Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 11.542
Filtrar
1.
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 , Plasminógeno , Unión Proteica , Fiebre Recurrente , Humanos , Borrelia/inmunología , Borrelia/metabolismo , Fiebre Recurrente/microbiología , Fiebre Recurrente/inmunología , Fiebre Recurrente/metabolismo , Plasminógeno/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Activación de Complemento , Evasión Inmune , Adhesión Bacteriana , Interacciones Huésped-Patógeno/inmunología , Fibronectinas/metabolismo , Fibrinolisina/metabolismo , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo
2.
Front Immunol ; 15: 1387808, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38745656

RESUMEN

Background: Tuberculous meningitis (TBM) is a devastating form of tuberculosis (TB) causing high mortality and disability. TBM arises due to immune dysregulation, but the underlying immune mechanisms are unclear. Methods: We performed single-cell RNA sequencing on peripheral blood mononuclear cells (PBMCs) and cerebrospinal fluid (CSF) cells isolated from children (n=6) with TBM using 10 xGenomics platform. We used unsupervised clustering of cells and cluster visualization based on the gene expression profiles, and validated the protein and cytokines by ELISA analysis. Results: We revealed for the first time 33 monocyte populations across the CSF cells and PBMCs of children with TBM. Within these populations, we saw that CD4_C04 cells with Th17 and Th1 phenotypes and Macro_C01 cells with a microglia phenotype, were enriched in the CSF. Lineage tracking analysis of monocyte populations revealed myeloid cell populations, as well as subsets of CD4 and CD8 T-cell populations with distinct effector functions. Importantly, we discovered that complement-activated microglial Macro_C01 cells are associated with a neuroinflammatory response that leads to persistent meningitis. Consistently, we saw an increase in complement protein (C1Q), inflammatory markers (CRP) and inflammatory factor (TNF-α and IL-6) in CSF cells but not blood. Finally, we inferred that Macro_C01 cells recruit CD4_C04 cells through CXCL16/CXCR6. Discussion: We proposed that the microglial Macro_C01 subset activates complement and interacts with the CD4_C04 cell subset to amplify inflammatory signals, which could potentially contribute to augment inflammatory signals, resulting in hyperinflammation and an immune response elicited by Mtb-infected tissues.


Asunto(s)
Microglía , Análisis de la Célula Individual , Transcriptoma , Tuberculosis Meníngea , Humanos , Tuberculosis Meníngea/inmunología , Microglía/inmunología , Microglía/metabolismo , Niño , Masculino , Femenino , Preescolar , Citocinas/metabolismo , Activación de Complemento/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Perfilación de la Expresión Génica , Mycobacterium tuberculosis/inmunología
3.
J Am Chem Soc ; 146(19): 13455-13466, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38703132

RESUMEN

The classical complement pathway is activated by antigen-bound IgG antibodies. Monomeric IgG must oligomerize to activate complement via the hexameric C1q complex, and hexamerizing mutants of IgG appear as promising therapeutic candidates. However, structural data have shown that it is not necessary to bind all six C1q arms to initiate complement, revealing a symmetry mismatch between C1 and the hexameric IgG complex that has not been adequately explained. Here, we use DNA nanotechnology to produce specific nanostructures to template antigens and thereby spatially control IgG valency. These DNA-nanotemplated IgG complexes can activate complement on cell-mimetic lipid membranes, which enabled us to determine the effect of IgG valency on complement activation without the requirement to mutate antibodies. We investigated this using biophysical assays together with 3D cryo-electron tomography. Our data revealed the importance of interantigen distance on antibody-mediated complement activation, and that the cleavage of complement component C4 by the C1 complex is proportional to the number of ideally spaced antigens. Increased IgG valency also translated to better terminal pathway activation and membrane attack complex formation. Together, these data provide insights into how nanopatterning antigen-antibody complexes influence the activation of the C1 complex and suggest routes to modulate complement activation by antibody engineering. Furthermore, to our knowledge, this is the first time DNA nanotechnology has been used to study the activation of the complement system.


