiotherapy, survival was markedly improved, and TV, not radiotherapy or chemotherapy, was the main driver of antitumor activity. Interestingly, TV-induced cell death in xenograft tumors showed an influx of macrophages indicative of a potential immune-mediated mode-of-action. In conclusion, on the basis of these preclinical data, TV may be a novel treatment modality for patients suffering from head and neck cancer and is hypothesized to improve efficacy of chemoradiotherapy. SIGNIFICANCE: This work shows preclinical in vitro and in vivo antitumor activity of the antibody-drug conjugate Tisotumab vedotin in head and neck cancer models, and enhanced activity in combination with chemoradiotherapy, supporting further clinical development for this cancer type.


Asunto(s)
Neoplasias de Cabeza y Cuello , Inmunoconjugados , Humanos , Línea Celular Tumoral , Quimioradioterapia , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Tromboplastina , Ensayos Antitumor por Modelo de Xenoinjerto , Animales
2.
Front Immunol ; 13: 732977, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35371001

RESUMEN

Immunoglobulin A (IgA) is generally considered as a non-inflammatory regulator of mucosal immunity, and its importance in diversifying the gut microbiota is increasingly appreciated. IgA autoantibodies have been found in several autoimmune or chronic inflammatory diseases, but their role in pathophysiology is ill-understood. IgA can interact with the Fc receptor FcαRI on immune cells. We now established a novel IgA autoimmune blistering model, which closely resembles the human disease linear IgA bullous disease (LABD) by using genetically modified mice that produce human IgA and express human FcαRI. Intravital microscopy demonstrated that presence of IgA anti-collagen XVII, - the auto-antigen in LABD-, resulted in neutrophil activation and extravasation from blood vessels into skin tissue. Continued exposure to anti-collagen XVII IgA led to massive neutrophil accumulation, severe tissue damage and blister formation. Importantly, treatment with anti-FcαRI monoclonal antibodies not only prevented disease, but was also able to resolve existing inflammation and tissue damage. Collectively, our data reveal a novel role of neutrophil FcαRI in IgA autoantibody-mediated disease and identify FcαRI as promising new therapeutic target to resolve chronic inflammation and tissue damage.


Asunto(s)
Inmunoglobulina A , Receptores Fc , Animales , Anticuerpos Monoclonales/uso terapéutico , Autoanticuerpos , Inflamación/tratamiento farmacológico , Ratones
3.
Int J Mol Sci ; 22(20)2021 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-34681717

RESUMEN

The immunosuppressive character of head and neck cancers may explain the relatively low response rates to antibody therapy targeting a tumor antigen, such as cetuximab, and anti-PD-1 checkpoint inhibition. Immunostimulatory agents that overcome tumor-derived inhibitory signals could augment therapeutic efficacy, thereby enhancing tumor elimination and improving patient survival. Here, we demonstrate that cetuximab treatment combined with immunostimulatory agonists for Toll-like receptor (TLR) 2 induces profound immune responses. Natural killer (NK) cells, isolated from healthy individuals or patients with head and neck cancer, harbored enhanced cytotoxic capacity and increased tumor-killing potential in vitro. Additionally, combination treatment increased the release of several pro-inflammatory cytokines and chemokines by NK cells. Tumor-bearing mice that received cetuximab and the TLR2 ligand Pam3CSK4 showed increased infiltration of immune cells into the tumors compared to mice that received cetuximab monotherapy, resulting in a significant delay in tumor growth or even complete tumor regression. Moreover, combination treatment resulted in improved overall survival in vivo. In conclusion, combining tumor-targeting antibody-based immunotherapy with TLR stimulation represents a promising treatment strategy to improve the clinical outcomes of cancer patients. This treatment could well be applied together with other therapeutic strategies such as anti-PD-(L)1 checkpoint inhibition to further overcome immunosuppression.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Neoplasias de Cabeza y Cuello/terapia , Células Asesinas Naturales/inmunología , Receptor Toll-Like 2/agonistas , Animales , Línea Celular Tumoral , Cetuximab/farmacología , Cetuximab/uso terapéutico , Citocinas/metabolismo , Quimioterapia Combinada , Femenino , Humanos , Inmunoterapia , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Lipopéptidos/farmacología , Lipopéptidos/uso terapéutico , Ratones , Ratones Desnudos , Receptores de IgG/agonistas , Receptores de IgG/metabolismo , Receptor Toll-Like 2/metabolismo , Trasplante Heterólogo
4.
J Clin Invest ; 131(6)2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33561014

