Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.482
Filtrar
Más filtros

Intervalo de año de publicación
1.
Nat Commun ; 15(1): 5028, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38866748

RESUMEN

Cholesterol-dependent cytolysins (CDCs) comprise a large family of pore-forming toxins produced by Gram-positive bacteria, which are used to attack eukaryotic cells. Here, we functionally characterize a family of 2-component CDC-like (CDCL) toxins produced by the Gram-negative Bacteroidota that form pores by a mechanism only described for the mammalian complement membrane attack complex (MAC). We further show that the Bacteroides CDCLs are not eukaryotic cell toxins like the CDCs, but instead bind to and are proteolytically activated on the surface of closely related species, resulting in pore formation and cell death. The CDCL-producing Bacteroides is protected from the effects of its own CDCL by the presence of a surface lipoprotein that blocks CDCL pore formation. These studies suggest a prevalent mode of bacterial antagonism by a family of two-component CDCLs that function like mammalian MAC and that are wide-spread in the gut microbiota of diverse human populations.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento , Humanos , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Bacteroides/genética , Bacteroides/metabolismo , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Citotoxinas/metabolismo , Microbioma Gastrointestinal , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas del Sistema Complemento/metabolismo , Proteínas del Sistema Complemento/inmunología , Animales , Células Eucariotas/metabolismo
2.
Neurol Neuroimmunol Neuroinflamm ; 11(4): e200270, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38912898

RESUMEN

BACKGROUND AND OBJECTIVES: The complement system is known to play a role in multiple sclerosis (MS) pathogenesis. However, its contribution to disease progression remains elusive. The study investigated the role of the complement system in disability progression of patients with primary progressive MS (PPMS). METHODS: Sixty-eight patients with PPMS from 12 European MS centers were included in the study. Serum and CSF levels of a panel of complement components (CCs) were measured by multiplex enzyme-linked immunosorbent assay at a baseline time point (i.e., sampling). Mean (SD) follow-up time from baseline was 9.6 (4.8) years. Only one patient (1.5%) was treated during follow-up. Univariable and multivariable logistic regressions adjusted for age, sex, and albumin quotient were performed to assess the association between baseline CC levels and disability progression in short term (2 years), medium term (6 years), and long term (at the time of the last follow-up). RESULTS: In short term, CC played little or no role in disability progression. In medium term, an elevated serum C3a/C3 ratio was associated with a higher risk of disability progression (adjusted OR 2.30; 95% CI 1.17-6.03; p = 0.040). By contrast, increased CSF C1q levels were associated with a trend toward reduced risk of disability progression (adjusted OR 0.43; 95% CI 0.17-0.98; p = 0.054). Similarly, in long term, an elevated serum C3a/C3 ratio was associated with higher risk of disability progression (adjusted OR 1.81; 95% CI 1.09-3.40; p = 0.037), and increased CSF C1q levels predicted lower disability progression (adjusted OR 0.41; 95% CI 0.17-0.86; p = 0.025). DISCUSSION: Proteins involved in the activation of early complement cascades play a role in disability progression as risk (elevated serum C3a/C3 ratio) or protective (elevated CSF C1q) factors after 6 or more years of follow-up in patients with PPMS. The protective effects associated with C1q levels in CSF may be related to its neuroprotective and anti-inflammatory properties.


Asunto(s)
Progresión de la Enfermedad , Esclerosis Múltiple Crónica Progresiva , Humanos , Masculino , Femenino , Esclerosis Múltiple Crónica Progresiva/líquido cefalorraquídeo , Esclerosis Múltiple Crónica Progresiva/sangre , Esclerosis Múltiple Crónica Progresiva/fisiopatología , Persona de Mediana Edad , Adulto , Estudios de Seguimiento , Complemento C3/metabolismo , Complemento C3/análisis , Complemento C3a/metabolismo , Complemento C3a/líquido cefalorraquídeo , Evaluación de la Discapacidad , Proteínas del Sistema Complemento/líquido cefalorraquídeo , Proteínas del Sistema Complemento/metabolismo
3.
Front Immunol ; 15: 1357869, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38895123

RESUMEN

The complement system, an important part of the innate system, is known to play a central role in many immune mediated kidney diseases. All parts of the complement system including the classical, alternative, and mannose-binding lectin pathways have been implicated in complement-mediated kidney injury. Although complement components are thought to be mainly synthesized in the liver and activated in the circulation, emerging data suggest that complement is synthesized and activated inside the kidney leading to direct injury. Urinary complement biomarkers are likely a better reflection of inflammation within the kidneys as compared to traditional serum complement biomarkers which may be influenced by systemic inflammation. In addition, urinary complement biomarkers have the advantage of being non-invasive and easily accessible. With the rise of therapies targeting the complement pathways, there is a critical need to better understand the role of complement in kidney diseases and to develop reliable and non-invasive biomarkers to assess disease activity, predict treatment response and guide therapeutic interventions. In this review, we summarized the current knowledge on urinary complement biomarkers of kidney diseases due to immune complex deposition (lupus nephritis, primary membranous nephropathy, IgA nephropathy) and due to activation of the alternative pathway (C3 glomerulopathy, thrombotic microangiography, ANCA-associated vasculitis). We also address the limitations of current research and propose future directions for the discovery of urinary complement biomarkers.


