Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 177
Filtrar
1.
Fish Shellfish Immunol ; 151: 109706, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38897310

RESUMEN

The complement component 5a/complement component 5 receptor 1 (C5a/C5aR1) pathway plays a crucial role in the onset and development of inflammation, but relevant studies in fish are lacking. In this study, we successfully characterized the relationship between half-smooth tongue sole (Cynoglossus semilaevis) C5aR1 (CsC5aR1) and bacterial inflammation. First, we showed that the overexpression of CsC5aR1 significantly increased bacterial pathological damage in the liver and intestine, whereas inhibition attenuated the damage. The in vitro experiments suggested that CsC5aR1 was able to positively regulate the phagocytic activity and respiratory burst of tongue sole macrophages. In terms of both transcriptional and translational levels, overexpression/inhibition of CsC5aR1 was followed by a highly consistent up-regulation/decrease of its downstream canonical inflammatory factor interleukin-6 (CsIL-6). Furthermore, we stimulated macrophages by lipopolysaccharide (LPS) and lipoteichoic acid (LTA) and found a broad-spectrum response to bacterial infections by the C5a/C5aR1 complement pathway together with the downstream inflammatory factor CsIL-6. Subsequently, we directly elucidated that CsIL-6 is an indicator of C5a/C5aR1-mediated inflammation at different infection concentrations, different infectious bacteria (Vibrio anguillarum and Mycobacterium marinum), and different detection levels. These results might provide a new inflammation bio-marker for early warning of bacteria-induced hyperinflammation leading to fish mortality and a promising target for the treatment of bacterial inflammation in teleost.


Asunto(s)
Enfermedades de los Peces , Proteínas de Peces , Peces Planos , Interleucina-6 , Receptor de Anafilatoxina C5a , Animales , Peces Planos/inmunología , Peces Planos/genética , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/inmunología , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/microbiología , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Interleucina-6/metabolismo , Vibriosis/veterinaria , Vibriosis/inmunología , Vibrio/fisiología , Inflamación/inmunología , Inflamación/veterinaria , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética
2.
Sci Immunol ; 6(66): eabf2489, 2021 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-34932384

RESUMEN

While serum-circulating complement destroys invading pathogens, intracellularly active complement, termed the "complosome," functions as a vital orchestrator of cell-metabolic events underlying T cell effector responses. Whether intracellular complement is also nonredundant for the activity of myeloid immune cells is currently unknown. Here, we show that monocytes and macrophages constitutively express complement component (C) 5 and generate autocrine C5a via formation of an intracellular C5 convertase. Cholesterol crystal sensing by macrophages induced C5aR1 signaling on mitochondrial membranes, which shifted ATP production via reverse electron chain flux toward reactive oxygen species generation and anaerobic glycolysis to favor IL-1ß production, both at the transcriptional level and processing of pro­IL-1ß. Consequently, atherosclerosis-prone mice lacking macrophage-specific C5ar1 had ameliorated cardiovascular disease on a high-cholesterol diet. Conversely, inflammatory gene signatures and IL-1ß produced by cells in unstable atherosclerotic plaques of patients were normalized by a specific cell-permeable C5aR1 antagonist. Deficiency of the macrophage cell-autonomous C5 system also protected mice from crystal nephropathy mediated by folic acid. These data demonstrate the unexpected intracellular formation of a C5 convertase and identify C5aR1 as a direct modulator of mitochondrial function and inflammatory output from myeloid cells. Together, these findings suggest that the complosome is a contributor to the biologic processes underlying sterile inflammation and indicate that targeting this system could be beneficial in macrophage-dependent diseases, such as atherosclerosis.


Asunto(s)
Inflamación/inmunología , Interleucina-1beta/biosíntesis , Macrófagos/inmunología , Receptor de Anafilatoxina C5a/inmunología , Animales , Línea Celular , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Anafilatoxina C5a/deficiencia
3.
Front Immunol ; 12: 661290, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995384

RESUMEN

Intestinal immunity is coordinated by specialized mononuclear phagocyte populations, constituted by a diversity of cell subsets. Although the cell subsets constituting the mononuclear phagocyte network are thought to be similar in both small and large intestine, these organs have distinct anatomy, microbial composition, and immunological demands. Whether these distinctions demand organ-specific mononuclear phagocyte populations with dedicated organ-specific roles in immunity are unknown. Here we implement a new strategy to subset murine intestinal mononuclear phagocytes and identify two novel subsets which are colon-specific: a macrophage subset and a Th17-inducing dendritic cell (DC) subset. Colon-specific DCs and macrophages co-expressed CD24 and CD14, and surprisingly, both were dependent on the transcription factor IRF4. Novel IRF4-dependent CD14+CD24+ macrophages were markedly distinct from conventional macrophages and failed to express classical markers including CX3CR1, CD64 and CD88, and surprisingly expressed little IL-10, which was otherwise robustly expressed by all other intestinal macrophages. We further found that colon-specific CD14+CD24+ mononuclear phagocytes were essential for Th17 immunity in the colon, and provide definitive evidence that colon and small intestine have distinct antigen presenting cell requirements for Th17 immunity. Our findings reveal unappreciated organ-specific diversity of intestine-resident mononuclear phagocytes and organ-specific requirements for Th17 immunity.


