Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 13: 984678, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36177051

RESUMO

Progress in neuroimmunology established that the nervous and the immune systems are two functionally related physiological systems. Unique sensory and immune receptors enable them to control interactions of the organism with the inner and the outer worlds. Both systems undergo an experience-driven selection process during their ontogeny. They share the same mediators/neurotransmitters and use synapses for intercellular communication. They keep a memory of previous experiences. Immune cells can affect nervous cells, nervous cells can affect immune cells, and they regulate each other. I however argue that the two systems differ by three major points: 1) Unlike the nervous system, the immune system has a loose anatomical structure, in which molecular and cellular events mostly occur at random; 2) The immune system can respond to molecules of the living world whereas the nervous system can respond to phenomena of the physical world; 3) Responses of the immune system act both on the organism and on the stimulus that triggered the response, whereas responses of the nervous system act on the organism only. The nervous and the immune systems therefore appear as two complementary systems of relations that closely work together, and whose reactivities are well-suited to deal with physical and biological stimuli, respectively. Its ability both to adapt the organism to the living world and to adapt the living world to the organism endows the immune system with powerful adaptive properties that enable the organism to live in peace with itself and with other living beings, whether pathogens or commensals.


Assuntos
Sistema Imunitário , Neuroimunomodulação , Comunicação Celular , Sistema Imunitário/fisiologia , Sistema Nervoso , Neuroimunomodulação/fisiologia , Neurotransmissores/fisiologia
2.
Rev Prat ; 71(4): 374-379, 2021 Apr.
Artigo em Francês | MEDLINE | ID: mdl-34161001

RESUMO

"Immune checkpoint inhibitors in oncology.Cancer immunotherapy has revolutionized oncology. It results from a long history that went along with that of immunology, with its hesitations and failures, and which led to two complementary approaches. One consists in manufacturing cancer-specific monoclonal antibodies or cytotoxic T cells that are injected into patients. The other consists in relieving control mechanisms that prevent normally immune effectors from being pathogenic, and thus, freeing anti-cancer immunity from its constraints. These immune checkpoint inhibitors proved markedly efficient, especially in cancers with a poor prognosis. They opened an unprecedented increase in novel immunotherapeutic approaches and clinical trials."


"Les inhibiteurs du contrôle immunitaire en oncologie. L'immunothérapie des cancers est en train de révolutionner l'oncologie. Elle est le fruit d'une longue histoire qui a accompagné celle de l'immunologie, avec ses hésitations et ses échecs, et qui a abouti à deux approches complémentaires. L'une consiste à fabriquer au laboratoire des anticorps monoclonaux ou des cellules T cytotoxiques spécifiques du cancer des patients et à les leur administrer. L'autre consiste à lever les mécanismes de contrôle qui empêchent normalement les effecteurs immunitaires d'être pathogènes, et de libérer ainsi l'immunité anticancer. Ces inhibiteurs de contrôle immunitaire se sont révélés d'une grande efficacité, notamment sur des cancers de mauvais pronostic. Ils ont été à l'origine d'une recherche sans précédent de nouvelles approches d'immunothérapie et à une multiplication des essais cliniques."


Assuntos
Inibidores de Checkpoint Imunológico , Neoplasias , Anticorpos Monoclonais/uso terapêutico , Humanos , Fatores Imunológicos/uso terapêutico , Imunoterapia , Neoplasias/tratamento farmacológico
3.
Nature ; 576(7785): E3, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31745371

RESUMO

An Amendment to this paper has been published and can be accessed via a link at the top of the paper.

4.
Nature ; 574(7776): 45-56, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31578484

RESUMO

New therapies that promote antitumour immunity have been recently developed. Most of these immunomodulatory approaches have focused on enhancing T-cell responses, either by targeting inhibitory pathways with immune checkpoint inhibitors, or by targeting activating pathways, as with chimeric antigen receptor T cells or bispecific antibodies. Although these therapies have led to unprecedented successes, only a minority of patients with cancer benefit from these treatments, highlighting the need to identify new cells and molecules that could be exploited in the next generation of immunotherapy. Given the crucial role of innate immune responses in immunity, harnessing these responses opens up new possibilities for long-lasting, multilayered tumour control.


