Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.062
Filtrar
1.
FASEB J ; 36(11): e22599, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36250902

RESUMEN

Emerging evidence suggest that C3aR plays important roles in homeostasis, host defense and disease. Although it is known that C3aR is protective in several models of acute bacterial infections, the role for C3aR in chronic infection is largely unknown. Here we show that C3aR is protective in experimental chronic pyelonephritis. Global C3aR deficient (C3ar-/- ) mice had higher renal bacterial load, more pronounced renal histological lesions, increased renal apoptotic cell accumulation, tissue inflammation and extracellular matrix deposition following renal infection with uropathogenic E. coli (UPEC) strain IH11128, compared to WT control mice. Myeloid C3aR deficient (Lyz2-C3ar-/- ) mice exhibited a similar disease phenotype to global C3ar-/- mice. Pharmacological treatment with a C3aR agonist reduced disease severity in experimental chronic pyelonephritis. Furthermore, macrophages of C3ar-/- mice exhibited impaired ability to phagocytose UPEC. Our data clearly demonstrate a protective role for C3aR against experimental chronic pyelonephritis, macrophage C3aR plays a major role in the protection, and C3aR is necessary for phagocytosis of UPEC by macrophages. Our observation that C3aR agonist curtailed the pathology suggests a therapeutic potential for activation of C3aR in chronic infection.


Asunto(s)
Infecciones por Escherichia coli , Pielonefritis , Receptores de Complemento , Animales , Ratones , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/patología , Inflamación/inmunología , Inflamación/microbiología , Inflamación/patología , Riñón/microbiología , Riñón/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Pielonefritis/inmunología , Pielonefritis/microbiología , Pielonefritis/patología , Pielonefritis/prevención & control , Escherichia coli Uropatógena/patogenicidad , Receptores de Complemento/agonistas , Receptores de Complemento/deficiencia , Receptores de Complemento/genética , Receptores de Complemento/inmunología , Matriz Extracelular/metabolismo
2.
Nat Commun ; 12(1): 7172, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34887405

RESUMEN

Complement receptor of immunoglobulin superfamily (CRIg) is expressed on liver macrophages and directly binds complement component C3b or Gram-positive bacteria to mediate phagocytosis. CRIg plays important roles in several immune-mediated diseases, but it is not clear how its pathogen recognition and phagocytic functions maintain homeostasis and prevent disease. We previously associated cytolysin-positive Enterococcus faecalis with severity of alcohol-related liver disease. Here, we demonstrate that CRIg is reduced in liver tissues from patients with alcohol-related liver disease. CRIg-deficient mice developed more severe ethanol-induced liver disease than wild-type mice; disease severity was reduced with loss of toll-like receptor 2. CRIg-deficient mice were less efficient than wild-type mice at clearing Gram-positive bacteria such as Enterococcus faecalis that had translocated from gut to liver. Administration of the soluble extracellular domain CRIg-Ig protein protected mice from ethanol-induced steatohepatitis. Our findings indicate that ethanol impairs hepatic clearance of translocated pathobionts, via decreased hepatic CRIg, which facilitates progression of liver disease.


Asunto(s)
Enterococcus faecalis/inmunología , Infecciones por Bacterias Grampositivas/inmunología , Hepatopatías Alcohólicas/inmunología , Macrófagos/inmunología , Receptores de Complemento 3b/inmunología , Receptores de Complemento/inmunología , Animales , Traslocación Bacteriana , Complemento C3b/inmunología , Enterococcus faecalis/fisiología , Etanol/efectos adversos , Femenino , Tracto Gastrointestinal/microbiología , Infecciones por Bacterias Grampositivas/genética , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/microbiología , Hepatopatías Alcohólicas/etiología , Hepatopatías Alcohólicas/genética , Hepatopatías Alcohólicas/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Complemento/deficiencia , Receptores de Complemento/genética , Receptores de Complemento 3b/genética
3.
Mol Immunol ; 137: 75-83, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34229135