RESUMEN

Most clinically used anticancer mAbs are of the IgG isotype, which can eliminate tumor cells through NK cell-mediated antibody-dependent cellular cytotoxicity and macrophage-mediated antibody-dependent phagocytosis. IgG, however, ineffectively recruits neutrophils as effector cells. IgA mAbs induce migration and activation of neutrophils through the IgA Fc receptor (FcαRI) but are unable to activate NK cells and have poorer half-life. Here, we combined the agonistic activity of IgG mAbs and FcαRI targeting in a therapeutic bispecific antibody format. The resulting TrisomAb molecules recruited NK cells, macrophages, and neutrophils as effector cells for eradication of tumor cells in vitro and in vivo. Moreover, TrisomAb had long in vivo half-life and strongly decreased B16F10gp75 tumor outgrowth in mice. Importantly, neutrophils of colorectal cancer patients effectively eliminated tumor cells in the presence of anti-EGFR TrisomAb but were less efficient in mediating killing in the presence of IgG anti-EGFR mAb (cetuximab). The clinical application of TrisomAb may provide potential alternatives for cancer patients who do not benefit from current IgG mAb therapy.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antineoplásicos Inmunológicos/farmacología , Neutrófilos/inmunología , Animales , Anticuerpos Biespecíficos/farmacología , Anticuerpos Monoclonales/farmacología , Antígenos CD/inmunología , Línea Celular Tumoral , Cetuximab/farmacología , Femenino , Células HCT116 , Humanos , Inmunoglobulina G/inmunología , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Masculino , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Inmunológicos , Receptores Fc/inmunología
5.
Front Immunol ; 9: 3191, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30766540

RESUMEN

IgA binding to FcαRI (CD89) is rapidly enhanced by cytokine induced inside-out signaling. Dephosphorylation of serine 263 in the intracellular tail of FcαRI by PP2A and PI3K activation are instrumental in this process. To further investigate these signaling pathways, we targeted downstream kinases of PI3K. Our experiments revealed that PI3K activates PKCζ, which subsequently inhibits GSK-3, a constitutively active kinase in resting cells and found here to be associated with FcαRI. We propose that GSK-3 maintains FcαRI in an inactive state at homeostatic conditions. Upon cytokine stimulation, GSK-3 is inactivated through a PI3K-PKCζ pathway, preventing the maintenance of phosphorylated inactive FcαRI. The concomitantly activated PP2A is then able to dephosphorylate and activate FcαRI. Moreover, FRAP and FLIP studies showed that FcαRI activation coincides with an increased mobile fraction of the receptor. This can enhance FcαRI valency and contribute to stronger avidity for IgA immune complexes. This tightly regulated inside-out signaling pathway allows leukocytes to respond rapidly and efficiently to their environment and could be exploited to enhance the efficacy of future IgA therapeutics.


Asunto(s)
Citocinas/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Proteína Quinasa C/metabolismo , Receptores Fc/metabolismo , Transducción de Señal , Animales , Membrana Celular/metabolismo , Humanos , Inmunoglobulina A/inmunología , Inmunoglobulina A/metabolismo , Ratones , Modelos Biológicos , Fosforilación , Unión Proteica
6.
Eur J Immunol ; 47(10): 1835-1845, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28736835