Asunto(s)
Biomarcadores , Proteínas del Sistema Complemento , Enfermedades Renales , Humanos , Biomarcadores/orina , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/orina , Proteínas del Sistema Complemento/metabolismo , Enfermedades Renales/orina , Enfermedades Renales/inmunología , Enfermedades Renales/diagnóstico , Animales , Activación de Complemento
4.
Int J Mol Sci ; 25(11)2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38892429

RESUMEN

South Africa is the epicentre of the global HIV pandemic, with 13.9% of its population infected. Preeclampsia (PE), a hypertensive disorder of pregnancy, is often comorbid with HIV infection, leading to multi-organ dysfunction and convulsions. The exact pathophysiology of preeclampsia is triggered by an altered maternal immune response or defective development of maternal tolerance to the semi-allogenic foetus via the complement system. The complement system plays a vital role in the innate immune system, generating inflammation, mediating the clearance of microbes and injured tissue materials, and a mediator of adaptive immunity. Moreover, the complement system has a dual effect, of protecting the host against HIV infection and enhancing HIV infectivity. An upregulation of regulatory proteins has been implicated as an adaptive phenomenon in response to elevated complement-mediated cell lysis in HIV infection, further aggravated by preeclamptic complement activation. In light of the high prevalence of HIV infection and preeclampsia in South Africa, this review discusses the association of complement proteins and their role in the synergy of HIV infection and preeclampsia in South Africa. It aims to identify women at elevated risk, leading to early diagnosis and better management with targeted drug therapy, thereby improving the understanding of immunological dysregulation.


Asunto(s)
Proteínas del Sistema Complemento , Infecciones por VIH , Preeclampsia , Humanos , Preeclampsia/inmunología , Preeclampsia/epidemiología , Embarazo , Infecciones por VIH/complicaciones , Infecciones por VIH/inmunología , Femenino , Proteínas del Sistema Complemento/metabolismo , Proteínas del Sistema Complemento/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/epidemiología , Sudáfrica/epidemiología , Comorbilidad , Activación de Complemento
5.
PLoS One ; 19(6): e0305851, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38935768

RESUMEN

We investigated the interactions of unopsonized and opsonized Mycoplasma mycoides subsp. mycoides (Mmm) with bovine macrophages in vitro. Mmm survived and proliferated extracellularly on bovine macrophage cell layers in the absence of Mmm-specific antisera. Bovine complement used at non-bactericidal concentrations did neither have opsonizing effect nor promoted intracellular survival, whereas Mmm-specific antisera substantially increased phagocytosis and Mmm killing. A phagocytosis-independent uptake of Mmm by macrophages occurred at a high multiplicity of infection, also found to induce the production of TNF, and both responses were unaffected by non-bactericidal doses of bovine complement. Bovine complement used at higher doses killed Mmm in cell-free cultures and completely abrogated TNF responses by macrophages. These results provide a framework to identify Mmm antigens involved in interactions with macrophages and targeted by potentially protective antibodies and point towards a pivotal role of complement in the control of inflammatory responses in contagious bovine pleuropneumonia.


Asunto(s)
Macrófagos , Fagocitosis , Animales , Bovinos , Macrófagos/microbiología , Macrófagos/inmunología , Macrófagos/metabolismo , Proteínas del Sistema Complemento/metabolismo , Proteínas del Sistema Complemento/inmunología , Mycoplasma/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Pleuroneumonía Contagiosa/microbiología , Pleuroneumonía Contagiosa/inmunología , Mycoplasma mycoides/inmunología
6.
Front Immunol ; 15: 1422370, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38938578