Asunto(s)
Colon/inmunología , Células Dendríticas/inmunología , Macrófagos/inmunología , Fagocitos/inmunología , Células Th17/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígeno CD24/inmunología , Antígeno CD24/metabolismo , Colon/citología , Colon/metabolismo , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Células Dendríticas/metabolismo , Expresión Génica/inmunología , Factores Reguladores del Interferón/inmunología , Factores Reguladores del Interferón/metabolismo , Intestino Delgado/inmunología , Receptores de Lipopolisacáridos/inmunología , Receptores de Lipopolisacáridos/metabolismo , Macrófagos/metabolismo , Ratones de la Cepa 129 , Ratones Noqueados , Ratones Transgénicos , Fagocitos/metabolismo , Receptor de Anafilatoxina C5a/inmunología , Receptor de Anafilatoxina C5a/metabolismo , Células Th17/metabolismo
4.
Cell Rep ; 35(2): 108995, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33852847

RESUMEN

The complement fragment C5a is closely associated with adaptive immune induction in the mucosa. However, the mechanisms that control CD8+ T cell responses by C5a have not been extensively explored. This study reveals that C5/C5a in the Peyer's patch (PP) subepithelial dome increases upon oral Listeria infection. We hypothesize that C5aR+ PP cells play an important role in the induction of antigen-specific T cell immunity. Using single-cell RNA sequencing, we identify C5aR- and lysozyme-expressing dendritic cells (C5aR+ LysoDCs) in PP and examine their role in CD8+ T cell immune induction. Stimulation of C5aR+ LysoDCs by C5a increases reactive oxygen species levels, leading to efficient antigen cross-presentation, which elicits an antigen-specific CD8+ T cell response. In C5-deficient mice, oral co-administration of C5a and Listeria enhances Listeria-specific cytotoxic T cell levels. Collectively, these findings suggest a role of the complement system in intestinal T cell immunity.


Asunto(s)
Complemento C5a/inmunología , Reactividad Cruzada , Mucosa Intestinal/inmunología , Listeria monocytogenes/inmunología , Ganglios Linfáticos Agregados/inmunología , Receptor de Anafilatoxina C5a/genética , Linfocitos T Citotóxicos/inmunología , Inmunidad Adaptativa , Animales , Presentación de Antígeno , Complemento C5a/genética , Complemento C5a/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Inmunidad Mucosa , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Listeria monocytogenes/patogenicidad , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/microbiología , Muramidasa/genética , Muramidasa/inmunología , Ganglios Linfáticos Agregados/efectos de los fármacos , Ganglios Linfáticos Agregados/microbiología , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Receptor de Anafilatoxina C5a/inmunología , Análisis de la Célula Individual , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/microbiología
5.
J Allergy Clin Immunol ; 147(6): 2083-2097.e6, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33852936

RESUMEN

BACKGROUND: Excessive inflammation triggered by a hitherto undescribed mechanism is a hallmark of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections and is associated with enhanced pathogenicity and mortality. OBJECTIVE: Complement hyperactivation promotes lung injury and was observed in patients suffering from Middle East respiratory syndrome-related coronavirus, SARS-CoV-1, and SARS-CoV-2 infections. Therefore, we investigated the very first interactions of primary human airway epithelial cells on exposure to SARS-CoV-2 in terms of complement component 3 (C3)-mediated effects. METHODS: For this, we used highly differentiated primary human 3-dimensional tissue models infected with SARS-CoV-2 patient isolates. On infection, viral load, viral infectivity, intracellular complement activation, inflammatory mechanisms, and tissue destruction were analyzed by real-time RT-PCR, high content screening, plaque assays, luminex analyses, and transepithelial electrical resistance measurements. RESULTS: Here, we show that primary normal human bronchial and small airway epithelial cells respond to SARS-CoV-2 infection by an inflated local C3 mobilization. SARS-CoV-2 infection resulted in exaggerated intracellular complement activation and destruction of the epithelial integrity in monolayer cultures of primary human airway cells and highly differentiated, pseudostratified, mucus-producing, ciliated respiratory tissue models. SARS-CoV-2-infected 3-dimensional cultures secreted significantly higher levels of C3a and the proinflammatory cytokines IL-6, monocyte chemoattractant protein 1, IL-1α, and RANTES. CONCLUSIONS: Crucially, we illustrate here for the first time that targeting the anaphylotoxin receptors C3a receptor and C5a receptor in nonimmune respiratory cells can prevent intrinsic lung inflammation and tissue damage. This opens up the exciting possibility in the treatment of COVID-19.