RESUMEN

The cross-linking of the IgA Fc receptor (FcαRI) by IgA induces release of the chemoattractant LTB4, thereby recruiting neutrophils in a positive feedback loop. IgA autoantibodies of patients with autoimmune blistering skin diseases therefore induce massive recruitment of neutrophils, resulting in severe tissue damage. To interfere with neutrophil mobilization and reduce disease morbidity, we developed a panel of specific peptides mimicking either IgA or FcαRI sequences. CLIPS technology was used to stabilize three-dimensional structures and to increase peptides' half-life. IgA and FcαRI peptides reduced phagocytosis of IgA-coated beads, as well as IgA-induced ROS production and neutrophil migration in in vitro and ex vivo (human skin) experiments. Since topical application would be the preferential route of administration, Cetomacrogol cream containing an IgA CLIPS peptide was developed. In the presence of a skin permeation enhancer, peptides in this cream were shown to penetrate the skin, while not diffusing systemically. Finally, epitope mapping was used to discover sequences important for binding between IgA and FcαRI. In conclusion, a cream containing IgA or FcαRI peptide mimetics, which block IgA-induced neutrophil activation and migration in the skin may have therapeutic potential for patients with IgA-mediated blistering skin diseases.


Asunto(s)
Antígenos CD/química , Autoanticuerpos/química , Inmunoglobulina A/química , Activación Neutrófila/efectos de los fármacos , Peptidomiméticos/inmunología , Peptidomiméticos/metabolismo , Receptores Fc/química , Administración Tópica , Antígenos CD/inmunología , Antígenos CD/metabolismo , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Enfermedades Autoinmunes/terapia , Cetomacrogol/administración & dosificación , Cetomacrogol/química , Mapeo Epitopo , Semivida , Humanos , Enfermedades del Sistema Inmune/inmunología , Inmunoglobulina A/inmunología , Inmunoglobulina A/metabolismo , Trastornos Leucocíticos/inmunología , Leucotrieno B4/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Biblioteca de Péptidos , Peptidomiméticos/química , Fagocitosis , Unión Proteica , Especies Reactivas de Oxígeno/metabolismo , Receptores Fc/inmunología , Receptores Fc/metabolismo , Piel/efectos de los fármacos , Piel/inmunología , Piel/patología , Absorción Cutánea , Enfermedades de la Piel/inmunología , Enfermedades de la Piel/terapia
7.
Immunol Rev ; 268(1): 311-27, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26497530

RESUMEN

The individual role of pathogen-binding Toll-like receptors (TLRs) and antibody-binding Fc receptors (FcRs) during pathogenic infections has been studied extensively. However, combined activation of these different receptor classes has received little attention, even though they are triggered simultaneously when immune cells bind antibody-opsonized pathogens. In the last few years, it has become evident that joined activation of TLRs and FcRs substantially tailors inflammatory immune responses, which is an efficient and controlled mechanism of the host to act upon invading pathogens. In this review, we discuss the mechanisms of cross-talk between different TLRs and FcRs and the resulting inflammatory immune responses. Furthermore, we propose how chronic activation via this cross-talk might be detrimental in inflammatory (auto) immune diseases. We conclude with the potential exploitation of the interplay between TLRs and FcRs for monoclonal antibody therapy to target tumors. Future interests in this field of research include establishing a more detailed and mechanistic understanding of the mode of action of TLR and FcR cross-talk and exploration of its physiological importance in health and disease. This may furthermore open up novel therapeutic options for intervention in inflammatory diseases or cancer.


Asunto(s)
Interacciones Huésped-Patógeno , Inmunidad , Inmunoglobulinas/inmunología , Inmunoglobulinas/metabolismo , Receptores Fc/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Humanos , Infecciones/inmunología , Infecciones/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/terapia , Unión Proteica , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal/efectos de los fármacos , Receptores Toll-Like/antagonistas & inhibidores
8.
J Immunol ; 194(4): 1856-66, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25582855

RESUMEN

During secondary immune responses, Ab-opsonized bacteria are efficiently taken up via FcRs by dendritic cells. We now demonstrate that this process induces cross-talk between FcRs and TLRs, which results in synergistic release of several inflammatory cytokines, as well as altered lipid metabolite profiles. This altered inflammatory profile redirects Th1 polarization toward Th17 cell responses. Interestingly, GM-CSF-producing Th cells were synergistically evoked as well, which suggests the onset of polyfunctional Th17 cells. Synergistic cytokine release was dependent on activation via MyD88 and ITAM signaling pathways through TLRs and FcRs, respectively. Cytokine regulation occurred via transcription-dependent mechanisms for TNF-α and IL-23 and posttranscriptional mechanisms for caspase-1-dependent release of IL-1ß. Furthermore, cross-talk between TLRs and FcRs was not restricted to dendritic cells. In conclusion, our results support that bacteria alone initiate fundamentally different immune responses compared with Ab-opsonized bacteria through the combined action of two classes of receptors and, ultimately, may refine new therapies for inflammatory diseases.