RESUMEN

Introduction: Hematopoietic stem cell transplantation (HSCT) is associated with immune complications and endothelial dysfunction due to intricate donor-recipient interactions, conditioning regimens, and inflammatory responses. Methods: This study investigated the role of the complement system during HSCT and its interaction with the cytokine network. Seventeen acute myeloid leukemia patients undergoing HSCT were monitored, including blood sampling from the start of the conditioning regimen until four weeks post-transplant. Clinical follow-up was 200 days. Results: Total complement functional activity was measured by WIELISA and the degree of complement activation by ELISA measurement of sC5b-9. Cytokine release was measured using a 27-multiplex immuno-assay. At all time-points during HSCT complement functional activity remained comparable to healthy controls. Complement activation was continuously stable except for two patients demonstrating increased activation, consistent with severe endotheliopathy and infections. In vitro experiments with post-HSCT whole blood challenged with Escherichia coli, revealed a hyperinflammatory cytokine response with increased TNF, IL-1ß, IL-6 and IL-8 formation. Complement C3 inhibition markedly reduced the cytokine response induced by Staphylococcus aureus, Aspergillus fumigatus, and cholesterol crystals. Discussion: In conclusion, HSCT patients generally retained a fully functional complement system, whereas activation occurred in patients with severe complications. The complement-cytokine interaction indicates the potential for new complement-targeting therapeutic strategies in HSCT.


Asunto(s)
Activación de Complemento , Citocinas , Trasplante de Células Madre Hematopoyéticas , Trasplante Homólogo , Humanos , Masculino , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Femenino , Persona de Mediana Edad , Adulto , Citocinas/metabolismo , Anciano , Leucemia Mieloide Aguda/terapia , Leucemia Mieloide Aguda/inmunología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Acondicionamiento Pretrasplante/métodos , Adulto Joven
7.
Microb Pathog ; 192: 106682, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38750776

RESUMEN

Porcine reproductive and respiratory syndrome virus (PRRSV) causes a highly transmissible disease of significant concern in the pig industry. Previous studies have demonstrated that the XM-2020 strain (a lineage 1.8 PRRSV IA/2012/NADC30) can induce special hemorrhagic injury in the small intestines. However, the specific mechanism underlying this injurious effect remains incompletely understood. In this study, we examined the pathogenic properties of XM-2020 and YC-2020 strains (a lineage 1.5 PRRSV IA/2014/NADC34) in piglets. Animal pathogenic tests revealed that with either Lineage 1 PRRSVs strains XM-2020 or YC-2020 demonstrated pronounced intestinal hemorrhage and suppression of peripheral immunological organs, comparing to JXA1 infection. Transcriptome analysis of diseased small intestines unveiled that PRRSV infection stimulated oxidative and inflammatory reactions. Remarkably, we also observed activation of the complement system alongside a notable down-regulation of complement and coagulation cascade pathways in the Lineage 1 PRRSVs infection group. Based on these findings, we propose that the primary mechanism driving the hemorrhagic injury of the small intestine caused by Lineage 1 PRRSVs is the suppression of complement and coagulation cascades resulting from immunosuppression. This discovery deepens our understanding of the pathogenicity of PRRSV in the small intestine and provides promising ways out for the development of innovative strategies aimed at controlling PRRSV.


Asunto(s)
Proteínas del Sistema Complemento , Síndrome Respiratorio y de la Reproducción Porcina , Virus del Síndrome Respiratorio y Reproductivo Porcino , Animales , Porcinos , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Virus del Síndrome Respiratorio y Reproductivo Porcino/patogenicidad , Síndrome Respiratorio y de la Reproducción Porcina/virología , Síndrome Respiratorio y de la Reproducción Porcina/patología , Coagulación Sanguínea , Intestino Delgado/virología , Intestino Delgado/patología , Intestinos/virología , Intestinos/patología , Perfilación de la Expresión Génica , Hemorragia
8.
J Clin Invest ; 134(9)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38690727

RESUMEN

Careful regulation of the complement system is critical for enabling complement proteins to titrate immune defense while also preventing collateral tissue damage from poorly controlled inflammation. In the eye, this balance between complement activity and inhibition is crucial, as a low level of basal complement activity is necessary to support ocular immune privilege, a prerequisite for maintaining vision. Dysregulated complement activation contributes to parainflammation, a low level of inflammation triggered by cellular damage that functions to reestablish homeostasis, or outright inflammation that disrupts the visual axis. Complement dysregulation has been implicated in many ocular diseases, including glaucoma, diabetic retinopathy, and age-related macular degeneration (AMD). In the last two decades, complement activity has been the focus of intense investigation in AMD pathogenesis, leading to the development of novel therapeutics for the treatment of atrophic AMD. This Review outlines recent advances and challenges, highlighting therapeutic approaches that have advanced to clinical trials, as well as providing a general overview of the complement system in the posterior segment of the eye and selected ocular diseases.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento , Degeneración Macular , Humanos , Degeneración Macular/inmunología , Degeneración Macular/patología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Activación de Complemento/inmunología , Animales , Ojo/inmunología , Ojo/patología
9.
Expert Rev Hematol ; 17(4-5): 107-116, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38708453