Asunto(s)
Bronquios/inmunología , COVID-19/inmunología , Activación de Complemento , Células Epiteliales/inmunología , Receptor de Anafilatoxina C5a/inmunología , Mucosa Respiratoria/inmunología , SARS-CoV-2/inmunología , Bronquios/patología , Bronquios/virología , COVID-19/patología , COVID-19/virología , Línea Celular , Complemento C3/inmunología , Citocinas/inmunología , Células Epiteliales/patología , Células Epiteliales/virología , Humanos , Inflamación/inmunología , Inflamación/patología , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología
6.
Mol Immunol ; 131: 112-120, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33446393

RESUMEN

BACKGROUND: Networks formed of numerous autoantibodies (aabs) directed against G-protein coupled receptors (GPCR) have been suggested to play important role in autoimmune disorders. In present study, we aimed to evaluate the association between anti-GPCR antibodies and primary Sjogren's syndrome (pSS) to determine the potential pathogenic factors. METHODS: By applying a cell membrane-based ELISA technique, which is capable of detecting aabs against conformational epitopes within GPCR, serum levels of fourteen GPCR were determined in well-characterized patients with pSS (n = 52) and gender-matched healthy controls (n = 54). Comparisons between groups were analyzed by two-tailed Mann-Whitney U test, Bonferroni correction was applied for multiple comparisons. Spearman`s rank correlation coefficients were calculated between variables and visualized by heat map. RESULTS: Compared to healthy subjects, sera of patients with pSS showed significantly higher binding to ß2AR and ETAR, but lower binding to C5aR1, C3aR1, CXCR3, and CXCR4. Autoantibodies against C5aR1, C3aR1, CXCR3, and CXCR4 were also decreased in patients with rheumatoid arthritis. In pSS patients, levels of anti-CXCR3 and anti-CXCR4 antibodies were negatively correlated with circulating lymphocyte counts. Furthermore, correlation signatures of anti-GPCR antibodies changed dramatically in the patients with pulmonary involvement. CONCLUSIONS: This study demonstrates an association between pSS and autoantibodies recognizing GPCR, especially those functionally involved in immune cell migration and exocrine glandular secretion.


Asunto(s)
Autoanticuerpos/inmunología , Receptor de Anafilatoxina C5a/inmunología , Receptores CXCR3/inmunología , Receptores CXCR4/inmunología , Receptores de Complemento/inmunología , Síndrome de Sjögren/inmunología , Adulto , Anciano , Animales , Artritis Reumatoide/inmunología , Células CHO , Estudios de Casos y Controles , Cricetulus , Femenino , Humanos , Linfocitos/inmunología , Masculino , Persona de Mediana Edad
7.
J Clin Invest ; 131(5)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33351779

RESUMEN

Primary membranous nephropathy (pMN) is a leading cause of nephrotic syndrome in adults. In most cases, this autoimmune kidney disease is associated with autoantibodies against the M-type phospholipase A2 receptor (PLA2R1) expressed on kidney podocytes, but the mechanisms leading to glomerular damage remain elusive. Here, we developed a cell culture model using human podocytes and found that anti-PLA2R1-positive pMN patient sera or isolated IgG4, but not IgG4-depleted sera, induced proteolysis of the 2 essential podocyte proteins synaptopodin and NEPH1 in the presence of complement, resulting in perturbations of the podocyte cytoskeleton. Specific blockade of the lectin pathway prevented degradation of synaptopodin and NEPH1. Anti-PLA2R1 IgG4 directly bound mannose-binding lectin in a glycosylation-dependent manner. In a cohort of pMN patients, we identified increased levels of galactose-deficient IgG4, which correlated with anti-PLA2R1 titers and podocyte damage induced by patient sera. Assembly of the terminal C5b-9 complement complex and activation of the complement receptors C3aR1 or C5aR1 were required to induce proteolysis of synaptopodin and NEPH1 by 2 distinct proteolytic pathways mediated by cysteine and aspartic proteinases, respectively. Together, these results demonstrated a mechanism by which aberrantly glycosylated IgG4 activated the lectin pathway and induced podocyte injury in primary membranous nephropathy.


Asunto(s)
Autoanticuerpos/inmunología , Enfermedades Autoinmunes/inmunología , Lectina de Unión a Manosa de la Vía del Complemento/inmunología , Glomerulonefritis Membranosa/inmunología , Inmunoglobulina G/inmunología , Síndrome Nefrótico/inmunología , Podocitos/inmunología , Receptores de Fosfolipasa A2/inmunología , Adulto , Enfermedades Autoinmunes/patología , Proteínas Portadoras/inmunología , Línea Celular Transformada , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Glomerulonefritis Membranosa/patología , Humanos , Proteínas de la Membrana/inmunología , Proteínas de Microfilamentos/inmunología , Síndrome Nefrótico/patología , Podocitos/patología , Receptor de Anafilatoxina C5a/inmunología , Receptores de Complemento/inmunología
9.
Front Immunol ; 11: 1397, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32733463