Asunto(s)
Activación de Complemento , ADN , Inmunoglobulina G , Nanoestructuras , Nanoestructuras/química , Humanos , ADN/química , ADN/inmunología , Inmunoglobulina G/química , Inmunoglobulina G/inmunología , Complejo Antígeno-Anticuerpo/química , Complejo Antígeno-Anticuerpo/inmunología
4.
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
5.
Front Immunol ; 15: 1337215, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38715618

RESUMEN

Background: Mortalin/GRP75 is a ubiquitous mitochondrial chaperone related to the cytosolic heat shock protein 70. It protects cells from various types of damages and from senescence. Our goal was to determine whether COVID-19 patients have circulating mortalin in their blood and to assess its prognostic value in anticipating disease severity. Methods: Mortalin was determined by ELISA in the sera of 83 COVID-19 patients enrolled in the study. Patients were categorized into 4 groups: critical patients who died (FATAL) or required intensive care and survived (ICU), patients of mild severity (hospitalized but not critical) who required nasal oxygen support (HOSP+O2), and patients who did not need oxygen therapy (HOSP). Results: The mortalin concentration in the serum of all COVID-19 patients in the cohort was 194-2324 pg/mL. A comparison of the mortalin levels by peak severity among the various patient groups showed a highly significant difference between the HOSP and FATAL groups and a significant difference between the HOSP and the ICU groups. COVID-19 patients who eventually failed to survive had at hospitalization a markedly higher level of mortalin in their sera. Cox regression analysis revealed a high mortality hazard (HR=3.96, p<0.01) in patients with high mortalin circulating levels (above the median, ≥651 pg/mL). This was confirmed in survival curve analysis (Kaplan-Meier; p=0.0032, log-rank test). Mortalin remained an independent predictor of mortality even after adjusting for age and sex or various complement activation products. Complement activation data collected in an earlier study in the same cohort was compared regarding the mortalin levels. Patients with higher circulating mortalin levels also had higher levels of complement C3a but reduced levels of properdin. Discussion: This is the first report on circulating mortalin in COVID-19 patients. Higher mortalin levels were associated with more severe illnesses and a higher risk of death. We claim that quantifying the blood levels of mortalin and activated complement proteins will provide important information on the prognosis of COVID-19 patients and will serve as a useful tool for guiding their clinical management and treatment.


Asunto(s)
COVID-19 , Proteínas HSP70 de Choque Térmico , SARS-CoV-2 , Humanos , COVID-19/mortalidad , COVID-19/sangre , COVID-19/inmunología , Proteínas HSP70 de Choque Térmico/sangre , Masculino , Femenino , Persona de Mediana Edad , SARS-CoV-2/fisiología , SARS-CoV-2/inmunología , Anciano , Pronóstico , Índice de Severidad de la Enfermedad , Adulto , Biomarcadores/sangre , Activación de Complemento , Anciano de 80 o más Años
6.
Front Immunol ; 15: 1404752, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38690267

RESUMEN

Helminths produce calreticulin (CRT) to immunomodulate the host immune system as a survival strategy. However, the structure of helminth-derived CRT and the structural basis of the immune evasion process remains unclarified. Previous study found that the tissue-dwelling helminth Trichinella spiralis produces calreticulin (TsCRT), which binds C1q to inhibit activation of the complement classical pathway. Here, we used x-ray crystallography to resolve the structure of truncated TsCRT (TsCRTΔ), the first structure of helminth-derived CRT. TsCRTΔ was observed to share the same binding region on C1q with IgG based on the structure and molecular docking, which explains the inhibitory effect of TsCRT on C1q-IgG-initiated classical complement activation. Based on the key residues in TsCRTΔ involved in the binding activity to C1q, a 24 amino acid peptide called PTsCRT was constructed that displayed strong C1q-binding activity and inhibited C1q-IgG-initiated classical complement activation. This study is the first to elucidate the structural basis of the role of TsCRT in immune evasion, providing an approach to develop helminth-derived bifunctional peptides as vaccine target to prevent parasite infections or as a therapeutic agent to treat complement-related autoimmune diseases.