RESUMEN

INTRODUCTION: Bystander hemolysis occurs when antigen-negative red blood cells (RBCs) are lysed by the complement system. Many clinical entities including passenger lymphocyte syndrome, hyperhemolysis following blood transfusion, and paroxysmal nocturnal hemoglobinuria are complicated by bystander hemolysis. AREAS COVERED: The review provides data about the role of the complement system in the pathogenesis of bystander hemolysis. Moreover, future perspectives on the understanding and management of this syndrome are described. EXPERT OPINION: Complement system can be activated via classical, alternative, and lectin pathways. Classical pathway activation is mediated by antigen-antibody (autoantibodies and alloantibodies against autologous RBCs, infectious agents) complexes. Alternative pathway initiation is triggered by heme, RBC microvesicles, and endothelial injury that is a result of intravascular hemolysis. Thus, C5b is formed, binds with C6-C9 compomers, and MAC (C5b-9) is formulated in bystander RBCs membranes, leading to cell lysis. Intravascular hemolysis, results in activation of the alternative pathway, establishing a vicious cycle between complement activation and bystander hemolysis. C5 inhibitors have been used effectively in patients with hyperhemolysis syndrome and other entities characterized by bystander hemolysis.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento , Eritrocitos , Hemólisis , Humanos , Hemólisis/inmunología , Eritrocitos/inmunología , Eritrocitos/metabolismo , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Efecto Espectador , Hemoglobinuria Paroxística/inmunología , Hemoglobinuria Paroxística/terapia
10.
Int J Mol Sci ; 25(10)2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38791517

RESUMEN

Maternal immune activation (MIA) is a risk factor for multiple neurodevelopmental disorders; however, animal models developed to explore MIA mechanisms are sensitive to experimental factors, which has led to complexity in previous reports of the MIA phenotype. We sought to characterize an MIA protocol throughout development to understand how prenatal immune insult alters the trajectory of important neurodevelopmental processes, including the microglial regulation of synaptic spines and complement signaling. We used polyinosinic:polycytidylic acid (polyI:C) to induce MIA on gestational day 9.5 in CD-1 mice, and measured their synaptic spine density, microglial synaptic pruning, and complement protein expression. We found reduced dendritic spine density in the somatosensory cortex starting at 3-weeks-of-age with requisite increases in microglial synaptic pruning and phagocytosis, suggesting spine density loss was caused by increased microglial synaptic pruning. Additionally, we showed dysregulation in complement protein expression persisting into adulthood. Our findings highlight disruptions in the prenatal environment leading to alterations in multiple dynamic processes through to postnatal development. This could potentially suggest developmental time points during which synaptic processes could be measured as risk factors or targeted with therapeutics for neurodevelopmental disorders.


Asunto(s)
Proteínas del Sistema Complemento , Espinas Dendríticas , Microglía , Poli I-C , Animales , Microglía/metabolismo , Microglía/efectos de los fármacos , Microglía/inmunología , Ratones , Femenino , Embarazo , Espinas Dendríticas/metabolismo , Poli I-C/farmacología , Proteínas del Sistema Complemento/metabolismo , Proteínas del Sistema Complemento/inmunología , Efectos Tardíos de la Exposición Prenatal , Fagocitosis , Modelos Animales de Enfermedad , Corteza Somatosensorial/efectos de los fármacos , Corteza Somatosensorial/metabolismo , Sinapsis/metabolismo , Sinapsis/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos
11.
Front Immunol ; 15: 1390468, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38726006

RESUMEN

Introduction: Relapsing fever (RF) remains a neglected human disease that is caused by a number of diverse pathogenic Borrelia (B.) species. Characterized by high cell densities in human blood, relapsing fever spirochetes have developed plentiful strategies to avoid recognition by the host defense mechanisms. In this scenario, spirochetal lipoproteins exhibiting multifunctional binding properties in the interaction with host-derived molecules are known to play a key role in adhesion, fibrinolysis and complement activation. Methods: Binding of CihC/FbpC orthologs to different human proteins and conversion of protein-bound plasminogen to proteolytic active plasmin were examined by ELISA. To analyze the inhibitory capacity of CihC/FbpC orthologs on complement activation, a microtiter-based approach was performed. Finally, AlphaFold predictions were utilized to identified the complement-interacting residues. Results and discussion: Here, we elucidate the binding properties of CihC/FbpC-orthologs from distinct RF spirochetes including B. parkeri, B. hermsii, B. turicatae, and B. recurrentis to human fibronectin, plasminogen, and complement component C1r. All CihC/FbpC-orthologs displayed similar binding properties to fibronectin, plasminogen, and C1r, respectively. Functional studies revealed a dose dependent binding of plasminogen to all borrelial proteins and conversion to active plasmin. The proteolytic activity of plasmin was almost completely abrogated by tranexamic acid, indicating that lysine residues are involved in the interaction with this serine protease. In addition, a strong inactivation capacity toward the classical pathway could be demonstrated for the wild-type CihC/FbpC-orthologs as well as for the C-terminal CihC fragment of B. recurrentis. Pre-incubation of human serum with borrelial molecules except CihC/FbpC variants lacking the C-terminal region protected serum-susceptible Borrelia cells from complement-mediated lysis. Utilizing AlphaFold2 predictions and existing crystal structures, we mapped the putative key residues involved in C1r binding on the CihC/FbpC orthologs attempting to explain the relatively small differences in C1r binding affinity despite the substitutions of key residues. Collectively, our data advance the understanding of the multiple binding properties of structural and functional highly similar molecules of relapsing fever spirochetes proposed to be involved in pathogenesis and virulence.