Assuntos
Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Imunoterapia/métodos , Imunoterapia/tendências , Neoplasias/imunologia , Neoplasias/terapia , Animais , Humanos , Neoplasias/tratamento farmacológico , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
5.
J Allergy Clin Immunol ; 142(5): 1548-1557, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29366701

RESUMO

BACKGROUND: Basophil activation contributes to inflammatory reactions, especially in allergy. It is controlled, both positively and negatively, by several mechanisms. High-affinity IgE receptors (FcεRI) generate a mixture of activation and inhibition signals when aggregated, the ratio of which depends on the concentration of allergen recognized by receptor-bound IgE. Low-affinity IgG receptors (FcγRIIA/B) generate inhibition signals when coengaged with FcεRI by allergen-antibody immune complexes. Commensal and probiotic bacteria, such as Lactobacillus paracasei, generate inhibition signals through still unclear mechanisms. OBJECTIVE: We sought to investigate whether mechanisms that control, both positively and negatively, basophil activation, which were unraveled and studied in basophils from healthy donors, are functional in allergic patients. METHODS: FcεRI and FcγRIIA/B expression, FcεRI-dependent activation, FcεRI-dependent inhibition, and FcγRIIB-dependent inhibition were examined in blood basophils incubated overnight with or without L paracasei and challenged under 10 experimental conditions. Basophils from healthy donors were compared with basophils from patients who consulted an allergology outpatient clinic over a period of 3 months with respiratory allergy, anaphylaxis antecedents, chronic urticaria, and/or atopic dermatitis. RESULTS: Patients' basophils expressed neither more FcεRI nor less FcγRIIB than basophils from healthy donors. They were neither hyperreactive to positive regulation nor hyporeactive to negative regulation, irrespective of the receptors or mechanisms involved and the allergic manifestations of the patients. CONCLUSION: Regulatory mechanisms that control basophil activation are fully functional in allergic patients. Intrinsic defects in these mechanisms do not explain allergic manifestations. Based on these mechanisms, immune checkpoint modifiers can be developed as novel therapeutic tools for allergy.


Assuntos
Basófilos/imunologia , Hipersensibilidade/imunologia , Adolescente , Adulto , Idoso , Feminino , Humanos , Lacticaseibacillus paracasei/imunologia , Masculino , Pessoa de Meia-Idade , Receptores de IgE/imunologia , Receptores de IgG/imunologia , Adulto Jovem
6.
Sci Signal ; 9(459): c24, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-27999172

RESUMO

This Podcast features an interview with Marc Daëron, author of a Research Article that appears in the 20 December 2016 issue of Science Signaling, about a mechanism by which an Fc receptor can inhibit signaling by other receptors without aggregating with those other receptors. Engagement of Fc receptors on basophils and mast cells can either activate these cells, which promotes autoimmune and allergic inflammation, or prevent these cells from being activated. Whether these cells are activated depends upon which Fc receptors are present in clusters, because some Fc receptors can inhibit signaling by other Fc receptors that are present in the same signalosome, a phenomenon known as cis-inhibition. Malbec et al. identified a mechanism whereby inhibitory Fc receptors limit signaling by activating Fc receptors without being present in the same signalosome. This mechanism of trans-inhibition also allowed inhibitory Fc receptors to limit signaling by growth factor receptors in mast cells and oncogene-induced proliferation in mastocytoma cells.Listen to Podcast.