RESUMEN

Macrophages polarize into functionally divergent phenotypes - M1 and M2 - which express distinct receptors. These cells are known to express complement receptors, including CR1, CR3, and CR4. However, whether these complement receptors are differentially expressed on M1 and M2 macrophages is not yet known. Herein, we have examined the expression of CR1 to CR4 on murine bone marrow-derived M1 (stimulated with IFN-γ or LPS) and M2 (stimulated with IL-4 or IL-4 + IL-13) macrophages. We show that M1 cells exhibit increased expression of CR1/2, whereas M2 cells display the higher expression of CR4; CR3 is equally expressed on both the phenotypes. Thus, M1 cells are CR1/2+CR4+, and M2 are CR1/2-CR4+. Functional probing of these cells for their phagocytic ability indicates that M1 cells, which express higher CR1/2, internalize a significantly greater number of C3b-opsonized erythrocytes. Both M1 and M2 cells, on the other hand, internalize iC3b-opsonized erythrocytes to a similar extent. Interestingly, the phagocytic receptor involved in phagocytosis of iC3b-opsonized erythrocytes is only CR3 with no contribution of CR4. We, thus, propose that complement receptor expression can be used in combination with the expression of other known polarization markers to better locate a macrophage along its phenotypic spectrum.


Asunto(s)
Macrófagos/inmunología , Receptores de Complemento/inmunología , Animales , Médula Ósea/inmunología , Eritrocitos/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fagocitosis/inmunología
4.
Immunol Lett ; 237: 42-57, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34186155

RESUMEN

The involvement of complement in the regulation of antibody responses has been known for long. By now several additional B cell functions - including cytokine production and antigen presentation - have also been shown to be regulated by complement proteins. Most of these important activities are mediated by receptors interacting with activation fragments of the central component of the complement system C3, such as C3b, iC3b and C3d, which are covalently attached to antigens and immune complexes. This review summarizes the role of complement receptors interacting with these ligands, namely CR1 (CD35), CR2 (CD21), CR3 (CD11b/CD18) and CR4 (CD11c/CD18) expressed by B cells in health and disease. Although we focus on human B lymphocytes, we also aim to call the attention to important differences between human and mouse systems.


Asunto(s)
Linfocitos B/inmunología , Complemento C3/inmunología , Receptores de Complemento/inmunología , Animales , Formación de Anticuerpos , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , División Celular , Expresión Génica , Humanos , Memoria Inmunológica , Ligandos , Ratones , Especificidad de Órganos , Receptores de Antígenos de Linfocitos B/inmunología , Receptores de Complemento/química , Receptores de Complemento/genética , Especificidad de la Especie , Relación Estructura-Actividad
5.
PLoS Pathog ; 17(6): e1009655, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34125873

RESUMEN

Microbial pathogens bind host complement regulatory proteins to evade the immune system. The bacterial pathogen Neisseria meningitidis, or meningococcus, binds several complement regulators, including human Factor H (FH). FH binding protein (FHbp) is a component of two licensed meningococcal vaccines and in mice FHbp elicits antibodies that inhibit binding of FH to FHbp, which defeat the bacterial evasion mechanism. However, humans vaccinated with FHbp develop antibodies that enhance binding of FH to the bacteria, which could limit the effectiveness of the vaccines. In the present study, we show that two vaccine-elicited antibody fragments (Fabs) isolated from different human subjects increase binding of complement FH to meningococcal FHbp by ELISA. The two Fabs have different effects on the kinetics of FH binding to immobilized FHbp as measured by surface plasmon resonance. The 1.7- and 2.0-Å resolution X-ray crystal structures of the Fabs in complexes with FHbp illustrate that the two Fabs bind to similar epitopes on the amino-terminal domain of FHbp, adjacent to the FH binding site. Superposition models of ternary complexes of each Fab with FHbp and FH show that there is likely minimal contact between the Fabs and FH. Collectively, the structures reveal that the Fabs enhance binding of FH to FHbp by altering the conformations and mobilities of two loops adjacent to the FH binding site of FHbp. In addition, the 1.5 Å-resolution structure of one of the isolated Fabs defines the structural rearrangements associated with binding to FHbp. The FH-enhancing human Fabs, which are mirrored in the human polyclonal antibody responses, have important implications for tuning the effectiveness of FHbp-based vaccines.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Factor H de Complemento/inmunología , Vacunas Meningococicas/inmunología , Anticuerpos Antibacterianos/metabolismo , Factor H de Complemento/metabolismo , Humanos , Infecciones Meningocócicas/prevención & control , Neisseria meningitidis , Receptores de Complemento/inmunología , Receptores de Complemento/metabolismo , Factores de Virulencia/inmunología
6.
Int Immunopharmacol ; 96: 107616, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34162127