Asunto(s)
Proteínas Bacterianas , Borrelia , Fibrinólisis , Interacciones Huésped-Patógeno , Plasminógeno , Humanos , Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Borrelia/inmunología , Borrelia/metabolismo , Activación de Complemento , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Fibrinolisina/metabolismo , Fibronectinas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Evasión Inmune , Plasminógeno/metabolismo , Unión Proteica , Fiebre Recurrente/inmunología , Fiebre Recurrente/microbiología
12.
J Innate Immun ; 16(1): 337-353, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38815564

RESUMEN

BACKGROUND: Sepsis-associated coagulopathy specifically refers to widespread systemic coagulation activation accompanied by a high risk of hemorrhage and organ damage, which in severe cases manifests as disseminated intravascular coagulation (DIC), or even develops into multiple organ dysfunction syndrome (MODS). The complement system and the coagulation system as the main columns of innate immunity and hemostasis, respectively, undergo substantial activation after sepsis. SUMMARY: Dysfunction of the complement, coagulation/fibrinolytic cascades caused by sepsis leads to "thromboinflammation," which ultimately amplifies the systemic inflammatory response and accelerates the development of MODS. Recent studies have revealed that massive activation of the complement system exacerbates sepsis-induced coagulation and even results in DIC, which suggests that inhibition of complement activation may have therapeutic potential in the treatment of septic coagulopathy. KEY MESSAGES: Sepsis-associated thrombosis involves the upregulation or activation of procoagulant factors, down-regulation or inactivation of anticoagulant factors, and impairment of the fibrinolytic mechanism. This review aims to summarize the latest literature and analyze the underlying molecular mechanisms of the activation of the complement system on the abnormal coagulation cascades in sepsis.


Asunto(s)
Activación de Complemento , Sepsis , Humanos , Sepsis/inmunología , Activación de Complemento/inmunología , Animales , Coagulación Sanguínea , Coagulación Intravascular Diseminada/inmunología , Coagulación Intravascular Diseminada/etiología , Inmunidad Innata , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Insuficiencia Multiorgánica/inmunología , Insuficiencia Multiorgánica/etiología , Fibrinólisis , Trastornos de la Coagulación Sanguínea/inmunología , Trastornos de la Coagulación Sanguínea/etiología , Trombosis/inmunología , Trombosis/etiología
13.
Clin Immunol ; 263: 110232, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38701960

RESUMEN

IgA nephropathy (IgAN), which has been confirmed as a complement mediated autoimmune disease, is also one form of glomerulonephritis associated with COVID-19. Here, we aim to investigate the clinical and immunological characteristics of patients with IgAN after COVID-19. The level of plasma level of C5a (p < 0.001), soluble C5b-9 (p = 0.018), FHR5 (p < 0.001) were all significantly higher in Group CoV (33 patients with renal biopsy-proven IgAN experienced COVID-19) compared with Group non-CoV (44 patients with IgAN without COVID-19), respectively. Compared with Group non-CoV, the intensity of glomerular C4d (p = 0.017) and MAC deposition (p < 0.001) and Gd-IgA1 deposition (p = 0.005) were much stronger in Group CoV. Our finding revealed that for IgAN after COVID-19, mucosal immune responses to SARS-CoV-2 infection may result in the overactivation of systemic and renal local complement system, and increased glomerular deposition of Gd-IgA1, which may lead to renal dysfunction and promote renal progression in IgAN patients.