Assuntos
Basófilos/metabolismo , Proliferação de Células , Mastócitos/metabolismo , Mastocitoma/metabolismo , Proteínas de Neoplasias/metabolismo , Oncogenes , Receptores Fc/metabolismo , Animais , Basófilos/patologia , Humanos , Mastócitos/patologia , Mastocitoma/genética , Mastocitoma/patologia , Proteínas de Neoplasias/genética , Receptores Fc/genética
7.
Sci Signal ; 9(459): ra126, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-27999175

RESUMO

Allergic and autoimmune inflammation are associated with the activation of mast cells and basophils by antibodies against allergens or auto-antigens, respectively. Both cell types express several receptors for the Fc portion of antibodies, the engagement of which by antigen-antibody complexes controls their responses. When aggregated on the plasma membrane, high-affinity immunoglobulin E (IgE) receptors (FcεRI) and low-affinity IgG receptors (FcγRIIIA in mice, FcγRIIA in humans) induce these cells to release and secrete proinflammatory mediators, chemokines, and cytokines that account for clinical symptoms. When coaggregated with activating receptors on the same cells, other low-affinity IgG receptors (FcγRIIB in both species) inhibit mast cell and basophil activation. We found that FcγRIIB inhibited not only signals triggered by activating receptors with which they were coengaged (cis-inhibition), but also signals triggered by receptors engaged independently (trans-inhibition). Trans-inhibition acted upon the FcεRI-dependent activation of mouse mast cells, mouse basophils, and human basophils, and upon growth factor receptor (Kit)-dependent normal mouse mast cell proliferation, as well as the constitutive in vitro proliferation and the in vivo growth of oncogene (v-Abl)-transformed mastocytoma cells. Trans-inhibition was induced by receptors, whether inhibitory (FcγRIIB) or activating (FcεRI), which recruited the lipid phosphatase SHIP1. By hydrolyzing PI(3,4,5)P3, SHIP1 induced a global unresponsiveness that affected biological responses triggered by receptors that use phosphoinositide 3-kinase to signal. These data suggest that trans-inhibition controls numerous physiological and pathological processes, and that it may be used as a therapeutic tool in inflammation, especially but not exclusively, in allergy and autoimmunity.


Assuntos
Basófilos/imunologia , Proliferação de Células , Receptores de IgE/imunologia , Receptores de IgG/imunologia , Transdução de Sinais/imunologia , Animais , Humanos , Mastócitos , Camundongos , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/imunologia , Fosfatos de Fosfatidilinositol/genética , Fosfatos de Fosfatidilinositol/imunologia , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases/genética , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases/imunologia , Receptores de IgG/genética
8.
Immun Inflamm Dis ; 4(3): 289-99, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27621812

RESUMO

INTRODUCTION: The microbiota controls a variety of biological functions, including immunity, and alterations of the microbiota in early life are associated with a higher risk of developing allergies later in life. Several probiotic bacteria, and particularly lactic acid bacteria, were described to reduce both the induction of allergic responses and allergic manifestations. Although specific probiotic strains were used in these studies, their protective effects on allergic responses also might be common for all lactobacilli. METHODS: To determine whether allergic effector cells inhibition is a common feature of lactobacilli or whether it varies among lactobacilli strains, we compared the ability of 40 strains of the same Lactobacillus paracasei species to inhibit IgE-dependent mouse mast cell and human basophil activation. RESULTS: We uncovered a marked heterogeneity in the inhibitory properties of the 40 Lactobacillus strains tested. These segregated into three to four clusters depending on the intensity of inhibition. Some strains inhibited both mouse mast cell and human basophil activation, others strains inhibited only one cell type and another group induced no inhibition of activation for either cell type. CONCLUSIONS: Individual Lactobacillus strains of the same species differentially inhibit IgE-dependent activation of mouse mast cells and human basophils, two cell types that are critical in the onset of allergic manifestations. Although we failed to identify specific bacterial genes associated with inhibition by gene-trait matching analysis, our findings demonstrate the complexity of the interactions between the microbiota and the host. These results suggest that some L. paracasei strains might be more beneficial in allergies than others strains and provide the bases for a rational screening of lactic acid bacteria strains as next-generation probiotics in the field of allergy.

9.
Curr Opin Immunol ; 38: 101-8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26745401

RESUMO

Mast cells and basophils are mostly known as the initiators of IgE-dependent allergic reactions. They, however, contribute to innate immunity against pathogens and venoms. Like other myeloid cells, they also express receptors for the Fc portion of IgG antibodies. These include activating receptors and inhibitory receptors. Because IgG antibodies are produced in exceedingly higher amounts than IgE antibodies, IgG receptors are co-engaged with IgE receptors under physiological conditions. Mast cells and basophils are examples of the many innate myeloid cells whose effector functions are used and finely tuned by antibodies. They can be thus enrolled in a variety of adaptive immune responses, their activation can be regulated, positively and negatively and their biological responses can be modulated qualitatively by antibodies.