RESUMEN

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is the main pathological subtype of esophageal cancer with high incidence and mortality. Immune and stromal cells in the tumor microenvironment (TME) profoundly affect the development of ESCC. METHODS: In this study, we used the ESTIMATE algorithm to calculate the immune and stromal scores of ESCC samples in The Cancer Genome Atlas (TCGA) database. Next, we used the R package limma to identify differentially expressed genes (DEGs) from high- versus low-immune/stromal score groups and these DEGs were further utilized to analyze the functional annotations, protein-protein interaction (PPI) networks and overall survival of patients with ESCC. Finally, we identified the biological roles of core gene C3AR1 in the TME of ESCC using the TCGA database and in vitro experiments. RESULTS: We obtained the immune and stromal scores of ESCC samples and further evaluated the impact of these scores on the prognosis and clinical parameters of patients with ESCC. Next, we identified 410 DEGs from high- versus low-immune/stromal score groups and to gain better understanding of the biological functions and characteristics of DEGs. Among these DEGs, 69 were correlated with the overall survival of patients with ESCC and C3AR1 was identified as a core gene for the regulation of most genes in the network. We found that C3AR1 was positively correlated with M2 macrophages and immune inhibitory molecules (T-cell immunoglobulin and mucin domain 3 (TIM-3), programmed cell death-1 (PD-1)), but not with M1 macrophages. We also observed a higher expression of CD163 and CD206, which were the markers for M2 macrophages in the TLQP-21 TFA (the agonist of C3AR1)groups than in the control groups. CONCLUSION: Based on the ESTIMATE algorithm, we obtained and characterized prognosis-related genes in the TME of ESCC samples from the TCGA database. We have further revealed that C3AR1 may cause an immunosuppressive microenvironment by affecting the polarization of macrophages to M2 phenotype and lead to the progression of ESCC, which indicates that C3AR1 may be a potential target for immunotherapy.


Asunto(s)
Neoplasias Esofágicas/genética , Neoplasias Esofágicas/inmunología , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/inmunología , Receptores de Complemento/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Biomarcadores de Tumor/inmunología , Biología Computacional , Bases de Datos Factuales , Neoplasias Esofágicas/mortalidad , Carcinoma de Células Escamosas de Esófago/mortalidad , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Humanos , Estimación de Kaplan-Meier , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Persona de Mediana Edad , Pronóstico , Receptor de Muerte Celular Programada 1/metabolismo , Mapas de Interacción de Proteínas/genética , Mapas de Interacción de Proteínas/inmunología , Receptores de Superficie Celular/metabolismo , Receptores de Complemento/agonistas , Receptores de Complemento/metabolismo , Receptores Inmunológicos/metabolismo
7.
Front Immunol ; 12: 662164, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995387

RESUMEN

The ß2-integrin receptor family has a broad spectrum of physiological functions ranging from leukocyte adhesion, cell migration, activation, and communication to the phagocytic uptake of cells and particles. Among the members of this family, complement receptor 3 (CR3; CD11b/CD18, Mac-1, αMß2) is particularly promiscuous in its functional profile and ligand selectivity. There are close to 100 reported structurally unrelated ligands for CR3, and while many ligands appear to cluster at the αMI domain, molecular details about binding modes remain largely elusive. The versatility of CR3 is reflected in its functional portfolio, which includes prominent roles in the removal of invaders and cell debris, induction of tolerance and synaptic pruning, and involvement in the pathogenesis of numerous autoimmune and chronic inflammatory pathologies. While CR3 is an interesting therapeutic target for immune modulation due to these known pathophysiological associations, drug development efforts are limited by concerns of potential interference with host defense functions and, most importantly, an insufficient molecular understanding of the interplay between ligand binding and functional impact. Here, we provide a systematic summary of the various interaction partners of CR3 with a focus on binding mechanisms and functional implications. We also discuss the roles of CR3 as an immune receptor in health and disease, as an activation marker in research and diagnostics, and as a therapeutic target.