Asunto(s)
Células Dendríticas/inmunología , Receptor Cross-Talk/inmunología , Receptores Fc/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Receptores Toll-Like/inmunología , Anticuerpos Antibacterianos/inmunología , Western Blotting , Diferenciación Celular/inmunología , Separación Celular , Infecciones por Enterobacteriaceae/inmunología , Ensayo de Inmunoadsorción Enzimática , Escherichia coli/inmunología , Citometría de Flujo , Humanos , Memoria Inmunológica/inmunología , Inflamación/inmunología , Activación de Linfocitos/inmunología , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/inmunología
9.
Curr Top Microbiol Immunol ; 382: 373-92, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25116109

RESUMEN

Targeted therapies like treatment with monoclonal antibodies (mAbs) have entered the arsenal of modern anticancer drugs. mAbs combine specificity with multiple effector functions that can lead to reduction of tumour burden. Direct mechanisms of action, including induction of apoptosis or growth inhibition, depend on the biology of the target antigen. Fc tails of mAbs have furthermore the potential to initiate complement-dependent lysis as well as immune effector cell-mediated tumour cell killing via binding to Fc receptors. Natural killer cells can induce apoptosis via antibody-dependent cellular cytotoxicity (ADCC), whereas macrophages are able to phagocytose mAb-opsonized tumour cells (antibody-dependent cellular phagocytosis; ADCP). Finally, neutrophils can induce non-apoptotic tumour cell death, especially in the presence of immunoglobulin A (IgA) antitumour mAbs. In spite of promising clinical successes in some malignancies, improvement of mAb immunotherapy is required to achieve overall complete remission in cancer patients. New strategies to enhance Fc receptor-mediated mechanisms of action or to overcome the immunosuppressive microenvironment of the tumour in mAb therapy of cancer are therefore currently being explored and will be addressed in this chapter.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias/tratamiento farmacológico , Receptores Fc/fisiología , Animales , Humanos , Neoplasias/inmunología
10.
J Immunol ; 192(5): 2374-83, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24493821

RESUMEN

Polymorphonuclear cells (neutrophils) are the first cells that arrive at sites of infections. According to the current dogma, they are involved in eliminating bacteria, after which they die through apoptosis. We now demonstrate that enhanced IgA-induced phagocytosis of bacteria or beads by neutrophils led to increased cell death. Nuclear changes and positivity for the general cell death marker 7-aminoactinomycin D were observed, but the absence of annexin V membrane staining supported that neutrophils did not die via apoptosis, in contrast to neutrophils that had not phagocytosed bacteria. Moreover, increased release of neutrophil extracellular traps (NETs) was observed, which was most likely due to augmented production of reactive oxygen species after uptake of IgA-opsonized particles. Blocking the IgA Fc receptor FcαRI abrogated phagocytosis and NET formation. Thus, FcαRI triggering on neutrophils resulted in a rapid form of cell death that is referred to as NETosis, as it is accompanied by the release of NETs. As such, IgA may play a prominent role in mucosal inflammatory responses, where it is the most prominent Ab, because it enhanced both phagocytosis of bacteria and formation of NETs, which are effective mechanisms that neutrophils employ to eliminate pathogens.