Assuntos
Imunidade Adaptativa , Hipersensibilidade/imunologia , Imunidade Inata , Neoplasias/imunologia , Receptores de IgE/imunologia , Receptores de IgG/imunologia , Infecções por Strongylida/imunologia , Animais , Apresentação de Antígeno , Basófilos/citologia , Basófilos/imunologia , Células Dendríticas/imunologia , Células Dendríticas/patologia , Regulação da Expressão Gênica/imunologia , Humanos , Hipersensibilidade/genética , Hipersensibilidade/patologia , Imunoglobulina E/genética , Imunoglobulina G/genética , Mastócitos/citologia , Mastócitos/imunologia , Neoplasias/genética , Neoplasias/patologia , Receptores de IgE/genética , Receptores de IgG/genética , Infecções por Strongylida/genética , Infecções por Strongylida/patologia , Linfócitos T/imunologia , Linfócitos T/patologia
11.
J Clin Invest ; 124(10): 4577-89, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25180604

RESUMO

Controlling the overwhelming inflammatory reaction associated with polymicrobial sepsis remains a prevalent clinical challenge with few treatment options. In septic peritonitis, blood neutrophils and monocytes are rapidly recruited into the peritoneal cavity to control infection, but the role of resident sentinel cells during the early phase of infection is less clear. In particular, the influence of mast cells on other tissue-resident cells remains poorly understood. Here, we developed a mouse model that allows both visualization and conditional ablation of mast cells and basophils to investigate the role of mast cells in severe septic peritonitis. Specific depletion of mast cells led to increased survival rates in mice with acute sepsis. Furthermore, we determined that mast cells impair the phagocytic action of resident macrophages, thereby allowing local and systemic bacterial proliferation. Mast cells did not influence local recruitment of neutrophils and monocytes or the release of inflammatory cytokines. Phagocytosis inhibition by mast cells involved their ability to release prestored IL-4 within 15 minutes after bacterial encounter, and treatment with an IL-4-neutralizing antibody prevented this inhibitory effect and improved survival of septic mice. Our study uncovers a local crosstalk between mast cells and macrophages during the early phase of sepsis development that aggravates the outcome of severe bacterial infection.


Assuntos
Inflamação/imunologia , Macrófagos Peritoneais/citologia , Mastócitos/citologia , Fagocitose , Sepse/imunologia , Animais , Infecções Bacterianas , Células da Medula Óssea/citologia , Proliferação de Células , Separação Celular , Citometria de Fluxo , Interleucina-4/metabolismo , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Peritonite/patologia
12.
Curr Top Microbiol Immunol ; 382: 69-93, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25116096

RESUMO

A global and rigorous understanding of the signaling pathways and cross-regulatory processes involved in mast cell activation requires the integration of published information with novel functional datasets into a comprehensive computational model. Based on an exhaustive curation of the existing literature and using the software CellDesigner, we have built and annotated a comprehensive molecular map for the FcεRI signaling network. This map can be used to visualize and interpret high-throughput expression data. Furthermore, leaning on this map and using the logical modeling software GINsim, we have derived a qualitative dynamical model, which recapitulates the most salient features of mast cell activation. The resulting logical model can be used to explore the dynamical properties of the system and its responses to different stimuli, in normal or mutant conditions.