RESUMEN

Toxoplasma gondii (T. gondii) is a parasite infecting animals and humans. In intermediate hosts, such as humans or rodents, rapidly replicating tachyzoites drive vigorous innate and adaptive immune responses resulting in bradyzoites that survive within tissue cysts. Activation of the innate immune system is critical during the early phase of infection to limit pathogen growth and to instruct parasite-specific adaptive immunity. In rodents, dendritic cells (DCs) sense T. gondii through TLR11/12, leading to IL-12 production, which activates NK cells to produce IFN-γ as an essential mechanism for early parasite control. Further, C3 can bind to T. gondii resulting in limited complement activation. Here, we determined the role of C5a/C5aR1 axis activation for the early innate immune response in a mouse model of peritoneal T. gondii infection. We found that C5ar1-/- animals suffered from significantly higher weight loss, disease severity, mortality, and parasite burden in the brain than wild type control animals. Severe infection in C5ar1-/- mice was associated with diminished serum concentrations of IL-12, IL-27, and IFN-γ. Importantly, the serum levels of pro-inflammatory cytokines, including IL-1α, IL-6, and TNF-α, as well as several CXC and CC chemokines, were decreased in comparison to wt animals, whereas anti-inflammatory IL-10 was elevated. The defect in IFN-γ production was associated with diminished Ifng mRNA expression in the spleen and the brain, reduced frequency of IFN-γ+ NK cells in the spleen, and decreased Nos2 expression in the brain of C5ar1-/- mice. Mechanistically, DCs from the spleen of C5ar1-/- mice produced significantly less IL-12 in response to soluble tachyzoite antigen (STAg) stimulation in vivo and in vitro. Our findings suggest a model in which the C5a/C5aR1 axis promotes IL-12 induction in splenic DCs that is critical for IFN-γ production from NK cells and subsequent iNOS expression in the brain as a critical mechanism to control acute T. gondii infection.


Asunto(s)
Activación de Complemento/inmunología , Inmunidad Innata/inmunología , Interferón gamma/inmunología , Receptor de Anafilatoxina C5a/inmunología , Toxoplasmosis Animal/inmunología , Animales , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/inmunología , Toxoplasma/inmunología
10.
J Clin Invest ; 130(11): 6151-6157, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32759504

RESUMEN

Emerging data indicate that complement and neutrophils contribute to the maladaptive immune response that fuels hyperinflammation and thrombotic microangiopathy, thereby increasing coronavirus 2019 (COVID-19) mortality. Here, we investigated how complement interacts with the platelet/neutrophil extracellular traps (NETs)/thrombin axis, using COVID-19 specimens, cell-based inhibition studies, and NET/human aortic endothelial cell (HAEC) cocultures. Increased plasma levels of NETs, tissue factor (TF) activity, and sC5b-9 were detected in patients. Neutrophils of patients yielded high TF expression and released NETs carrying active TF. Treatment of control neutrophils with COVID-19 platelet-rich plasma generated TF-bearing NETs that induced thrombotic activity of HAECs. Thrombin or NETosis inhibition or C5aR1 blockade attenuated platelet-mediated NET-driven thrombogenicity. COVID-19 serum induced complement activation in vitro, consistent with high complement activity in clinical samples. Complement C3 inhibition with compstatin Cp40 disrupted TF expression in neutrophils. In conclusion, we provide a mechanistic basis for a pivotal role of complement and NETs in COVID-19 immunothrombosis. This study supports strategies against severe acute respiratory syndrome coronavirus 2 that exploit complement or NETosis inhibition.


Asunto(s)
Betacoronavirus , Complejo de Ataque a Membrana del Sistema Complemento , Infecciones por Coronavirus , Trampas Extracelulares , Neutrófilos , Pandemias , Neumonía Viral , Tromboplastina , Trombosis , Anciano , Betacoronavirus/inmunología , Betacoronavirus/metabolismo , COVID-19 , Activación de Complemento/efectos de los fármacos , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Infecciones por Coronavirus/sangre , Infecciones por Coronavirus/inmunología , Trampas Extracelulares/inmunología , Trampas Extracelulares/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neutrófilos/inmunología , Neutrófilos/metabolismo , Péptidos Cíclicos/farmacología , Neumonía Viral/sangre , Neumonía Viral/inmunología , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/sangre , Receptor de Anafilatoxina C5a/inmunología , Síndrome de Dificultad Respiratoria/sangre , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/virología , SARS-CoV-2 , Trombina/inmunología , Trombina/metabolismo , Tromboplastina/inmunología , Tromboplastina/metabolismo , Trombosis/sangre , Trombosis/inmunología , Trombosis/virología
11.
Nature ; 588(7836): 146-150, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32726800

RESUMEN

Coronavirus disease 2019 (COVID-19) is a disease caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and has resulted in a pandemic1. The C5a complement factor and its receptor C5aR1 (also known as CD88) have a key role in the initiation and maintenance of several inflammatory responses by recruiting and activating neutrophils and monocytes1. Here we provide a longitudinal analysis of immune responses, including phenotypic analyses of immune cells and assessments of the soluble factors that are present in the blood and bronchoalveolar lavage fluid of patients at various stages of COVID-19 severity, including those who were paucisymptomatic or had pneumonia or acute respiratory distress syndrome. The levels of soluble C5a were increased in proportion to the severity of COVID-19 and high expression levels of C5aR1 receptors were found in blood and pulmonary myeloid cells, which supports a role for the C5a-C5aR1 axis in the pathophysiology of acute respiratory distress syndrome. Anti-C5aR1 therapeutic monoclonal antibodies prevented the C5a-mediated recruitment and activation of human myeloid cells, and inhibited acute lung injury in human C5aR1 knock-in mice. These results suggest that blockade of the C5a-C5aR1 axis could be used to limit the infiltration of myeloid cells in damaged organs and prevent the excessive lung inflammation and endothelialitis that are associated with acute respiratory distress syndrome in patients with COVID-19.