Asunto(s)
Receptores de Complemento/inmunología , Receptores de Complemento/metabolismo , Animales , Movimiento Celular , Desarrollo de Medicamentos , Humanos , Integrinas/inmunología , Leucocitos/metabolismo , Ligandos , Ratones , Neutrófilos/inmunología , Receptores de Complemento/clasificación
8.
Front Immunol ; 12: 660194, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33868311

RESUMEN

Inflammation is a common denominator of diseases. The complement system, an intrinsic part of the innate immune system, is a key driver of inflammation in numerous disorders. Recently, a family of proteins has been suggested to be of vital importance in conditions characterized by complement dysregulation: the human Factor H (FH) family. This group of proteins consists of FH, Factor H-like protein 1 and five Factor H-related proteins. The FH family has been linked to infectious, vascular, eye, kidney and autoimmune diseases. In contrast to FH, the functions of the other highly homologous proteins are largely unknown and, hence, their role in the different disease-specific pathogenic mechanisms remains elusive. In this perspective review, we address the major challenges ahead in this emerging area, including 1) the controversies about the functional roles of the FH protein family, 2) the discrepancies in quantification of the FH protein family, 3) the unmet needs for validated tools and 4) limitations of animal models. Next, we also discuss the opportunities that exist for the immunology community. A strong multidisciplinary approach is required to solve these obstacles and is only possible through interdisciplinary collaboration between biologists, chemists, geneticists and physicians. We position this review in light of our own perspective, as principal investigators of the SciFiMed Consortium, a consortium aiming to create a comprehensive analytical system for the quantitative and functional assessment of the entire FH protein family.


Asunto(s)
Activación de Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Inflamación/inmunología , Degeneración Macular/inmunología , Receptores de Complemento/inmunología , Animales , Factor H de Complemento/análisis , Factor H de Complemento/inmunología , Factor H de Complemento/metabolismo , Proteínas del Sistema Complemento/análisis , Proteínas del Sistema Complemento/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , Humanos , Inflamación/sangre , Inflamación/metabolismo , Degeneración Macular/metabolismo , Receptores de Complemento/metabolismo , Reproducibilidad de los Resultados
9.
Int J Mol Sci ; 22(6)2021 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-33799879

RESUMEN

The development of nanocarriers (NC) for biomedical applications has gained large interest due to their potential to co-deliver drugs in a cell-type-targeting manner. However, depending on their surface characteristics, NC accumulate serum factors, termed protein corona, which may affect their cellular binding. We have previously shown that NC coated with carbohydrates to enable biocompatibility triggered the lectin-dependent complement pathway, resulting in enhanced binding to B cells via complement receptor (CR)1/2. Here we show that such NC also engaged all types of splenic leukocytes known to express CR3 at a high rate when NC were pre-incubated with native mouse serum resulting in complement opsonization. By focusing on dendritic cells (DC) as an important antigen-presenting cell type, we show that CR3 was essential for binding/uptake of complement-opsonized NC, whereas CR4, which in mouse is specifically expressed by DC, played no role. Further, a minor B cell subpopulation (B-1), which is important for first-line pathogen responses, and co-expressed CR1/2 and CR3, in general, engaged NC to a much higher extent than normal B cells. Here, we identified CR-1/2 as necessary for binding of complement-opsonized NC, whereas CR3 was dispensable. Interestingly, the binding of complement-opsonized NC to both DC and B-1 cells affected the expression of activation markers. Our findings may have important implications for the design of nano-vaccines against infectious diseases, which codeliver pathogen-specific protein antigen and adjuvant, aimed to induce a broad adaptive cellular and humoral immune response by inducing cytotoxic T lymphocytes that kill infected cells and pathogen-neutralizing antibodies, respectively. Decoration of nano-vaccines either with carbohydrates to trigger complement activation in vivo or with active complement may result in concomitant targeting of DC and B cells and thereby may strongly enhance the extent of dual cellular/humoral immune responses.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Linfocitos B/inmunología , Antígeno CD11b/inmunología , Proteínas del Sistema Complemento/inmunología , Células Dendríticas/inmunología , Receptores de Complemento/inmunología , Animales , Subgrupos de Linfocitos B/metabolismo , Linfocitos B/metabolismo , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Células Cultivadas , Activación de Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Células Dendríticas/metabolismo , Dextranos/química , Portadores de Fármacos/química , Humanos , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Nanopartículas/química , Proteínas Opsoninas/inmunología , Proteínas Opsoninas/metabolismo , Fagocitosis/inmunología , Receptores de Complemento/metabolismo
10.
Front Immunol ; 12: 580594, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33767691