Assuntos
Simulação por Computador , Mastócitos/fisiologia , Transdução de Sinais/fisiologia , Animais , Humanos , Receptores Fc/fisiologia , Software
13.
Curr Top Microbiol Immunol ; 382: 131-64, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25116099

RESUMO

Most biological activities of antibodies depend on their ability to engage Receptors for the Fc portion of immunoglobulins (FcRs) on a variety of cell types. As FcRs can trigger positive and negative signals, as these signals control several biological activities in individual cells, as FcRs are expressed by many cells of hematopoietic origin, mostly of the myeloid lineage, as these cells express various combinations of FcRs, and as FcR-expressing cells have different functional repertoires, antibodies can exert a wide spectrum of biological activities. Like B and T Cell Receptors (BCRs and TCRs), FcRs are bona fide immunoreceptors. Unlike BCRs and TCRs, however, FcRs are immunoreceptors with an adaptive specificity for antigen, with an adaptive affinity for antibodies, with an adaptive structure and with an adaptive signaling. They induce adaptive biological responses that depend on their tissue distribution and on FcR-expressing cells that are selected locally by antibodies. They critically determine health and disease. They are thus exquisitely adaptive therapeutic tools.


Assuntos
Imunidade Adaptativa , Receptores Fc/fisiologia , Animais , Afinidade de Anticorpos , Autoimunidade , Humanos , Hipersensibilidade/imunologia , Infecções/imunologia , Transdução de Sinais
14.
Virol J ; 11: 82, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24885320

RESUMO

BACKGROUND: Public health risks associated to infection by human coronaviruses remain considerable and vaccination is a key option for preventing the resurgence of severe acute respiratory syndrome coronavirus (SARS-CoV). We have previously reported that antibodies elicited by a SARS-CoV vaccine candidate based on recombinant, full-length SARS-CoV Spike-protein trimers, trigger infection of immune cell lines. These observations prompted us to investigate the molecular mechanisms and responses to antibody-mediated infection in human macrophages. METHODS: We have used primary human immune cells to evaluate their susceptibility to infection by SARS-CoV in the presence of anti-Spike antibodies. Fluorescence microscopy and real-time quantitative reverse transcriptase polymerase chain reaction (RT-PCR) were utilized to assess occurrence and consequences of infection. To gain insight into the underlying molecular mechanism, we performed mutational analysis with a series of truncated and chimeric constructs of fragment crystallizable γ receptors (FcγR), which bind antibody-coated pathogens. RESULTS: We show here that anti-Spike immune serum increased infection of human monocyte-derived macrophages by replication-competent SARS-CoV as well as Spike-pseudotyped lentiviral particles (SARS-CoVpp). Macrophages infected with SARS-CoV, however, did not support productive replication of the virus. Purified anti-viral IgGs, but not other soluble factor(s) from heat-inactivated mouse immune serum, were sufficient to enhance infection. Antibody-mediated infection was dependent on signaling-competent members of the human FcγRII family, which were shown to confer susceptibility to otherwise naïve ST486 cells, as binding of immune complexes to cell surface FcγRII was necessary but not sufficient to trigger antibody-dependent enhancement (ADE) of infection. Furthermore, only FcγRII with intact cytoplasmic signaling domains were competent to sustain ADE of SARS-CoVpp infection, thus providing additional information on the role of downstream signaling by FcγRII. CONCLUSIONS: These results demonstrate that human macrophages can be infected by SARS-CoV as a result of IgG-mediated ADE and indicate that this infection route requires signaling pathways activated downstream of binding to FcγRII receptors.


Assuntos
Anticorpos Antivirais/imunologia , Endocitose , Macrófagos/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Células Cultivadas , Humanos , Microscopia de Fluorescência , Reação em Cadeia da Polimerase em Tempo Real
18.
Mol Cell Proteomics ; 12(10): 2874-89, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23820730