Asunto(s)
COVID-19/complicaciones , COVID-19/inmunología , Complemento C5a/inmunología , Inflamación/complicaciones , Inflamación/inmunología , Receptor de Anafilatoxina C5a/inmunología , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/prevención & control , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/inmunología , Antígeno CD11b/inmunología , Antígeno CD11b/metabolismo , COVID-19/sangre , COVID-19/patología , Complemento C5a/antagonistas & inhibidores , Complemento C5a/biosíntesis , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/prevención & control , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación/tratamiento farmacológico , Inflamación/patología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Células Mieloides/efectos de los fármacos , Células Mieloides/inmunología , Células Mieloides/patología , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/sangre , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/inmunología , Síndrome de Dificultad Respiratoria/prevención & control , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad
12.
FASEB J ; 34(6): 7540-7560, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32301538

RESUMEN

The vascular endothelium has been discovered in the past several years to be important in shaping the cellular immune response. During the immune response the vascular endothelium is constantly perturbed by biologically potent molecules, including the complement activation peptides, C3a and C5a. Despite the importance of C3a and C5a in inflammation and immunity, their role in modulating lymphocyte function via activation of vascular endothelial cells is unknown. Accordingly, we investigated the regulated expression of the C3a and C5a receptors (complement anaphylatoxin C3a receptor [C3aR] and complement anaphylatoxin C5a receptor 1 [C5aR1]) on human umbilical vascular endothelial cells (HUVECs) and examined how C3a or C5a activation of HUVECs affects the activation and polarization of lymphatic cells. Our findings demonstrated that C3a and C5a increase C3aR and C5aR1 expression by HUVECs as well as directing their cellular transmigration and spreading through transwell filters. Moreover, C3a- or C5a-stimulated endothelial cells: (1) caused activation of B-lymphoblasts with significant increase in Fas Ligand (CD95L) (FasL), CD69, and IL-R1 expression, and (2) skewed T-lymphoblast cells toward a Th1 subtype, (CD4+ /CCR5+ ) that correlated with significant increase of IFN-γ. Collectively, these data indicate that C3a and C5a signaling is important in the activation and polarization of lymphocytes as they traffic through the vascular endothelium during the immune response.


Asunto(s)
Anafilatoxinas/inmunología , Linfocitos B/inmunología , Complemento C3a/inmunología , Complemento C5a/inmunología , Péptidos/inmunología , Linfocitos T/inmunología , Células Cultivadas , Activación de Complemento/inmunología , Endotelio Vascular/inmunología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/inmunología , Receptor de Anafilatoxina C5a/inmunología , Receptores de Complemento/inmunología , Transducción de Señal/inmunología
13.
Vet Microbiol ; 242: 108599, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32122603

RESUMEN

Streptococcus suis is an emerging swine and human pathogen causing severe infections and sudden death. During infection, complement C3a and C5a were reported to induce immune cells towards infection and injury sites via their corresponding receptors C3aR and C5aR. However, how S. suis evade immune surveillance mediated by C3aR and C5aR remains unclear. In this study, we analyze and construct an S. suis bacterial two-hybrid prey library containing 39 LPXTG motif anchored proteins and 18 secreted proteins. Two highly possible C3aR-binding proteins: thiol-activated toxin Suilysin, putative RTX family exoprotein A gene and three highly possible C5aR-binding proteins: thiol-activated toxin Suilysin, putative 5'-nucleotidase and subtilisin-like serine protease are identified through bacterial two-hybrid assay. Far-western blot assay confirms that a cholesterol-binding cytolysin Suilysin can interact with both C3aR and C5aR. Chemotaxis assays demonstrate that recombinant and natural Suilysin can inhibit monocyte chemotaxis mediated by C3a and C5a. These findings enlarge our knowledge of suilysin biological significance and provide a new perspective on S. suis complement evasion.