Asunto(s)
COVID-19 , Glomerulonefritis por IGA , SARS-CoV-2 , Humanos , Glomerulonefritis por IGA/inmunología , Glomerulonefritis por IGA/sangre , COVID-19/inmunología , COVID-19/complicaciones , Femenino , Masculino , Adulto , SARS-CoV-2/inmunología , Persona de Mediana Edad , Activación de Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Glomérulos Renales/patología , Glomérulos Renales/inmunología , Complemento C5a/inmunología , Complemento C5a/metabolismo
14.
Front Immunol ; 15: 1288597, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38817607

RESUMEN

Complement activation protects against infection but also contributes to pathological mechanisms in a range of clinical conditions such as autoimmune diseases and transplant rejection. Complement-inhibitory drugs, either approved or in development, usually act systemically, thereby increasing the risk for infections. We therefore envisioned a novel class of bispecific antibodies (bsAbs) which are capable of site-directed complement inhibition by bringing endogenous complement regulators in the vicinity of defined cell surface antigens. Here, we analyzed a comprehensive set of obligate bsAbs designed to crosslink a specific target with either complement regulator factor H (FH) or C4b-binding protein (C4BP). The bsAbs were assessed for their capacity to inhibit complement activation and cell lysis in an antigen-targeted manner. We observed that the bsAbs inhibited classical, lectin, and alternative pathway complement activation in which sufficient endogenous serum FH and C4BP could be recruited to achieve local inhibition. Importantly, the bsAbs effectively protected antigen-positive liposomes, erythrocytes, and human leukocytes from complement-mediated lysis. In conclusion, localized complement inhibition by bsAbs capable of recruiting endogenous human complement regulators (such as FH or C4BP) to cell surfaces potentially provides a novel therapeutic approach for the targeted treatment of complement-mediated diseases.


Asunto(s)
Anticuerpos Biespecíficos , Activación de Complemento , Proteína de Unión al Complemento C4b , Factor H de Complemento , Anticuerpos Biespecíficos/inmunología , Anticuerpos Biespecíficos/farmacología , Humanos , Activación de Complemento/inmunología , Proteína de Unión al Complemento C4b/inmunología , Proteína de Unión al Complemento C4b/metabolismo , Factor H de Complemento/inmunología , Factor H de Complemento/metabolismo , Antígenos/inmunología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Unión Proteica
15.
Med Microbiol Immunol ; 213(1): 8, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38767707

RESUMEN

Bacterial resistance to serum is a key virulence factor for the development of systemic infections. The amount of lipopolysaccharide (LPS) and the O-antigen chain length distribution on the outer membrane, predispose Salmonella to escape complement-mediated killing. In Salmonella enterica serovar Enteritidis (S. Enteritidis) a modal distribution of the LPS O-antigen length can be observed. It is characterized by the presence of distinct fractions: low molecular weight LPS, long LPS and very long LPS. In the present work, we investigated the effect of the O-antigen modal length composition of LPS molecules on the surface of S. Enteritidis cells on its ability to evade host complement responses. Therefore, we examined systematically, by using specific deletion mutants, roles of different O-antigen fractions in complement evasion. We developed a method to analyze the average LPS lengths and investigated the interaction of the bacteria and isolated LPS molecules with complement components. Additionally, we assessed the aspect of LPS O-antigen chain length distribution in S. Enteritidis virulence in vivo in the Galleria mellonella infection model. The obtained results of the measurements of the average LPS length confirmed that the method is suitable for measuring the average LPS length in bacterial cells as well as isolated LPS molecules and allows the comparison between strains. In contrast to earlier studies we have used much more precise methodology to assess the LPS molecules average length and modal distribution, also conducted more subtle analysis of complement system activation by lipopolysaccharides of various molecular mass. Data obtained in the complement activation assays clearly demonstrated that S. Enteritidis bacteria require LPS with long O-antigen to resist the complement system and to survive in the G. mellonella infection model.


Asunto(s)
Proteínas del Sistema Complemento , Modelos Animales de Enfermedad , Lipopolisacáridos , Antígenos O , Salmonella enteritidis , Salmonella enteritidis/inmunología , Salmonella enteritidis/patogenicidad , Animales , Antígenos O/inmunología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Lipopolisacáridos/inmunología , Evasión Inmune , Viabilidad Microbiana , Mariposas Nocturnas/microbiología , Mariposas Nocturnas/inmunología , Virulencia , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/microbiología , Salmonelosis Animal/inmunología , Salmonelosis Animal/microbiología , Activación de Complemento , Lepidópteros/inmunología , Lepidópteros/microbiología
16.
RMD Open ; 10(2)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38749532