Asunto(s)
Calreticulina , Complemento C1q , Evasión Inmune , Trichinella spiralis , Trichinella spiralis/inmunología , Complemento C1q/inmunología , Complemento C1q/metabolismo , Complemento C1q/química , Animales , Calreticulina/inmunología , Calreticulina/química , Calreticulina/metabolismo , Cristalografía por Rayos X , Unión Proteica , Simulación del Acoplamiento Molecular , Proteínas del Helminto/inmunología , Proteínas del Helminto/química , Activación de Complemento/inmunología , Inmunoglobulina G/inmunología , Humanos , Antígenos Helmínticos/inmunología , Antígenos Helmínticos/química , Triquinelosis/inmunología , Triquinelosis/parasitología , Vía Clásica del Complemento/inmunología , Conformación Proteica
7.
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
8.
Front Immunol ; 15: 1352330, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38694513

RESUMEN

Introduction: COVID-19 patients can develop autoantibodies against a variety of secreted and membrane proteins, including some expressed on lymphocytes. However, it is unclear what proportion of patients might develop anti-lymphocyte antibodies (ALAb) and what functional relevance they might have. Methods: We evaluated the presence and lytic function of ALAb in the sera of a cohort of 85 COVID-19 patients (68 unvaccinated and 17 vaccinated) assigned to mild (N=63), or moderate/severe disease (N=22) groups. Thirty-seven patients were followed-up after recovery. We also analyzed in vivo complement deposition on COVID-19 patients' lymphocytes and examined its correlation with lymphocyte numbers during acute disease. Results: Compared with healthy donors (HD), patients had an increased prevalence of IgM ALAb, which was significantly higher in moderate/severe disease patients and persisted after recovery. Sera from IgM ALAb+ patients exhibited complement-dependent cytotoxicity (CDC) against HD lymphocytes. Complement protein C3b deposition on patients' CD4 T cells was inversely correlated with CD4 T cell numbers. This correlation was stronger in moderate/severe disease patients. Discussion: IgM ALAb and complement activation against lymphocytes may contribute to the acute lymphopenia observed in COVID-19 patients.


Asunto(s)
Autoanticuerpos , COVID-19 , Activación de Complemento , Inmunoglobulina M , SARS-CoV-2 , Humanos , COVID-19/inmunología , COVID-19/sangre , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Masculino , Femenino , Persona de Mediana Edad , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Activación de Complemento/inmunología , SARS-CoV-2/inmunología , Anciano , Adulto , Linfocitos/inmunología , Prevalencia , Linfocitos T CD4-Positivos/inmunología , Linfopenia/inmunología , Linfopenia/sangre , Complemento C3b/inmunología
9.
Mol Immunol ; 170: 19-25, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38598870

RESUMEN

The assembly of tissue-damaging membrane attack complexes (MACs; C5b-9) is a major mechanism by which excessive complement activation causes diseases. We previously developed a mouse anti-human C6 monoclonal antibody (mAb) 1C9 that selectively inhibits the assembly of MACs in human and non-human primates. In this project, we found that 1C9 also cross-reacted with rat and guinea pig C6, and determined its binding domains on C6 using different truncated C6 proteins. We then humanized the anti-C6 mAb by molecular modeling and complementarity-determining region grafting. After screening a library of 276 humanized variants with different combinations of humanized light and heavy chains in biophysical assays, we identified clone 3713 with the best developability profile, and an increased affinity against C6 when compared with the parental 1C9 mAb. This humanized 3713 mAb inhibited human, monkey, and rat complement-mediated hemolysis in vitro, and more importantly, it significantly reduced complement-mediated hemolysis in vivo in rats. These results demonstrated the successful humanization of the anti-C6 mAb and suggested that the humanized 3713 mAb could be further developed as a new therapeutic that selectively targets MAC for certain complement-mediated pathological conditions.