RESUMEN

The zoonotic intracellular bacterium Chlamydia psittaci causes life-threatening pneumonia in humans. During mouse lung infection, complement factor C3 and the anaphylatoxin C3a augment protection against C. psittaci by a so far unknown mechanism. To clarify how complement contributes to the early, innate and the late, specific immune response and resulting protection, this study addresses the amount of C3, the timing when its presence is required as well as the anaphylatoxin receptor(s) mediating its effects and the complement-dependent migration of dendritic cells. Challenge experiments with C. psittaci on various complement KO mice were combined with transient decomplementation by pharmacological treatment, as well as the analysis of in vivo dendritic cells migration. Our findings reveal that a plasma concentration of C3 close to wildtype levels was required to achieve full protection. The diminished levels of C3 of heterozygote C3+/- mice permitted already relative effective protection and improved survival as compared to C3-/- mice, but overall recovery of these animals was delayed. Complement was in particular required during the first days of infection. However, additionally, it seems to support protection at later stages. Migration of CD103+ dendritic cells from the infected lung to the draining lymph node-as prerequisite of antigen presentation-depended on C3 and C3aR and/or C5aR. Our results provide unique mechanistic insight in various aspects of complement-dependent immune responses under almost identical, rather physiological experimental conditions. Our study contributes to an improved understanding of the role of complement, and C3a in particular, in infections by intracellular bacteria.


Asunto(s)
Movimiento Celular/inmunología , Infecciones por Chlamydiaceae/inmunología , Chlamydophila psittaci/inmunología , Complemento C3a/inmunología , Células Dendríticas/inmunología , Pulmón/inmunología , Anafilatoxinas/inmunología , Anafilatoxinas/metabolismo , Animales , Línea Celular , Infecciones por Chlamydiaceae/metabolismo , Infecciones por Chlamydiaceae/microbiología , Chlamydophila psittaci/fisiología , Activación de Complemento/inmunología , Complemento C3a/genética , Complemento C3a/metabolismo , Células Dendríticas/citología , Células Dendríticas/microbiología , Pulmón/metabolismo , Pulmón/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Complemento/genética , Receptores de Complemento/inmunología , Receptores de Complemento/metabolismo , Transducción de Señal/inmunología , Análisis de Supervivencia
11.
Curr Opin Virol ; 47: 52-67, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33581646

RESUMEN

Pathogen-specific immunity evolves in the context of the infected tissue. However, current immune correlates analyses and vaccine efficacy metrics are based on immune functions from peripheral cells. Less is known about tissue-resident mechanisms of immunity. While antibodies represent the primary correlate of immunity following most clinically approved vaccines, how antibodies interact with localized, compartment-specific immune functions to fight infections, remains unclear. Emerging data demonstrate a unique community of immune cells that reside within different tissues. These tissue-specific immunological communities enable antibodies to direct both expected and unexpected local attack strategies to control, disrupt, and eliminate infection in a tissue-specific manner. Defining the full breadth of antibody effector functions, how they selectively contribute to control at the site of infection may provide clues for the design of next-generation vaccines able to direct the control, elimination, and prevention of compartment specific diseases of both infectious and non-infectious etiologies.