RESUMEN

OBJECTIVES: To investigate lectin pathway proteins (LPPs) as biomarkers for axial spondyloarthritis (axSpA) in a cross-sectional cohort with a suspicion of axSpA, comprising newly diagnosed axSpA and chronic low back pain (cLBP) individuals. METHODS: Serum samples from 515 participants within the OptiRef cohort, including 151 axSpA patients and 364 cLBP patients, were measured using immunoassays for LPPs (mannan-binding lectin (MBL), collectin liver-1 (CL-L1), M-ficolin, H-ficolin and L-ficolin, MBL-associated serine proteases (MASP)-1, -2 and -3, MBL-associated proteins (MAp19 and MAp44) and the complement activation product C3dg). RESULTS: Serum levels of L-ficolin, MASP-2 and C3dg were elevated in axSpA patients, whereas levels of MASP-3 and CL-L1 were decreased, and this remained significant for C3dg and MASP-3 after adjustment for C reactive protein (CRP). A univariate regression analysis showed serum levels of CL-L1, MASP-2, MASP-3 and C3dg to predict the diagnosis of axSpA, and MASP-3 and C3dg remained significant in a multivariate logistic regression analysis. Assessment of the diagnostic potential showed that a combination of human leukocyte antigen B27 (HLA-B27) and measurements of L-ficolin, MASP-3 and C3dg increased the diagnostic specificity for axSpA, however, with a concomitant loss of sensitivity. CONCLUSIONS: Serum levels of complement activation, that is, C3dg, and MASP-3 differed significantly between axSpA and cLBP patients after adjustment for CRP. Although combining HLA-B27 with measurements of L-ficolin, MASP-3 and C3dg increased the diagnostic specificity for axSpA, this seems unjustified due to the concomitant loss of sensitivity. However, both C3dg and MASP-3 were associated with axSpA diagnosis in multivariate logistic regression, suggesting an involvement of complement in the inflammatory processes and possibly pathogenesis in axSpA.


Asunto(s)
Espondiloartritis Axial , Biomarcadores , Proteínas del Sistema Complemento , Humanos , Biomarcadores/sangre , Masculino , Femenino , Adulto , Persona de Mediana Edad , Estudios Transversales , Proteínas del Sistema Complemento/metabolismo , Proteínas del Sistema Complemento/análisis , Espondiloartritis Axial/diagnóstico , Espondiloartritis Axial/sangre , Espondiloartritis Axial/etiología , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/metabolismo , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/análisis , Lectinas/sangre , Activación de Complemento
17.
Pathologie (Heidelb) ; 45(4): 246-253, 2024 Jul.
Artículo en Alemán | MEDLINE | ID: mdl-38578365

RESUMEN

The complement cascade comprises a variety of soluble and cell surface proteins and is an important component of the innate immune system. When the cascade is triggered by any of the three activation pathways, the complement system rapidly produces large amounts of protein fragments that are potent mediators of inflammatory, vasoactive, and metabolic responses. All activation pathways lead to the terminal complement cascade with the formation of the membrane attack complex, which lyses cells by forming membrane pores. Although the complement system is essential for pathogen defense and homeostasis, excessive or uncontrolled activation can lead to tissue damage. Recent research shows that the complement system is activated in almost all kidney diseases, even those not traditionally considered immune-mediated. In directly complement-mediated kidney diseases, complement factors or regulators are defective, afunctional or inactivated by antibodies. In many other renal diseases, the complement system is activated secondarily as a result of renal damage and is therefore involved in the pathogenesis of the disease, but is not the trigger. The detection of complement deposits is also used to diagnose kidney disease. This review describes the structure of the complement system and the effects of its dysregulation as a cause and modulator of renal disease.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento , Enfermedades Renales , Humanos , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Activación de Complemento/inmunología , Enfermedades Renales/inmunología , Enfermedades Renales/patología , Enfermedades Renales/metabolismo , Riñón/patología , Riñón/inmunología , Riñón/metabolismo , Inmunidad Innata , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/inmunología
18.
Nanomedicine (Lond) ; 19(11): 995-1012, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38593053

RESUMEN

Aim: To investigate the influence of fluorine in reducing the adsorption of immune-reactive proteins onto PEGylated gold nanoparticles. Methods: Reversible addition fragmentation chain transfer polymerization, the Turkevich method and ligand exchange were used to prepare polymer-coated gold nanoparticles. Subsequent in vitro physicochemical and biological characterizations and proteomic analysis were performed. Results: Fluorine-modified polymers reduced the adsorption of complement and other immune-reactive proteins while potentially improving circulatory times and modulating liver toxicity by reducing apolipoprotein E adsorption. Fluorine actively discouraged phagocytosis while encouraging the adsorption of therapeutic targets, CD209 and signaling molecule calreticulin. Conclusion: This study suggests that the addition of fluorine in the surface coating of nanoparticles could lead to improved performance in nanomedicine designed for the intravenous delivery of cargos.