Asunto(s)
Proteínas Bacterianas/inmunología , Quimiotaxis , Proteínas Hemolisinas/inmunología , Monocitos/inmunología , Receptor de Anafilatoxina C5a/inmunología , Receptores de Complemento/inmunología , Animales , Células Cultivadas , Humanos , Infecciones Estreptocócicas/microbiología , Streptococcus suis/química , Streptococcus suis/inmunología , Porcinos/inmunología , Porcinos/microbiología , Células THP-1
14.
Cells ; 9(2)2020 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-31991941

RESUMEN

Activation of the C5/C5a/C5a receptor 1 (C5aR1) axis during allergen sensitization protects from maladaptive T cell activation. To explore the underlying regulatory mechanisms, we analyzed the impact of C5aR1 activation on pulmonary CD11b+ conventional dendritic cells (cDCs) in the context of house-dust-mite (HDM) exposure. BALB/c mice were intratracheally immunized with an HDM/ovalbumin (OVA) mixture. After 24 h, we detected two CD11b+ cDC populations that could be distinguished on the basis of C5aR1 expression. C5aR1- but not C5aR1+ cDCs strongly induced T cell proliferation of OVA-reactive transgenic CD4+ T cells after re-exposure to antigen in vitro. C5aR1- cDCs expressed higher levels of MHC-II and CD40 than their C5aR1+ counterparts, which correlated directly with a higher frequency of interactions with cognate CD4+ T cells. Priming of OVA-specific T cells by C5aR1+ cDCs could be markedly increased by in vitro blockade of C5aR1 and this was associated with increased CD40 expression. Simultaneous blockade of C5aR1 and CD40L on C5aR1+ cDCs decreased T cell proliferation. Finally, pulsing with OVA-induced C5 production and its cleavage into C5a by both populations of CD11b+ cDCs. Thus, we propose a model in which allergen-induced autocrine C5a generation and subsequent C5aR1 activation in pulmonary CD11b+ cDCs promotes tolerance towards aeroallergens through downregulation of CD40.


Asunto(s)
Alérgenos/inmunología , Antígeno CD11b/metabolismo , Linfocitos T CD4-Positivos/inmunología , Antígenos CD40/metabolismo , Células Dendríticas/inmunología , Tolerancia Inmunológica , Pulmón/inmunología , Receptor de Anafilatoxina C5a/metabolismo , Animales , Antígeno CD11b/inmunología , Antígenos CD40/inmunología , Diferenciación Celular/inmunología , Proliferación Celular/genética , Técnicas de Cocultivo , Complemento C5a/fisiología , Células Dendríticas/metabolismo , Femenino , Antígenos de Histocompatibilidad Clase II/metabolismo , Tolerancia Inmunológica/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Pyroglyphidae/inmunología , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/inmunología , Receptores CCR7/metabolismo
15.
Front Immunol ; 10: 1866, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31447855

RESUMEN

Psoriasis is one of the most common chronic inflammatory skin diseases, affecting ~2% of the population. The lack of characterization of the pathogenesis of psoriasis has hindered efficient clinical treatment of the disease. In our study, we observed that expression of complement component 5a receptor 1(C5aR1) was significantly increased in skin lesions of both imiquimod (IMQ) and IL23-induced psoriatic mice and patients with psoriasis. C5aR1 deficiency or treatment with C5a receptor 1 antagonist (C5aR1a) in mice significantly attenuated psoriasis-like skin lesions and expression of inflammatory cytokines and chemokines. Moreover, C5aR1 deficiency significantly decreased IMQ-induced infiltration of plasmacytoid dendritic cells (pDCs), monocytes and neutrophils in psoriatic skin lesions and functions of pDCs, evidenced by the remarkable reduction in the IMQ-induced production of interferon-α (IFN-α) and tumor necrosis factor α (TNF-α), and FMS-like tyrosine kinase 3 ligand (FLT3L)-dependent pDCs differentiation. Accordingly, in vitro treatment with recombinant C5a accelerated pDCs migration and the differentiation of bone marrow cells into pDCs. Furthermore, biopsies of psoriatic patients showed a dramatic increase of C5aR1+ pDCs infiltration in psoriatic skin lesions, compared to healthy subjects. Our results provide direct evidence that C5a/C5aR1 signaling plays a critical role in the pathogenesis of psoriasis. Inhibition of C5a/C5aR1 pathway is expected to be beneficial in the treatment of patients with psoriasis.


Asunto(s)
Complemento C5a/inmunología , Psoriasis/inmunología , Receptor de Anafilatoxina C5a/inmunología , Animales , Complemento C5a/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Psoriasis/metabolismo , Psoriasis/patología , Receptor de Anafilatoxina C5a/metabolismo
16.
Front Immunol ; 10: 1574, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31379815