Asunto(s)
Anticuerpos/inmunología , Animales , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/fisiología , Humanos , Inmunidad Innata , Fragmentos Fc de Inmunoglobulinas/inmunología , Intestinos/inmunología , Intestinos/microbiología , Hígado/inmunología , Hígado/microbiología , Especificidad de Órganos , Receptores de Complemento/inmunología , Piel/inmunología , Piel/microbiología , Vacunas/inmunología
12.
Microbiol Mol Biol Rev ; 85(1)2021 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-33504655

RESUMEN

The complement system is an evolutionarily ancient defense mechanism against foreign substances. Consisting of three proteolytic activation pathways, complement converges on a common effector cascade terminating in the formation of a lytic pore on the target surface. The classical and lectin pathways are initiated by pattern recognition molecules binding to specific ligands, while the alternative pathway is constitutively active at low levels in circulation. Complement-mediated killing is essential for defense against many Gram-negative bacterial pathogens, and genetic deficiencies in complement can render individuals highly susceptible to infection, for example, invasive meningococcal disease. In contrast, Gram-positive bacteria are inherently resistant to the direct bactericidal activity of complement due to their thick layer of cell wall peptidoglycan. However, complement also serves diverse roles in immune defense against all bacteria by flagging them for opsonization and killing by professional phagocytes, synergizing with neutrophils, modulating inflammatory responses, regulating T cell development, and cross talk with coagulation cascades. In this review, we discuss newly appreciated roles for complement beyond direct membrane lysis, incorporate nonlytic roles of complement into immunological paradigms of host-pathogen interactions, and identify bacterial strategies for complement evasion.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Bacterias Gramnegativas/inmunología , Bacterias Grampositivas/inmunología , Interacciones Huésped-Patógeno/inmunología , Receptores de Complemento/inmunología , Humanos , Neutrófilos/inmunología , Fagocitosis/inmunología , Transducción de Señal/inmunología
13.
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
14.
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
15.
Front Immunol ; 11: 583853, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33117397

RESUMEN

Genetic deficiency in C1q is a strong susceptibility factor for systemic lupus erythematosus (SLE). There are two major hypotheses that potentially explain the role of C1q in SLE. The first postulates that C1q deficiency abrogates apoptotic cell clearance, leading to persistently high loads of potentially immunogenic self-antigens that trigger autoimmune responses. While C1q undoubtedly plays an important role in apoptotic clearance, an essential biological process such as removal of self- waste is so critical for host survival that multiple ligand-receptor combinations do fortunately exist to ensure that proper disposal of apoptotic debris is accomplished even in the absence of C1q. The second hypothesis is based on the observation that locally synthesized C1q plays a critical role in regulating the earliest stages of monocyte to dendritic cell (DC) differentiation and function. Indeed, circulating C1q has been shown to keep monocytes in a pre-dendritic state by silencing key molecular players and ensuring that unwarranted DC-driven immune responses do not occur. Monocytes are also able to display macromolecular C1 on their surface, representing a novel mechanism for the recognition of circulating "danger." Translation of this danger signal in turn, provides the requisite "license" to trigger a differentiation pathway that leads to adaptive immune response. Based on this evidence, the second hypothesis proposes that deficiency in C1q dysregulates monocyte-to-DC differentiation and causes inefficient or defective maintenance of self-tolerance. The fact that C1q receptors (cC1qR and gC1qR) are also expressed on the surface of both monocytes and DCs, suggests that C1q/C1qR may regulate DC differentiation and function through specific cell-signaling pathways. While their primary ligand is C1q, C1qRs can also independently recognize a vast array of plasma proteins as well as pathogen-associated molecular ligands, indicating that these molecules may collaborate in antigen recognition and processing, and thus regulate DC-differentiation. This review will therefore focus on the role of C1q and C1qRs in SLE and explore the gC1qR/C1q axis as a potential target for therapy.