Asunto(s)
Bacterias/inmunología , Inmunidad Mucosa/fisiología , Inmunoglobulina A/inmunología , Neutrófilos/inmunología , Fagocitosis/inmunología , Anexina A5/inmunología , Antígenos CD/inmunología , Muerte Celular/efectos de los fármacos , Muerte Celular/inmunología , Dactinomicina/análogos & derivados , Dactinomicina/farmacología , Femenino , Colorantes Fluorescentes/farmacología , Humanos , Inflamación/inmunología , Masculino , Fagocitosis/efectos de los fármacos , Receptores Fc/inmunología
11.
Semin Cancer Biol ; 23(3): 190-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23287459

RESUMEN

The use of monoclonal antibodies (mAbs) has become a mainstream strategy for the treatment of cancer. After binding to the target antigen, they mediate a plethora of effector functions for elimination of tumour cells. These range from direct effects like induction of apoptosis or growth inhibition of tumour cells to activation of immune system components, including the initiation of complement-dependent lysis or recruitment of immune effector cells. Neutrophils are generally not considered as potential effector cells. However, the most abundant population of circulating white blood cells consists of neutrophils, which express Fc receptors for both immunoglobulin (Ig) G and IgA. In the presence of mAbs that are directed against tumour cells, they execute potent cytotoxic functions. Moreover, as they play a role in regulating adaptive immunity, neutrophil activation may lead to the generation of anti-tumour immune responses as well. This article addresses the possibility of exploiting neutrophils for mAb-based immunotherapy of cancer.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Neutrófilos/inmunología , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Citotoxicidad Inmunológica , Humanos
12.
J Immunol ; 189(4): 1594-601, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22802416

RESUMEN

IgA represents the most prominent Ab class at mucosal surfaces and the second most prevalent Ab in human blood after IgG. We recently demonstrated that cross-linking of the granulocyte IgA FcR (FcαRI) by IgA induces a chemotactic-driven positive-feedback migration loop, hereby amplifying recruitment of granulocytes to IgA deposits. Therefore, we postulated that aberrant IgA-Ag complexes, which can be found in tissues in IgA-mediated diseases, are responsible for tissue damage by inducing continuous granulocyte migration and activation. Using an IgA-dependent skin-blistering disease as a model system, we demonstrated colocalization of FcαRI-positive granulocyte infiltrates with IgA in cryosections of lesional skin of patients suffering from this disease. Furthermore, we showed granulocyte migration to IgA deposits injected in human skin explants and in murine skin of FcαRI transgenic mice in vivo. Importantly, ex vivo migration and tissue damage were inhibited by blocking FcαRI, indicating that these events are dependent on the interaction of IgA autoantibodies with FcαRI. Thus, interrupting the granulocyte migration loop by blocking FcαRI reduces tissue damage in diseases with aberrant IgA-immune complexes. As such, our results may lead to development of new therapies for IgA-mediated chronic inflammatory diseases, hereby decreasing severe morbidity and improving quality of life for these patients.


Asunto(s)
Autoanticuerpos/inmunología , Enfermedades Autoinmunes/inmunología , Granulocitos/inmunología , Inmunoglobulina A/inmunología , Receptores de IgG/inmunología , Animales , Autoanticuerpos/metabolismo , Enfermedades Autoinmunes/metabolismo , Quimiotaxis de Leucocito/inmunología , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Granulocitos/metabolismo , Humanos , Inmunoglobulina A/metabolismo , Inmunohistoquímica , Ratones , Ratones Transgénicos , Receptores de IgG/metabolismo , Enfermedades Cutáneas Vesiculoampollosas/inmunología , Enfermedades Cutáneas Vesiculoampollosas/metabolismo
13.
Eur J Immunol ; 42(7): 1815-21, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22535639

RESUMEN

Neutrophils potently kill tumour cells in the presence of anti-tumour antibodies in vitro. However, for in vivo targeting, the neutrophils need to extravasate from the circulation by passing through endothelial barriers. To study neutrophil migration in the presence of endothelial cells in vitro, we established a three-dimensional collagen culture in which SK-BR-3 tumour colonies were grown in the presence or absence of an endothelial barrier. We demonstrated that - in contrast to targeting FcγR on neutrophils with mAbs - targeting the immunoglobulin A Fc receptor (FcαRI) instead triggered neutrophil migration and degranulation leading to tumour destruction, which coincided with release of the pro-inflammatory cytokines interleukin (IL)-1ß and tumour necrosis factor (TNF)-α. Interestingly, neutrophil migration was enhanced in the presence of endothelial cells, which coincided with production of significant levels of the neutrophil chemokine IL-8. This supports the idea that stimulation of neutrophil FcαRI, but not FcγR, initiates cross-talk between neutrophils and endothelial cells, leading to enhanced neutrophil migration towards tumour colonies and subsequent tumour killing.