Asunto(s)
Anticuerpos Monoclonales , Complemento C6 , Hemólisis , Animales , Humanos , Ratas , Cobayas , Ratones , Hemólisis/efectos de los fármacos , Hemólisis/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Complemento C6/inmunología , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Activación de Complemento/inmunología , Activación de Complemento/efectos de los fármacos , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Reacciones Cruzadas/inmunología
10.
Colloids Surf B Biointerfaces ; 238: 113886, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38608461

RESUMEN

In this work, poly(lactide) nanoparticles were equipped with a bioinspired coating layer based on poly[2-(methacryloyloxy)ethyl phosphorylcholine] and then evaluated when administered to the lungs and after intravenous injection. Compared to the plain counterparts, the chosen zwitterionic polymer shell prevented the coated colloidal formulation from aggregation and conditioned it for lower cytotoxicity, protein adsorption, complement activation and phagocytic cell uptake. Consequently, no interference with the biophysical function of the lung surfactant system could be detected accompanied by negligible protein and cell influx into the bronchoalveolar space after intratracheal administration. When injected into the central compartment, the coated formulation showed a prolonged circulation half-life and a delayed biodistribution to the liver. Taken together, colloidal drug delivery vehicles would clearly benefit from the investigated poly[2-(methacryloyloxy)ethyl phosphorylcholine]-based polymer coatings.


Asunto(s)
Coloides , Sistemas de Liberación de Medicamentos , Fosforilcolina , Coloides/química , Animales , Fosforilcolina/química , Fosforilcolina/análogos & derivados , Nanopartículas/química , Poliésteres/química , Ratones , Polímeros/química , Polímeros/farmacología , Distribución Tisular , Pulmón/metabolismo , Ácidos Polimetacrílicos/química , Activación de Complemento/efectos de los fármacos , Metacrilatos/química , Humanos
11.
Int J Mol Sci ; 25(7)2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38612407

RESUMEN

A small fraction of people vaccinated with mRNA-lipid nanoparticle (mRNA-LNP)-based COVID-19 vaccines display acute or subacute inflammatory symptoms whose mechanism has not been clarified to date. To better understand the molecular mechanism of these adverse events (AEs), here, we analyzed in vitro the vaccine-induced induction and interrelations of the following two major inflammatory processes: complement (C) activation and release of proinflammatory cytokines. Incubation of Pfizer-BioNTech's Comirnaty and Moderna's Spikevax with 75% human serum led to significant increases in C5a, sC5b-9, and Bb but not C4d, indicating C activation mainly via the alternative pathway. Control PEGylated liposomes (Doxebo) also induced C activation, but, on a weight basis, it was ~5 times less effective than that of Comirnaty. Viral or synthetic naked mRNAs had no C-activating effects. In peripheral blood mononuclear cell (PBMC) cultures supplemented with 20% autologous serum, besides C activation, Comirnaty induced the secretion of proinflammatory cytokines in the following order: IL-1α < IFN-γ < IL-1ß < TNF-α < IL-6 < IL-8. Heat-inactivation of C in serum prevented a rise in IL-1α, IL-1ß, and TNF-α, suggesting C-dependence of these cytokines' induction, although the C5 blocker Soliris and C1 inhibitor Berinert, which effectively inhibited C activation in both systems, did not suppress the release of any cytokines. These findings suggest that the inflammatory AEs of mRNA-LNP vaccines are due, at least in part, to stimulation of both arms of the innate immune system, whereupon C activation may be causally involved in the induction of some, but not all, inflammatory cytokines. Thus, the pharmacological attenuation of inflammatory AEs may not be achieved via monotherapy with the tested C inhibitors; efficacy may require combination therapy with different C inhibitors and/or other anti-inflammatory agents.