RESUMEN

Cancer immunotherapy has made remarkable clinical advances in recent years. Antibodies targeting the immune checkpoint receptors PD-1 and CTLA-4 and adoptive cell therapy (ACT) based on ex vivo expanded peripheral CTLs, tumor infiltrating lymphocytes (TILs), gene-engineered TCR- and chimeric antigen receptor (CAR)-T cells have all shown durable clinical efficacies in multiple types of cancers. However, these immunotherapeutic approaches only benefit a small fraction of cancer patients as various immune resistance mechanisms and limitations make their effective use a challenge in the majority of cancer patients. For example, adaptive resistance to therapeutic PD-1 blockade is associated with an upregulation of some additional immune checkpoint receptors. The efficacy of transferred tumor-specific T cells under the current clinical ACT protocol is often limited by their inefficient engraftment, poor persistence, and weak capability to attack tumor cells. Recent studies demonstrate that the complement receptor C3aR and C5aR function as a new class of immune checkpoint receptors. Complement signaling through C3aR and C5aR expressed on effector T lymphocytes prevent the production of the cytokine interleukin-10 (IL-10). Removing C3aR/C5aR-mediated transcriptional suppression of IL-10 expression results in endogenous IL-10 production by antitumor effector T cells, which drives T cell expansion and enhances T cell-mediated antitumor immunity. Importantly, preclinical, and clinical data suggest that a signaling axis consisting of complement/C3aR/C5aR/IL-10 critically regulates T cell mediated antitumor immunity and manipulation of the pathway ex vivo and in vivo is an effective strategy for cancer immunotherapy. Furthermore, a combination of treatment strategies targeting the complement/C3aR/C5aR/IL-10 pathway with other treatment modalities may improve cancer therapeutic efficacy.


Asunto(s)
Neoplasias/inmunología , Receptor de Anafilatoxina C5a/inmunología , Receptores de Complemento/inmunología , Animales , Humanos , Inmunoterapia , Interleucina-10/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias/terapia
17.
Virulence ; 10(1): 677-694, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31274379

RESUMEN

The complement system is pivotal in the defense against invasive disease caused by Neisseria meningitidis (Nme, meningococcus), particularly via the membrane attack complex. Complement activation liberates the anaphylatoxins C3a and C5a, which activate three distinct G-protein coupled receptors, C3aR, C5aR1 and C5aR2 (anaphylatoxin receptors, ATRs). We recently discovered that C5aR1 exacerbates the course of the disease, revealing a downside of complement in Nme sepsis. Here, we compared the roles of all three ATRs during mouse nasal colonization, intraperitoneal infection and human whole blood infection with Nme. Deficiency of complement or ATRs did not alter nasal colonization, but significantly affected invasive disease: Compared to WT mice, the disease was aggravated in C3ar-/- mice, whereas C5ar1-/- and C5ar2-/- mice showed increased resistance to meningococcal sepsis. Surprisingly, deletion of either of the ATRs resulted in lower cytokine/chemokine responses, irrespective of the different susceptibilities of the mice. This was similar in ex vivo human whole blood infection using ATR inhibitors. Neutrophil responses to Nme were reduced in C5ar1-/- mouse blood. Upon stimulation with C5a plus Nme, mouse macrophages displayed reduced phosphorylation of ERK1/2, when C5aR1 or C5aR2 were ablated or inhibited, suggesting that both C5a-receptors prime an initial macrophage response to Nme. Finally, in vivo blockade of C5aR1 alone (PMX205) or along with C5aR2 (A8Δ71-73) resulted in ameliorated disease, whereas neither antagonizing C3aR (SB290157) nor its activation with a "super-agonist" peptide (WWGKKYRASKLGLAR) demonstrated a benefit. Thus, C5aR1 and C5aR2 augment disease pathology and are interesting targets for treatment, whereas C3aR is protective in experimental meningococcal sepsis.


Asunto(s)
Infecciones Meningocócicas/inmunología , Neisseria meningitidis/inmunología , Receptor de Anafilatoxina C5a/inmunología , Receptores de Complemento/inmunología , Anafilatoxinas/inmunología , Animales , Quimiocinas/inmunología , Citocinas/inmunología , Humanos , Macrófagos/inmunología , Macrófagos/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neisseria meningitidis/patogenicidad , Neutrófilos/inmunología , Neutrófilos/microbiología , Receptor de Anafilatoxina C5a/genética , Receptores de Complemento/genética , Sepsis
18.
Int J Mol Med ; 44(3): 823-834, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31257457

RESUMEN

Clinically, deep decay can lead to inflammation in the dental pulp. Apart from the use of various materials to sooth the inflamed pulp, there is currently no adequate treatment, and the gold standard, calcium hydroxide, that is used to cover the dentin/pulp, has limited effect. Sometimes the pulp will remain infected and cause pulpitis, and ultimately, the pulp will need to be removed. The first principle of oral treatment is to protect the pulp. Therefore, it is necessary to study the immune response and regeneration of pulp cells in conditions of deep decay. Of the terminal complement system proteins, complement 5a (C5a) has the most potent effect compared to complement 3a (C3a) and complement 4a (C4a). C5a is 20- to 2,500-fold stronger than C3a and C4a. The purpose of this study was to elucidate the association between C5a, secreted by complement activation, and the duration of inflammation. Another key goal was to detect the expression of C5a and its receptor, complement 5a receptor (C5aR). To this end, the cells were divided into 4 groups as per stimulation with lipoteichoic acid (LTA) or lipopolysaccharide (LPS) as follows: i) The 1 µg/ml LTA group; ii) the 1 µg/ml LPS group; iii) the 1 µg/ml LTA and 1 µg/ml LPS group; and iv) the PBS-only group, which served as a control. There were 5 time points for all 4 groups: 1, 2, 3, 5 and 7 days. Reverse transcription-quantitative polymerase chain reaction was used to detect the gene expression levels of C5a, C5aR and interleukin (IL)-6 at different time points. Western blot analyses was carried out to detect the expression of C5aR. Transmission electron microscopy was also conducted to assess the ultrastructural features of dental pulp cells. The gene expression trends of C5a and C5aR mRNA were identical. C5a and C5aR mRNA was highly expressed on the second day of LTA or LPS stimulation. However, in the LTA and LPS co-stimulation group, C5a and C5aR mRNA were highly expressed on both the first and second day, with higher levels on the second day. IL-6 expression decreased as time progressed in the LTA only and in the LTA + LPS co-stimulation groups. However, a peak in its expression was observed on the second day in the LPS group. On the whole, this study demonstrates that a 1 µg/ml concentration of LTA and LPS stimulates human dental pulp cells to activate the expression of C5a.