Asunto(s)
Antígenos CD/inmunología , Neoplasias de la Mama/inmunología , Quimiotaxis de Leucocito/inmunología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Neutrófilos/inmunología , Receptores Fc/inmunología , Neoplasias de la Mama/patología , Comunicación Celular/inmunología , Línea Celular Tumoral , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Inmunidad Innata/inmunología , Inmunohistoquímica , Interleucina-1beta/análisis , Interleucina-1beta/inmunología , Neutrófilos/citología , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/inmunología
14.
J Immunol ; 187(2): 726-32, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21653835

RESUMEN

Neutrophils are the most abundant circulating FcR-expressing WBCs with potent cytotoxic ability. Currently, they are recognized as promising effector cells for Ab-mediated immunotherapy of cancer, because their capacity to kill tumor cells is greatly enhanced by tumor Ag-specific mAbs. The FcαRI represents the most potent FcR on neutrophils for induction of Ab-mediated tumor cell killing. However, the mechanisms of cell death that are induced are poorly understood. Because these mechanisms can be used for modulation of anticancer treatment, we investigated the tumor cell death induced by neutrophil-mediated Ab-dependent killing via FcαRI. Human mammary carcinoma cells were efficiently killed when incubated with human neutrophils and tumor-specific FcαRI bispecific or IgA Abs. Interestingly, we observed characteristics of autophagy such as autophagic structures by electron microscopy and LC3B(+) autophagosomes in different human epithelial carcinoma cells, which resulted in tumor cell death. To a lesser extent, necrotic features, such as cellular membrane breakdown and spillage of intracellular content, were found. By contrast, apoptotic features including fragmented nuclei, Annexin V-positivity, and presence of cleaved caspase-3 were not observed. These findings indicate that neutrophils mainly facilitate autophagy to induce tumor cell death rather than the more commonly recognized apoptotic cell death mechanisms induced by NK cells or cytotoxic T cells. This knowledge not only reveals the type of tumor cell death induced in neutrophil-mediated, Ab-dependent cellular cytotoxicity, but importantly opens up additional perspectives for modulation of anticancer therapy in, for example, apoptosis-resistant tumor cells.


Asunto(s)
Anticuerpos Biespecíficos/fisiología , Antígenos CD/fisiología , Autofagia/inmunología , Marcación de Gen/métodos , Neutrófilos/inmunología , Neutrófilos/patología , Receptores Fc/fisiología , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/metabolismo , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antígenos CD/genética , Antígenos CD/metabolismo , Apoptosis/inmunología , Línea Celular Tumoral , Células Cultivadas , Técnicas de Cocultivo , Humanos , Inmunoglobulina A/fisiología , Neutrófilos/metabolismo , Receptores Fc/genética , Receptores Fc/metabolismo
15.
MAbs ; 3(4): 352-61, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21691145

RESUMEN

Although immunoglobulin (Ig) A is commonly recognized as the most prevalent antibody subclass at mucosal sites with an important role in mucosal defense, its potential as a therapeutic monoclonal antibody is less well known. However, IgA has multifaceted anti-, non-, and pro-inflammatory functions that can be exploited for different immunotherapeutical strategies, which will be the focus of this review.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inmunoglobulina A/uso terapéutico , Infecciones/terapia , Neoplasias/terapia , Receptores de IgE/inmunología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Humanos , Inmunidad Mucosa , Inmunoglobulina A/química , Inmunoglobulina A/inmunología , Inmunoterapia/métodos , Modelos Moleculares
16.
Eur J Immunol ; 40(7): 2035-40, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20411563

RESUMEN

The receptor for IgA, FcalphaRI or CD89, is expressed on myeloid cells and can trigger phagocytosis, tumor cell lysis, and release of inflammatory mediators. These functions critically depend on the associated FcR gamma-chain; however, some biological functions, like receptor internalization, are solely mediated by FcalphaRI alpha-chain. Little is known as to how FcalphaRI regulates these processes and the FcalphaRI intracellular domain does not contain recognized signalling motifs. We searched for associating proteins and identified c-Jun activating binding protein 1 (JAB1) as a binding partner specifically for FcalphaRI. We found increased FcalphaRI surface expression after ectopic expression of JAB1 as well as diminished protein levels of total FcR gamma-chain levels after JAB1 knock-down. These data functionally link JAB1 with controlling protein expression levels of FcalphaRI-FcR gamma-chain protein complex.


Asunto(s)
Antígenos CD/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptido Hidrolasas/metabolismo , Receptores Fc/metabolismo , Receptores de IgG/biosíntesis , Secuencias de Aminoácidos/genética , Animales , Antígenos CD/genética , Complejo del Señalosoma COP9 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Ratones , Células 3T3 NIH , Péptido Hidrolasas/genética , Péptido Hidrolasas/inmunología , Unión Proteica/genética , Dominios y Motivos de Interacción de Proteínas/genética , Estabilidad Proteica , ARN Interferente Pequeño/genética , Receptores Fc/genética , Receptores de IgG/genética , Activación Transcripcional/genética , Transgenes/genética , Células U937
17.
Cancer Res ; 70(8): 3209-17, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20354182

RESUMEN

Antibody-Fc receptor (FcR) interactions play an important role in the mechanism of action of most therapeutic antibodies against cancer. Effector cell activation through FcR triggering may induce tumor cell killing via antibody-dependent cellular cytotoxicity (ADCC). Reciprocally, FcR cross-linking of antibody may lead to the induction of apoptotic signaling in tumor cells. The relative importance of these bisecting pathways to in vivo antibody activity is unknown. To unravel these roles, we developed a novel mouse model with normal FcR expression but in which FcR signaling was inactivated by mutation of the associated gamma-chain. Transgenic mice showed similar immune complex binding compared with wild-type mice. In contrast, ADCC of cells expressing frequently used cancer targets, such as CD20, epidermal growth factor receptor, Her2, and gp75, was abrogated. Using the therapeutic CD20 antibodies ofatumumab and rituximab, we show that FcR cross-linking of antibody-antigen immune complexes in the absence of gamma-chain signaling is insufficient for their therapeutic activity in vivo. ADCC therefore represents an essential mechanism of action for immunotherapy of lymphoid tumors.


Asunto(s)
Anticuerpos/química , Antígenos CD20/química , Receptores Fc/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Anticuerpos Monoclonales de Origen Murino , Apoptosis , Receptores ErbB/metabolismo , Humanos , Inmunoterapia/métodos , Melanoma Experimental , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Mutación , Rituximab , Homología de Secuencia de Aminoácido , Transducción de Señal
18.
Gastroenterology ; 137(6): 2018-29.e1-3, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19555692

RESUMEN

BACKGROUND & AIMS: Exacerbations of ulcerative colitis (UC) are dominated by massive neutrophil influx in the lamina propria with concomitant mucosal ulceration. The prevalent antibody in this area is immunoglobulin A (IgA). Interestingly, the IgA Fc receptor (Fc(alpha)RI) potently activates neutrophils. As such, we investigated whether IgA-Fc(alpha)RI interaction contributes to tissue damage in UC. METHODS: Response of neutrophils to bovine serum albumin-, IgG-, or IgA-coated beads and Escherichia coli was investigated with 3-dimensional culture systems, real-time video microscopy, and (fluorescence) microscopy. In vivo studies were performed using human Fc(alpha)RI transgenic mice or nontransgenic littermates. Microscopic slides of UC patients were stained for IgA, Fc(alpha)RI, and neutrophils. RESULTS: In vitro and in vivo cross-linking of Fc(alpha)RI on neutrophils by serum IgA or uptake of IgA-coated E coli led to neutrophil migration. The responsible chemotactic factor was identified as leukotriene B4. Moreover, dimeric IgA (dIgA), which is produced in the lamina propria, but neither secretory IgA nor IgG, was equally capable of inducing neutrophil recruitment. We furthermore showed that Fc(alpha)RI(+)-neutrophils in the colon of UC patients had phagocytosed IgA-antigen complexes. CONCLUSIONS: Neutrophils are the first cells that arrive at inflammatory sites once pathogens have crossed the epithelial barrier. Fc(alpha)RI-dIgA interactions therefore may constitute an essential activation step to recruit more neutrophils, hereby eradicating impending infections. However, excessive IgA-antigen complexes can sustain a perpetuating inflammatory loop in UC, hereby seriously aggravating morbidity. Novel therapeutic strategies that block dIgA-Fc(alpha)RI interactions, and therefore diminish neutrophil migration and activation, may dampen the uncontrolled inflammatory processes in these patients.


Asunto(s)
Antígenos CD/metabolismo , Quimiotaxis de Leucocito , Colitis Ulcerosa/inmunología , Inmunoglobulina A/metabolismo , Leucotrieno B4/metabolismo , Activación Neutrófila , Infiltración Neutrófila , Neutrófilos/inmunología , Receptores Fc/metabolismo , Animales , Antígenos CD/genética , Células Cultivadas , Escherichia coli/inmunología , Humanos , Inmunoglobulina G/metabolismo , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Microscopía por Video , Fagocitosis , Receptores Fc/genética , Albúmina Sérica Bovina/metabolismo , Factores de Tiempo
19.
J Immunol ; 181(6): 4080-8, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18768864

RESUMEN

To achieve a correct cellular immune response toward pathogens, interaction between FcR and their ligands must be regulated. The Fc receptor for IgA, FcalphaRI, is pivotal for the inflammatory responses against IgA-opsonized pathogens. Cytokine-induced inside-out signaling through the intracellular FcalphaRI tail is important for FcalphaRI-IgA binding. However, the underlying molecular mechanism governing this process is not well understood. In this study, we report that PP2A can act as a molecular switch in FcalphaRI activation. PP2A binds to the intracellular tail of FcalphaRI and, upon cytokine stimulation, PP2A becomes activated. Subsequently, FcalphaRI is dephosphorylated on intracellular Serine 263, which we could link to receptor activation. PP2A inhibition, in contrast, decreased FcalphaRI ligand binding capacity in transfected cells but also in eosinophils and monocytes. Interestingly, PP2A activity was found crucial for IgA-mediated binding and phagocytosis of Neisseria meningitidis. The present findings demonstrate PP2A involvement as a molecular mechanism for FcalphaRI ligand binding regulation, a key step in initiating an immune response.


Asunto(s)
Antígenos CD/metabolismo , Proteína Fosfatasa 2/fisiología , Receptores Fc/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Células Cultivadas , Humanos , Líquido Intracelular/química , Líquido Intracelular/inmunología , Líquido Intracelular/metabolismo , Ratones , Datos de Secuencia Molecular , Monocitos/enzimología , Monocitos/inmunología , Monocitos/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/fisiología , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/metabolismo , Estructura Terciaria de Proteína , Transducción de Señal/inmunología , Células U937
20.
Mol Immunol ; 45(7): 2069-75, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18023480

RESUMEN

The C-terminal domain of protein 4.1G was identified to interact with the cytosolic tail of the high affinity IgG receptor, Fc gamma RI, in yeast two-hybrid screens. Proteins of the 4.1 family have previously been found to mediate receptor/cytoskeleton interactions. In the study presented here, we show an alternatively spliced 4.1G product to be associated with increased Fc gamma RI binding in yeast two-hybrid assays, and to be selectively enriched in most immune cells at the transcript level. In addition, a detailed analysis of the 4.1G 'docking site' within Fc gamma RI is provided by examining Fc gamma RI-CY-truncated and alanine-substituted mutants. These pointed to an Fc gamma RI membrane-proximal core motif of HxxBxxxBB (H represents hydrophobic residues, B basic residues and x represents any residue), followed by hydrophobic and (potentially) negatively charged residues to be central for interaction with protein 4.1G.


Asunto(s)
Secuencias de Aminoácidos , Citoplasma/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de IgG/química , Receptores de IgG/metabolismo , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Línea Celular , Membrana Celular/metabolismo , Humanos , Inmunoprecipitación , Ratones , Datos de Secuencia Molecular , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...