Asunto(s)
COVID-19 , Inactivadores del Complemento , Nanopartículas , Humanos , Liposomas , Vacunas contra la COVID-19/efectos adversos , Leucocitos Mononucleares , Citocinas , Factor de Necrosis Tumoral alfa , Vacuna BNT162 , Activación de Complemento , Lípidos
12.
Front Immunol ; 15: 1383936, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38638432

RESUMEN

In the quest to address the critical shortage of donor organs for transplantation, xenotransplantation stands out as a promising solution, offering a more abundant supply of donor organs. Yet, its widespread clinical adoption remains hindered by significant challenges, chief among them being immunological rejection. Central to this issue is the role of the complement system, an essential component of innate immunity that frequently triggers acute and chronic rejection through hyperacute immune responses. Such responses can rapidly lead to transplant embolism, compromising the function of the transplanted organ and ultimately causing graft failure. This review delves into three key areas of xenotransplantation research. It begins by examining the mechanisms through which xenotransplantation activates both the classical and alternative complement pathways. It then assesses the current landscape of xenotransplantation from donor pigs, with a particular emphasis on the innovative strides made in genetically engineering pigs to evade complement system activation. These modifications are critical in mitigating the discordance between pig endogenous retroviruses and human immune molecules. Additionally, the review discusses pharmacological interventions designed to support transplantation. By exploring the intricate relationship between the complement system and xenotransplantation, this retrospective analysis not only underscores the scientific and clinical importance of this field but also sheds light on the potential pathways to overcoming one of the major barriers to the success of xenografts. As such, the insights offered here hold significant promise for advancing xenotransplantation from a research concept to a viable clinical reality.


Asunto(s)
Activación de Complemento , Rechazo de Injerto , Animales , Humanos , Porcinos , Trasplante Heterólogo , Animales Modificados Genéticamente , Estudios Retrospectivos , Rechazo de Injerto/prevención & control , Proteínas del Sistema Complemento
13.
Ren Fail ; 46(1): 2344658, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38644359

RESUMEN

Previous studies have highlighted the significant role of complement activation in kidney injuries induced by rhabdomyolysis, intravascular hemolysis, sepsis, and ischemia-reperfusion. Nevertheless, the specific role and mechanism of complement activation in acute kidney injury (AKI) caused by wasp venom remain unclear. The aim of this study was to elucidate the specific complement pathway activated and investigate complement activation in AKI induced by wasp venom. In this study, a complement-depleted mouse model was used to investigate the role of complement in wasp venom-induced AKI. Mice were randomly categorized into control, cobra venom factor (CVF), AKI, and CVF + AKI groups. Compared to the AKI group, the CVF + AKI group showed improved pathological changes in kidneys and reduced blood urea nitrogen (BUN) levels. The expression levels of renal complement 3 (C3), complement 5 (C5), complement 1q (C1q), factor B (FB), mannose-binding lectin (MBL), and C5b-9 in AKI group were upregulated compared with the control group. Conversely, the renal tissue expression levels of C3, C5, C1q, FB, MBL, and C5b-9 were decreased in the CVF + AKI group compared to those in the AKI group. Complement activation occurs through all three pathways in AKI induced by wasp venom. Furthermore, complement depletion by CVF attenuates wasp venom-induced nephrotoxicity, suggesting that complement activation plays a primary role in the pathogenesis of wasp venom-induced AKI.


Asunto(s)
Lesión Renal Aguda , Activación de Complemento , Modelos Animales de Enfermedad , Venenos de Avispas , Animales , Lesión Renal Aguda/inmunología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/etiología , Lesión Renal Aguda/inducido químicamente , Ratones , Venenos de Avispas/inmunología , Venenos de Avispas/efectos adversos , Masculino , Riñón/patología , Venenos Elapídicos , Nitrógeno de la Urea Sanguínea , Complemento C3/metabolismo , Proteínas del Sistema Complemento/metabolismo
14.
J Neuroinflammation ; 21(1): 98, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38632569

RESUMEN

BACKGROUND: Traumatic brain injury (TBI) is associated with the development of visual system disorders. Visual deficits can present with delay and worsen over time, and may be associated with an ongoing neuroinflammatory response that is known to occur after TBI. Complement system activation is strongly associated with the neuroinflammatory response after TBI, but whether it contributes to vision loss after TBI is unexplored. METHODS: Acute and chronic neuroinflammatory changes within the dorsal lateral geniculate nucleus (dLGN) and retina were investigated subsequent to a moderate to severe murine unilateral controlled cortical impact. Neuroinflammatory and histopathological outcomes were interpreted in the context of behavioral and visual function data. To investigate the role of complement, cohorts were treated after TBI with the complement inhibitor, CR2-Crry. RESULTS: At 3 days after TBI, complement component C3 was deposited on retinogeniculate synapses in the dLGN both ipsilateral and contralateral to the lesion, which was reduced in CR2-Crry treated animals. This was associated with microglia morphological changes in both the ipsilateral and contralateral dLGN, with a less ramified phenotype in vehicle compared to CR2-Crry treated animals. Microglia in vehicle treated animals also had a greater internalized VGlut2 + synaptic volume after TBI compared to CR2-Crry treated animals. Microglia morphological changes seen acutely persisted for at least 49 days after injury. Complement inhibition also reduced microglial synaptic internalization in the contralateral dLGN and increased the association between VGLUT2 and PSD95 puncta, indicating preservation of intact synapses. Unexpectedly, there were no changes in the thickness of the inner retina, retinal nerve fiber layer or retinal ganglion layer. Neuropathological changes in the dLGN were accompanied by reduced visual acuity at subacute and chronic time points after TBI, with improvement seen in CR2-Crry treated animals. CONCLUSION: TBI induces complement activation within the dLGN and promotes microglial activation and synaptic internalization. Complement inhibition after TBI in a clinically relevant paradigm reduces complement activation, maintains a more surveillance-like microglia phenotype, and preserves synaptic density within the dLGN. Together, the data indicate that complement plays a key role in the development of visual deficits after TBI via complement-dependent microglial phagocytosis of synapses within the dLGN.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Animales , Ratones , Lesiones Traumáticas del Encéfalo/patología , Complemento C3/genética , Activación de Complemento , Células Ganglionares de la Retina/patología , Inflamación/complicaciones , Proteínas Recombinantes de Fusión
15.
Bratisl Lek Listy ; 125(5): 275-280, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38624051

RESUMEN

C4d, a split product of C4 activation in classical and lectin pathways of the complement system activation, has been regarded as a footprint of tissue damage in antibody-mediated rejection in transplantology. The introduction of C4d staining into daily clinical practice aroused an ever-increasing interest in the role of antibody-mediated mechanisms in kidney allograft rejection. However, this marker of complement activation is also important in other various kidney glomerular pathologies such as immunoglobulin A nephropathy, membranoproliferative glomerulonephritis, lupus nephritis, and others. In routine histopathological practice, C4d staining can be done by two histological methods, specifically by immunofluorescence on frozen tissue using monoclonal antibody to C4d (with the downside of unsteady availability of frozen tissue) or by immunohistochemistry using C4d antibodies on formalin-fixed and paraffin-embedded renal tissue. The aim of this narrative review is to summarize recent knowledge about the complement fragment C4d and its significance in different kidney pathologies, focusing on its immunohistochemical detection in renal tissue biopsies. We have supplemented this review with our experience with our proprietary methodology of preparation and practical use of antibodies such as anti-C4d, on a small national level. Immunohistochemical staining for C4d has revolutionized the field of renal histopathology. Despite being a simple diagnostic test, its utility can be of utmost importance, especially in a resource-poor setting where immunofluorescence and frozen tissue may not be available (Fig. 2, Ref. 53). Keywords: C4d deposition, immunohistochemistry, kidney glomerular diseases, kidney transplant, renal tubular damage.


Asunto(s)
Trasplante de Riñón , Glomérulos Renales/patología , Riñón/metabolismo , Anticuerpos Monoclonales , Fragmentos de Péptidos , Activación de Complemento , Biopsia
16.
J Immunol Methods ; 528: 113668, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38574804

RESUMEN

Complement plays a critical role in the immune response toward nanomaterials. The complement attack on a foreign surface results in the deposition of C3, assembly of C3 convertases, the release of anaphylatoxins C3a and C5a, and finally, the formation of membrane attack complex C5b-9. Various technologies can measure complement activation markers in the fluid phase, but measurements of surface C3 deposition are less common. Previously, we developed an ultracentrifugation-based dot blot immunoassay (DBI) to measure the deposition of C3 and other protein corona components on nanoparticles. Here, we validate the repeatability of the DBI and its correlation with pathway-specific and common fluid phase markers. Moreover, we discuss the advantages of DBI, such as cost-effectiveness and versatility, while addressing potential limitations. This study provides insights into complement activation at the nanosurface level, offering a valuable tool for nanomedicine researchers in the field.


Asunto(s)
Nanopartículas , Opsonización , Activación de Complemento , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Inmunoensayo , Complemento C3a , Complemento C5a , Complemento C5
17.
PLoS Negl Trop Dis ; 18(4): e0012048, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38564496

RESUMEN

BACKGROUND: Numerous studies indicate a potential protective role of helminths in diabetes mellitus (DM) progression. The complement system, vital for host defense, plays a crucial role in tissue homeostasis and immune surveillance. Dysregulated complement activation is implicated in diabetic complications. We aimed to investigate the influence of the helminth, Strongyloides stercoralis (Ss) on complement activation in individuals with type 2 DM (T2D). METHODOLOGY: We assessed circulating levels of complement proteins (C1q, C2, C3, C4, C4b, C5, C5a, and MBL (Lectin)) and their regulatory components (Factor B, Factor D, Factor H, and Factor I) in individuals with T2D with (n = 60) or without concomitant Ss infection (n = 58). Additionally, we evaluated the impact of anthelmintic therapy on these parameters after 6 months in Ss-infected individuals (n = 60). RESULTS: Ss+DM+ individuals demonstrated reduced levels of complement proteins (C1q, C4b, MBL (Lectin), C3, C5a, and C3b/iC3b) and complement regulatory proteins (Factor B and Factor D) compared to Ss-DM+ individuals. Following anthelmintic therapy, there was a partial reversal of these levels in Ss+DM+ individuals. CONCLUSION: Our findings indicate that Ss infection reduces complement activation, potentially mitigating inflammatory processes in individuals with T2D. The study underscores the complex interplay between helminth infections, complement regulation, and diabetes mellitus, offering insights into potential therapeutic avenues.


Asunto(s)
Antihelmínticos , Diabetes Mellitus Tipo 2 , Helmintos , Strongyloides stercoralis , Estrongiloidiasis , Animales , Humanos , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Factor B del Complemento , Factor D del Complemento/uso terapéutico , Complemento C1q , Estrongiloidiasis/complicaciones , Estrongiloidiasis/tratamiento farmacológico , Activación de Complemento , Antihelmínticos/uso terapéutico , Lectinas
18.
Invest Ophthalmol Vis Sci ; 65(4): 43, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38683564

RESUMEN

Purpose: Complement dysregulation is a key component in the pathogenesis of age-related macular degeneration (AMD) and related diseases such as early-onset macular drusen (EOMD). Although genetic variants of complement factor H (CFH) are associated with AMD risk, the impact of CFH and factor H-like protein 1 (FHL-1) expression on local complement activity in human retinal pigment epithelium (RPE) remains unclear. Methods: We identified a novel CFH variant in a family with EOMD and generated patient induced pluripotent stem cell (iPSC)-derived RPE cells. We assessed CFH and FHL-1 co-factor activity through C3b breakdown assays and measured complement activation by immunostaining for membrane attack complex (MAC) formation. Expression of CFH, FHL-1, local alternative pathway (AP) components, and regulators of complement activation (RCA) in EOMD RPE cells was determined by quantitative PCR, western blot, and immunostaining. Isogenic EOMD (cEOMD) RPE was generated using CRISPR/Cas9 gene editing. Results: The CFH variant (c.351-2A>G) resulted in loss of CFH and FHL-1 expression and significantly reduced CFH and FHL-1 protein expression (∼50%) in EOMD iPSC RPE cells. These cells exhibited increased MAC deposition upon exposure to normal human serum. Under inflammatory or oxidative stress conditions, CFH and FHL-1 expression in EOMD RPE cells paralleled that of controls, whereas RCA expression, including MAC formation inhibitors, was elevated. CRISPR/Cas9 correction restored CFH/FHL-1 expression and mitigated alternative pathway complement activity in cEOMD RPE cells. Conclusions: Identification of a novel CFH variant in patients with EOMD resulting in reduced CFH and FHL-1 and increased local complement activity in EOMD iPSC RPE supports the involvement of CFH haploinsufficiency in EOMD pathogenesis.


Asunto(s)
Factor H de Complemento , Haploinsuficiencia , Péptidos y Proteínas de Señalización Intracelular , Proteínas con Dominio LIM , Degeneración Macular , Proteínas Musculares , Epitelio Pigmentado de la Retina , Humanos , Factor H de Complemento/genética , Factor H de Complemento/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Degeneración Macular/genética , Degeneración Macular/metabolismo , Masculino , Femenino , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas Inactivadoras del Complemento C3b/genética , Proteínas Inactivadoras del Complemento C3b/metabolismo , Activación de Complemento/genética , Linaje , Western Blotting , Proteínas del Sistema Complemento/metabolismo , Proteínas del Sistema Complemento/genética , Drusas Retinianas/genética , Drusas Retinianas/metabolismo , Persona de Mediana Edad
19.
Mol Pharm ; 21(5): 2272-2283, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38607681

RESUMEN

Over the years, there has been significant interest in PEGylated lipid-based nanocarriers within the drug delivery field. The inevitable interplay between the nanocarriers and plasma protein plays a pivotal role in their in vivo biological fate. Understanding the factors influencing lipid-based nanocarrier and protein corona interactions is of paramount importance in the design and clinical translation of these nanocarriers. Herein, discoid-shaped lipid nanodiscs (sNDs) composed of different phospholipids with varied lipid tails and head groups were fabricated. We investigated the impact of phospholipid components on the interaction between sNDs and serum proteins, particle stability, and biodistribution. The results showed that all of these lipid nanodiscs remained stable over a 15 day storage period, while their stability in the blood serum demonstrated significant differences. The sND composed of POPG exhibited the least stability due to its potent complement activation capability, resulting in rapid blood clearance. Furthermore, a negative correlation between the complement activation capability and serum stability was identified. Pharmacokinetic and biodistribution experiments indicated that phospholipid composition did not influence the capability of sNDs to evade the accelerated blood clearance phenomenon. Complement deposition on the sND was inversely associated with the area under the curve. Additionally, all lipid nanodiscs exhibited dominant adsorption of apolipoprotein. Remarkably, the POPC-based lipid nanodisc displayed a significantly higher deposition of apolipoprotein E, contributing to an obvious brain distribution, which provides a promising tool for brain-targeted drug delivery.


Asunto(s)
Nanopartículas , Fosfolípidos , Corona de Proteínas , Corona de Proteínas/química , Animales , Fosfolípidos/química , Distribución Tisular , Ratones , Nanopartículas/química , Portadores de Fármacos/química , Nanoestructuras/química , Masculino , Activación de Complemento/efectos de los fármacos , Lípidos/química , Sistemas de Liberación de Medicamentos/métodos , Proteínas Sanguíneas/metabolismo , Proteínas Sanguíneas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...