Asunto(s)
Complemento C5a/genética , Complemento C5a/inmunología , Pulpa Dental/citología , Pulpa Dental/metabolismo , Regulación de la Expresión Génica , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/inmunología , Células Madre/metabolismo , Adipocitos/metabolismo , Adolescente , Adulto , Biomarcadores , Diferenciación Celular , Células Cultivadas , Complemento C5a/metabolismo , Citocinas/metabolismo , Femenino , Humanos , Lipopolisacáridos/inmunología , Masculino , Osteoblastos/citología , Osteoblastos/metabolismo , Receptor de Anafilatoxina C5a/metabolismo , Células Madre/inmunología , Ácidos Teicoicos/inmunología , Adulto Joven
19.
J Immunol ; 203(2): 379-388, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31217324

RESUMEN

The involvement of complement in B2 cell responses has been regarded as occurring strictly via complement components in plasma. In this study, we show that Ab production and class switch recombination (CSR) depend on autocrine C3a and C5a receptor (C3ar1/C5ar1) signaling in B2 cells. CD40 upregulation, IL-6 production, growth in response to BAFF or APRIL, and AID/Bcl-6 expression, as well as follicular CD4+ cell CD21 production, all depended on this signal transduction. OVA immunization of C3ar1-/-C5ar1-/- mice elicited IgM Ab but no other isotypes, whereas decay accelerating factor (Daf1)-/- mice elicited more robust Ab production and CSR than wild-type (WT) mice. Comparable differences occurred in OVA-immunized µMT recipients of WT, C3ar1-/-C5ar1-/- , and Daf1-/- B2 cells and in hen egg lysozyme-immunized µMT recipients of MD4 B2 cells on each genetic background. B2 cells produced factor I and C3 and autophosphorylated CD19. Immunized C3-/-C5-/- recipients of WT MD4 bone marrow efficiently produced Ab. Thus, B2 cell-produced complement participates in B2 cell activation.


Asunto(s)
Comunicación Autocrina/inmunología , Receptor de Anafilatoxina C5a/inmunología , Receptores de Complemento/inmunología , Animales , Antígenos CD19/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Cultivadas , Proteínas del Sistema Complemento/inmunología , Interleucina-6/inmunología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/inmunología
20.
Mol Immunol ; 112: 131-139, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31102985

RESUMEN

BACKGROUND: Sepsis is a dysregulated host response to infection. The aim of this study was to investigate the effects of complement- and CD14 inhibition on phagocytosis of live and dead Gram-negative and Gram-positive bacteria in human whole blood. METHODS: Lepirudin-anticoagulated blood was incubated with live or dead E. coli or S. aureus at 37 °C for 120 min with or without the C5aR1 antagonist PMX53 and/or anti-CD14. Granulocyte and monocyte phagocytosis were measured by flow cytometry, and five plasma cytokines by multiplex, yielding a total of 28 mediators of inflammation tested for. RESULTS: 16/28 conditions were reduced by PMX53, 7/28 by anti-CD14, and 24/28 by combined PMX53 and CD14 inhibition. The effect of complement inhibition was quantitatively more pronounced, in particular for the responses to S. aureus. The effect of anti-CD14 was modest, except for a marked reduction in INF-ß. The responses to live and dead S. aureus were equally inhibited, whereas the responses to live E. coli were inhibited less than those to dead E. coli. CONCLUSION: C5aR1 inhibited phagocytosis-induced inflammation by live and dead E. coli and S. aureus. CD14 blockade potentiated the effect of C5aR1 blockade, thus attenuating inflammation.


Asunto(s)
Escherichia coli/inmunología , Receptores de Lipopolisacáridos/antagonistas & inhibidores , Fagocitosis/inmunología , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Activación de Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Citocinas/inmunología , Infecciones por Escherichia coli/inmunología , Granulocitos/inmunología , Humanos , Inflamación/inmunología , Inflamación/microbiología , Interferón beta/inmunología , Receptores de Lipopolisacáridos/inmunología , Monocitos/inmunología , Monocitos/microbiología , Péptidos Cíclicos/inmunología , Receptor de Anafilatoxina C5a/inmunología , Sepsis/inmunología , Sepsis/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...