Asunto(s)
Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/terapia , Animales , Diferenciación Celular/inmunología , Complemento C1q/inmunología , Células Dendríticas/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Glicoproteínas de Membrana/inmunología , Monocitos/inmunología , Receptores de Complemento/inmunología , Transducción de Señal/inmunología
16.
Front Immunol ; 11: 1875, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32973774

RESUMEN

The pathogenesis of some kidney diseases is closely associated with complement activation, where the C3a/C3a receptor (C3aR) might play a crucial role. C3a/C3aR has dual roles and may exert anti-inflammatory or pro-inflammatory effects depending on different cell types and diseases. In the kidneys, C3aR is primarily expressed on the tubular epithelium and less in glomerular podocytes. C3aR expression is enhanced and the levels of C3a in the plasma and urine are increased in kidney diseases of several types, and are associated with disease progression and severity. The C3a/C3aR pathway facilitates the progression of glomerular and tubulointerstitial diseases, while it has opposite effects on urinary tract infections. Clinical trials targeting C3a/C3aR in kidney diseases are lacking. Here, we reviewed the studies on the C3a/C3aR pathway in kidney disease, with the aim of understanding in-depth its controversial roles and its potential therapeutic value.


Asunto(s)
Activación de Complemento/inmunología , Complemento C3a/inmunología , Enfermedades Renales/inmunología , Receptores de Complemento/inmunología , Animales , Humanos
17.
Front Immunol ; 11: 1066, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32582172

RESUMEN

Phagocytosis is a cellular process for ingesting and eliminating particles larger than 0.5 µm in diameter, including microorganisms, foreign substances, and apoptotic cells. Phagocytosis is found in many types of cells and it is, in consequence an essential process for tissue homeostasis. However, only specialized cells termed professional phagocytes accomplish phagocytosis with high efficiency. Macrophages, neutrophils, monocytes, dendritic cells, and osteoclasts are among these dedicated cells. These professional phagocytes express several phagocytic receptors that activate signaling pathways resulting in phagocytosis. The process of phagocytosis involves several phases: i) detection of the particle to be ingested, ii) activation of the internalization process, iii) formation of a specialized vacuole called phagosome, and iv) maturation of the phagosome to transform it into a phagolysosome. In this review, we present a general view of our current understanding on cells, phagocytic receptors and phases involved in phagocytosis.


Asunto(s)
Modelos Inmunológicos , Fagocitosis/inmunología , Apoptosis/inmunología , Humanos , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Fagocitos/inmunología , Fagocitos/fisiología , Fagocitosis/fisiología , Fagosomas/inmunología , Receptores de Complemento/inmunología , Receptores de IgG/inmunología , Receptores Inmunológicos/inmunología , Receptores de Reconocimiento de Patrones/inmunología , Transducción de Señal/inmunología
19.
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
20.
Mol Immunol ; 121: 99-110, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32199212

RESUMEN

The complement cascade consists of cell bound and serum proteins acting together to protect the host from pathogens, remove cancerous cells and effectively links innate and adaptive immune responses. Despite its usefulness in microbial neutralization and clearance of cancerous cells, excessive complement activation causes an immune imbalance and tissue damage in the host. Hence, a series of complement regulatory proteins present at a higher concentration in blood plasma and on cell surfaces tightly regulate the cascade. The complement cascade can be initiated by B-1 B cell production of natural antibodies. Natural antibodies arise spontaneously without any known exogenous antigenic or microbial stimulus and protect against invading pathogens, clear apoptotic cells, provide tissue homeostasis, and modulate adaptive immune functions. Natural IgM antibodies recognize microbial and cancer antigens and serve as an activator of complement mediated lysis. This review will discuss advances in complement activation and regulation in bacterial and viral infections, and cancer. We will also explore the crosstalk of natural antibodies with bacterial populations and cancer.


Asunto(s)
Infecciones Bacterianas/inmunología , Inmunidad Humoral , Inmunidad Innata , Neoplasias/inmunología , Virosis/inmunología , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Antígenos Virales/inmunología , Antígenos Virales/metabolismo , Apoptosis/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Activación de Complemento , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Humanos , Inmunoglobulina M/inmunología , Inmunoglobulina M/metabolismo , Receptores de Complemento/inmunología , Receptores de Complemento/metabolismo , Escape del Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...