RESUMO

We report the first proteomic analysis of the SLP76 interactome in resting and activated primary mouse mast cells. This was made possible by a novel genetic approach used for the first time here. It consists in generating knock-in mice that express signaling molecules bearing a C-terminal tag that has a high affinity for a streptavidin analog. Tagged molecules can be used as molecular baits to affinity-purify the molecular complex in which they are engaged, which can then be studied by mass spectrometry. We examined first SLP76 because, although this cytosolic adapter is critical for both T cell and mast cell activation, its role is well known in T cells but not in mast cells. Tagged SLP76 was expressed in physiological amounts and fully functional in mast cells. We unexpectedly found that SLP76 is exquisitely sensitive to mast cell granular proteases, that Zn(2+)-dependent metalloproteases are especially abundant in mast cells and that they were responsible for SLP76 degradation. Adding a Zn(2+) chelator fully protected SLP76 in mast cell lysates, thereby enabling an efficient affinity-purification of this adapter with its partners. Label-free quantitative mass spectrometry analysis of affinity-purified SLP76 interactomes uncovered both partners already described in T cells and novel partners seen in mast cells only. Noticeably, molecules inducibly recruited in both cell types primarily concur to activation signals, whereas molecules recruited in activated mast cells only are mostly associated with inhibition signals. The transmembrane adapter LAT2, and the serine/threonine kinase with an exchange factor activity Bcr were the most recruited molecules. Biochemical and functional validations established the unexpected finding that Bcr is recruited by SLP76 and positively regulates antigen-induced mast cell activation. Knock-in mice expressing tagged molecules with a normal tissue distribution and expression therefore provide potent novel tools to investigate signalosomes and to uncover novel signaling molecules in mast cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Mastócitos/metabolismo , Fosfoproteínas/metabolismo , Receptores de IgE/metabolismo , Animais , Células da Medula Óssea/citologia , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mapas de Interação de Proteínas , Proteômica , Proteínas Proto-Oncogênicas c-bcr/genética , Proteínas Proto-Oncogênicas c-bcr/metabolismo
19.
Curr Opin Immunol ; 25(3): 313-20, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23684445

RESUMO

Recent progress has begun to reveal the often complex and changing roles of phosphotyrosine and phosphoinositide phosphatases in regulation of immunoreceptor signaling. The resultant confusion has been further increased by discoveries of new players. Here we provide a review of recent progress in defining the roles of these enzymes in immunoreceptor-dependent mast cell, T cell and B cell activation.


Assuntos
Linfócitos B/imunologia , Mastócitos/imunologia , Monoéster Fosfórico Hidrolases/imunologia , Receptores Imunológicos/imunologia , Transdução de Sinais , Linfócitos T/imunologia , Animais , Humanos
20.
Blood ; 121(9): 1563-73, 2013 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-23293080

RESUMO

Receptors for the Fc portion of IgG (FcγRs) are mandatory for the induction of various IgG-dependent models of autoimmunity, inflammation, anaphylaxis, and cancer immunotherapy. A few FcγRs have the ability to bind monomeric IgG: high-affinity mouse mFcγRI, mFcγRIV, and human hFcγRI. All others bind IgG only when aggregated in complexes or bound to cells or surfaces: low-affinity mouse mFcγRIIB and mFcγRIII and human hFcγRIIA/B/C and hFcγRIIIA/B. Although it has been proposed that high-affinity FcγRs are occupied by circulating IgG, multiple roles for mFcγRI and mFcγRIV have been reported in vivo. However, the potential roles of hFcγRI that is expressed on monocytes, macrophages, and neutrophils have not been reported. In the present study, we therefore investigated the role of hFcγRI in antibody-mediated models of disease and therapy by generating hFcγRI-transgenic mice deficient for multiple endogenous FcRs. hFcγRI was sufficient to trigger autoimmune arthritis and thrombocytopenia, immune complex-induced airway inflammation, and active and passive systemic anaphylaxis. We found monocyte/macrophages to be responsible for thrombocytopenia, neutrophils to be responsible for systemic anaphylaxis, and both cell types to be responsible for arthritis induction. Finally, hFcγRI was capable of mediating antibody-induced immunotherapy of metastatic melanoma. Our results unravel novel capabilities of human FcγRI that confirm the role of high-affinity IgG receptors in vivo.


Assuntos
Anafilaxia/genética , Imunoglobulina G/fisiologia , Imunoterapia , Inflamação/genética , Neoplasias/terapia , Receptores de IgG/fisiologia , Anafilaxia/imunologia , Animais , Antineoplásicos/farmacologia , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Humanos , Imunoglobulina G/farmacologia , Imunoterapia/métodos , Inflamação/induzido quimicamente , Inflamação/imunologia , Camundongos , Camundongos Transgênicos , Metástase Neoplásica , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Receptores de IgG/genética , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...