Nanomedicines are based around the delivery of therapies by tiny, nanosized delivery vehicles. This method offers a much better way of specifically targeting life-threatening diseases. For fast delivery, nanomedicines can be injected into the blood (intravenously); however, this often leads to an unwanted and exaggerated immune response. The immune system is activated by proteins in the blood that attach themselves to nanoparticles through various chemical interactions (the protein corona effect). Fluorine is a chemical routinely used in surfactants such as firefighting foam and more recently in molecular imaging and nanoparticles designed for the delivery of therapies aimed at cancer. While fluorine has great potential to improve the cellular uptake of therapies, little is known about whether it can also help camouflage the nanoparticles against the immune system responses. Here, using fluorinated polymer-coated gold nanoparticles, the authors demonstrate that fluorine reduces uptake by immune cells and is highly effective at reducing the binding of immune system-initiating proteins. This work successfully illustrates the rationale for more widespread investigation of fluorine during the development of polymer-coated nanoparticles designed for the intravenous delivery of nanomedicines.


Asunto(s)
Flúor , Oro , Nanopartículas del Metal , Polietilenglicoles , Oro/química , Nanopartículas del Metal/química , Flúor/química , Adsorción , Polietilenglicoles/química , Humanos , Polímeros/química , Fagocitosis/efectos de los fármacos , Animales , Propiedades de Superficie , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Ratones
19.
Trends Immunol ; 45(6): 397-399, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38637200

RESUMEN

The persistence or recurrence of symptoms after acute SARS-CoV-2 infection, termed 'long COVID', presents a formidable challenge to global healthcare systems. Recent research by Cervia-Hasler and colleagues delves into the intricate immunological landscape in patients with long COVID, demonstrating an interplay between complement and coagulation, driven by antiviral antibodies and tissue damage.


Asunto(s)
COVID-19 , Proteínas del Sistema Complemento , SARS-CoV-2 , Humanos , COVID-19/inmunología , SARS-CoV-2/inmunología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Tromboinflamación/inmunología , Coagulación Sanguínea/inmunología , Síndrome Post Agudo de COVID-19 , Activación de Complemento/inmunología , Anticuerpos Antivirales/inmunología
20.
mBio ; 15(5): e0011924, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38587424

RESUMEN

Gonorrhea, caused by the bacterium Neisseria gonorrhoeae (Gc), is characterized by neutrophilic influx to infection sites. Gc has developed mechanisms to resist killing by neutrophils that include modifications to its surface lipooligosaccharide (LOS). One such LOS modification is sialylation: Gc sialylates its terminal LOS sugars with cytidine-5'-monophosphate-N-acetylneuraminic acid, which is scavenged from the host using LOS sialyltransferase (Lst) since Gc cannot make its sialic acid. Sialylation enables sensitive strains of Gc to resist complement-mediated killing in a serum-dependent manner. However, little is known about the contribution of sialylation to complement-independent, direct Gc-neutrophil interactions. In the absence of complement, we found sialylated Gc expressing opacity-associated (Opa) proteins decreased the oxidative burst and granule exocytosis from primary human neutrophils. In addition, sialylated Opa+ Gc survived better than vehicle treated or Δlst Gc when challenged with neutrophils. However, Gc sialylation did not significantly affect Opa-dependent association with or internalization of Gc by neutrophils. Previous studies have implicated sialic acid-binding immunoglobulin-type lectins (Siglecs) in modulating neutrophil interactions with sialylated Gc. Blocking neutrophil Siglecs with antibodies that bind to their extracellular domains eliminated the ability of sialylated Opa+ Gc to suppress the oxidative burst and resist neutrophil killing. These findings highlight a new role for sialylation in Gc evasion of human innate immunity, with implications for the development of vaccines and therapeutics for gonorrhea. IMPORTANCE: Neisseria gonorrhoeae, the bacterium that causes gonorrhea, is an urgent global health concern due to increasing infection rates, widespread antibiotic resistance, and its ability to thwart protective immune responses. The mechanisms by which Gc subverts protective immune responses remain poorly characterized. One way N. gonorrhoeae evades human immunity is by adding sialic acid that is scavenged from the host onto its lipooligosaccharide, using the sialyltransferase Lst. Here, we found that sialylation enhances N. gonorrhoeae survival from neutrophil assault and inhibits neutrophil activation, independently of the complement system. Our results implicate bacterial binding of sialic acid-binding lectins (Siglecs) on the neutrophil surface, which dampens neutrophil antimicrobial responses. This work identifies a new role for sialylation in protecting N. gonorrhoeae from cellular innate immunity, which can be targeted to enhance the human immune response in gonorrhea.


Asunto(s)
Gonorrea , Ácido N-Acetilneuramínico , Neisseria gonorrhoeae , Activación Neutrófila , Neutrófilos , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico , Neisseria gonorrhoeae/inmunología , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/metabolismo , Humanos , Ácido N-Acetilneuramínico/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/genética , Gonorrea/inmunología , Gonorrea/microbiología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Lipopolisacáridos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas de la Membrana Bacteriana Externa/genética , Estallido Respiratorio , Interacciones Huésped-Patógeno/inmunología , Evasión Inmune
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA