Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.740
Filtrar
1.
Front Immunol ; 15: 1449003, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39295874

RESUMEN

Pseudomonas aeruginosa is a leading cause of nosocomial bloodstream infections. The outcome of these infections depends on the virulence of the microorganism as well as host-related conditions and factors. The complement system plays a crucial role in defense against bloodstream infections. P. aeruginosa counteracts complement attack by recruiting Factor H (FH) that inhibits complement amplification on the bacterial surface. Factor H-related proteins (FHRs) are a group of plasma proteins evolutionarily related to FH that have been postulated to interfere this bacterial evasion mechanism. In this study, we demonstrate that FHR-3 competes with purified FH for binding to P. aeruginosa and identify EF-Tu as a common bacterial target for both complement regulator factors. Importantly, elevated levels of FHR-3 in human serum promote complement activation, leading to increased opsonization and killing of P. aeruginosa. Conversely, physiological concentrations of FHR-3 have no significant effect. Our findings suggest that FHR-3 may serve as a protective host factor against P. aeruginosa infections.


Asunto(s)
Factor H de Complemento , Infecciones por Pseudomonas , Pseudomonas aeruginosa , Pseudomonas aeruginosa/inmunología , Humanos , Infecciones por Pseudomonas/inmunología , Factor H de Complemento/metabolismo , Factor H de Complemento/inmunología , Bacteriemia/inmunología , Bacteriemia/microbiología , Activación de Complemento/inmunología , Interacciones Huésped-Patógeno/inmunología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Unión Proteica
2.
Front Immunol ; 15: 1447991, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39136026

RESUMEN

Background: Factor H (FH) is a major soluble inhibitor of the complement system and part of a family comprising five related proteins (FHRs 1-5). Deficiency of FHR1 was described to be linked to an elevated risk of systemic lupus erythematosus (SLE). As FHR1 can partially antagonize the functionality of FH, an altered FHR1/FH ratio could not only enhance SLE vulnerability but also affect the disease expression. This study focuses on the analysis of FH and FHR1 at a protein level, and the occurrence of anti-FH autoantibodies (anti-FH) in a large cohort of SLE patients to explore their association with disease activity and/or expression. Methods: We assessed FH and FHR1 levels in plasma from 378 SLE patients compared to 84 healthy controls (normal human plasma, NHP), and sera from another cohort of 84 healthy individuals (normal human serum, NHS), using RayBio® CFH and CFHR1 ELISA kits. Patients were recruited by the Swiss SLE Cohort Study (SSCS). Unmeasurable FHR1 levels were all confirmed by Western blot, and in a subgroup of patients by PCR. Anti-FH were measured in SLE patients with non-detectable FHR1 levels and matched control patients using Abnova's CFH IgG ELISA kit. Results: Overall, FH and FHR1 levels were significantly higher in healthy controls, but there was no significant difference in FHR1/FH ratios between SLE patients and NHPs. However, SLE patients showed a significantly higher prevalence of undetectable FHR1 compared to all healthy controls (35/378 SLE patients versus 6/168 healthy controls; p= 0.0214, OR=2.751, 95% CI = 1.115 - 8.164), with a consistent trend across all ethnic subgroups. Levels of FH and FHR1, FHR1/FH ratios and absence of FHR1 were not consistently associated with disease activity and/or specific disease manifestations, but absence of FHR1 (primarily equivalent to CFHR1 deficiency) was linked to the presence of anti-FH in SLE patients (p=0.039). Conclusions: Deficiency of FHR1 is associated with a markedly elevated risk of developing SLE. A small proportion of FHR1-deficient SLE patients was found to have autoantibodies against FH but did not show clinical signs of microangiopathy.


Asunto(s)
Autoanticuerpos , Factor H de Complemento , Lupus Eritematoso Sistémico , Humanos , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/sangre , Femenino , Masculino , Factor H de Complemento/metabolismo , Factor H de Complemento/inmunología , Adulto , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Persona de Mediana Edad , Proteínas Inactivadoras del Complemento C3b/genética , Adulto Joven , Anciano , Estudios de Casos y Controles , Adolescente , Proteínas Sanguíneas
3.
Mol Biol Rep ; 51(1): 888, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39105829

RESUMEN

OBJECTIVE: To explore the relationship between rs1410996 polymorphism of CFH gene and essential hypertension (EH) in the Yunnan Han population. METHODS: rs1410996 of CFH gene was genotyped based on the collected clinical phenotypes of the EH patients (n = 520) and healthy people (n = 494). RESULTS: On the genotype model and dominance model, there was no relationship between rs1410996 of CFH gene and EH after adjustment (P > 0.05). On the dominance model of male EH patients, the pulse pressure (PP) level of CC genotype carriers was higher than that of (CT + TT) genotype carriers after adjustment (P < 0.05). CONCLUSION: rs1410996 of CFH gene has no correlation with the genetic susceptibility to EH in the Yunnan Han population, but it is related to the PP level in male patients.


Asunto(s)
Pueblo Asiatico , Factor H de Complemento , Hipertensión Esencial , Predisposición Genética a la Enfermedad , Genotipo , Polimorfismo de Nucleótido Simple , Humanos , Masculino , Hipertensión Esencial/genética , Persona de Mediana Edad , Femenino , China , Factor H de Complemento/genética , Polimorfismo de Nucleótido Simple/genética , Pueblo Asiatico/genética , Frecuencia de los Genes/genética , Anciano , Estudios de Casos y Controles , Adulto , Hipertensión/genética , Estudios de Asociación Genética/métodos , Alelos , Presión Sanguínea/genética
4.
Front Immunol ; 15: 1379023, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39104533

RESUMEN

Antibody-mediated complement-dependent cytotoxicity (CDC) on malignant cells is regulated by several complement control proteins, including the inhibitory complement factor H (fH). fH consists of 20 short consensus repeat elements (SCRs) with specific functional domains. Previous research revealed that the fH-derived SCRs 19-20 (SCR1920) can displace full-length fH on the surface of chronic lymphocytic leukemia (CLL) cells, which sensitizes CLL cells for e.g. CD20-targeting therapeutic monoclonal antibody (mAb) induced CDC. Therefore, we constructed lentiviral vectors for the generation of cell lines that stably produce mAb-SCR-fusion variants starting from the clinically approved parental mAbs rituximab, obinutuzumab and ofatumumab, respectively. Flow-cytometry revealed that the modification of the mAbs by the SCRs does not impair the binding to CD20. Increased in vitro lysis potency compared to their parental mAbs was corroborated by showing specific and dose dependent target cell elimination by CDC when compared to their parental mAbs. Lysis of CLL cells was not affected by the depletion of NK cells, suggesting that antibody-dependent cellular cytotoxicity plays a minor role in this context. Overall, this study emphasizes the crucial role of CDC in the elimination of CLL cells by mAbs and introduces a novel approach for enhancing CDC by directly fusing fH SCR1920 with mAbs.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD20 , Factor H de Complemento , Leucemia Linfocítica Crónica de Células B , Rituximab , Humanos , Antígenos CD20/inmunología , Antígenos CD20/genética , Factor H de Complemento/inmunología , Factor H de Complemento/metabolismo , Factor H de Complemento/genética , Leucemia Linfocítica Crónica de Células B/inmunología , Rituximab/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Línea Celular Tumoral
5.
BMC Nephrol ; 25(1): 224, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39009967

RESUMEN

BACKGROUND: No reports have shown histological changes before and after anti-C5 monoclonal antibody treatment in patients with atypical hemolytic uremic syndrome (aHUS). Here, we report a rare case of complement-mediated aHUS with a complement factor H (CFH) mutation and anti-CFH antibodies who underwent multiple kidney biopsies. CASE PRESENTATION: A 53-year-old woman developed aHUS with CFH gene mutation [c.3572C > T (p. Ser1191 Leu)] and anti-CFH antibodies. Her father had succumbed to acute kidney injury (AKI) in his 30 s. She exhibited AKI, thrombocytopenia, and hemolytic anemia with schistocytes. After improving the platelet count with one session of plasma exchange, a kidney biopsy was performed one month after the onset of symptoms. Blood vessel thrombosis, obvious endothelial swelling, endocapillary hypercellularity, and subendothelial exudative lesions in the glomeruli and arterioles were detected. Anti-C5 monoclonal antibody treatment with eculizumab immediately improved disease activity. A second biopsy 3 months later revealed marked improvement of endothelial injuries with residual membrane double contours and exudative lesions. A third biopsy at 17 months after gradual improvement of kidney function showed a further decrease of double contours along with alterations of the exudative lesions to fibrous intimal thickening. CONCLUSIONS: This is the first report showing the pathophysiology of aHUS in the kidneys and the efficacy of anti-C5 monoclonal antibody treatment by presenting serial kidney pathological features before and after anti-C5 monoclonal antibody treatment. Since her CFH mutation was considered the most important pathological condition, treatment centered on eculizumab was administered, resulting in a good long-term prognosis. In addition, kidney pathological resolution in aHUS occurred over 1 year after anti-C5 monoclonal antibody treatment.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Síndrome Hemolítico Urémico Atípico , Factor H de Complemento , Humanos , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Femenino , Persona de Mediana Edad , Anticuerpos Monoclonales Humanizados/uso terapéutico , Complemento C5/antagonistas & inhibidores , Riñón/patología
6.
Pediatr Blood Cancer ; 71(9): e31175, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38961591

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is a complement-mediated thrombotic microangiopathy sometimes associated with germline variants in genes of the complement system. Clinical findings of microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury arise due to aberrant complement protein activation in the circulation. A 13-month-old boy with metastatic neuroblastoma (NB) developed aHUS during his first cycle of induction chemotherapy with germline testing revealing a complement factor H (CFH) gene mutation, currently classified as a variant of uncertain significance (VUS). Now he is in disease remission after successful complement blockade therapy, thus highlighting a unique presentation of aHUS in a patient with newly diagnosed NB.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Quimioterapia de Inducción , Neuroblastoma , Humanos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/patología , Neuroblastoma/genética , Masculino , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/patología , Quimioterapia de Inducción/efectos adversos , Lactante , Factor H de Complemento/genética , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Mutación de Línea Germinal
7.
Appl Microbiol Biotechnol ; 108(1): 425, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39042328

RESUMEN

Borrelia, spirochetes transmitted by ticks, are the etiological agents of numerous multisystemic diseases, such as Lyme borreliosis (LB) and tick-borne relapsing fever (TBRF). This study focuses on two surface proteins from two Borrelia subspecies involved in these diseases: CspZ, expressed by Borrelia burgdorferi sensu stricto (also named BbCRASP-2 for complement regulator-acquiring surface protein 2), and the factor H binding A (FhbA), expressed by Borrelia hermsii. Numerous subspecies of Borrelia, including these latter, are able to evade the immune defenses of a variety of potential vertebrate hosts in a number of ways. In this context, previous data suggested that both surface proteins play a role in the immune evasion of both Borrelia subspecies by interacting with key regulators of the alternative pathway of the human complement system, factor H (FH) and FH-like protein 1 (FHL-1). The recombinant proteins, CspZ and FhbA, were expressed in Escherichia coli and purified by one-step metal-affinity chromatography, with yields of 15 and 20 mg or pure protein for 1 L of cultured bacteria, respectively. The purity was evaluated by SDS-PAGE and HPLC and is close to about 95%. The mass of CspZ and FhbA was checked by mass spectrometry (MS). Proper folding of CspZ and FhbA was confirmed by circular dichroism (CD), and their biological activity, namely their interaction with purified FH from human serum (recombinant FH15-20 and recombinant FHL-1), was characterized by SPR. Such a study provides the basis for the biochemical characterization of the studied proteins and their biomolecular interactions which is a necessary prerequisite for the development of new approaches to improve the current diagnosis of LB and TBRF. KEY POINTS: • DLS, CD, SEC-MALS, NMR, HPLC, and MS are tools for protein quality assessment • Borrelia spp. possesses immune evasion mechanisms, including human host complement • CspZ and FhbA interact with high affinity (pM to nM) to human FH and rFHL-1.


Asunto(s)
Proteínas Bacterianas , Proteínas Recombinantes , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Humanos , Borrelia burgdorferi/genética , Borrelia burgdorferi/metabolismo , Borrelia burgdorferi/inmunología , Cromatografía de Afinidad , Escherichia coli/genética , Escherichia coli/metabolismo , Borrelia/genética , Borrelia/metabolismo , Borrelia/inmunología , Factor H de Complemento/metabolismo , Factor H de Complemento/genética , Enfermedad de Lyme/microbiología , Proteínas Inactivadoras del Complemento C3b/genética , Proteínas Inactivadoras del Complemento C3b/metabolismo , Expresión Génica
8.
Ren Fail ; 46(2): 2365396, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38874150

RESUMEN

OBJECTIVE: The aim of this study was to investigate the potential importance of complement system activation, with particular emphasis on the complement alternative pathway (AP), in the pathogenesis of hypertensive renal damage. METHODS: Serum complement C3, complement Factor H (CFH) and AP activation were assessed in 66 participants with established essential hypertension with renal damage (RD). Fifty-nine patients with age- and sex-matched essential hypertension without renal damage (NRD) and 58 healthy participants (normal) were selected. RESULTS: Our study revealed that C3 and AP50 continuously increased from normal to NRD to RD (p < 0.05, respectively), while CFH was significantly lower than that in NRD and healthy participants (p < 0.05, respectively). After multifactorial logistic regression analysis corrected for confounders, elevated serum C3 (p = 0.001) and decreased CFH (p < 0.001) were found to be independent risk factors for hypertension in healthy participants; elevated serum C3 (p = 0.034), elevated AP50 (p < 0.001), decreased CFH (p < 0.001), increased age (p = 0.011) and increased BMI (p = 0.013) were found to be independent risk factors for the progression of hypertension to hypertensive renal damage; elevated serum C3 (p = 0.017), elevated AP50 (p = 0.023), decreased CFH (p = 0.005) and increased age (p = 0.041) were found to be independent risk factors for the development of hypertensive renal damage in healthy participants. CONCLUSION: Abnormal activation of complement, particularly complement AP, may be a risk factor for the development and progression of hypertensive renal damage.


Asunto(s)
Complemento C3 , Factor H de Complemento , Humanos , Masculino , Femenino , Persona de Mediana Edad , Estudios de Casos y Controles , Complemento C3/metabolismo , Complemento C3/análisis , Factores de Riesgo , Anciano , Adulto , Hipertensión/complicaciones , Hipertensión/sangre , Activación de Complemento , Hipertensión Esencial/sangre , Hipertensión Esencial/complicaciones , Hipertensión Esencial/fisiopatología , Modelos Logísticos , Vía Alternativa del Complemento , Progresión de la Enfermedad
9.
Front Immunol ; 15: 1334151, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38919628

RESUMEN

Introduction: Complement factor H (FH) is a major regulator of the complement alternative pathway, its mutations predispose to an uncontrolled activation in the kidney and on blood cells and to secondary C3 deficiency. Plasma exchange has been used to correct for FH deficiency and although the therapeutic potential of purified FH has been suggested by in vivo experiments in animal models, a clinical approved FH concentrate is not yet available. We aimed to develop a purification process of FH from a waste fraction rather than whole plasma allowing a more efficient and ethical use of blood and plasma donations. Methods: Waste fractions from industrial plasma fractionation (pooled human plasma) were analyzed for FH content by ELISA. FH was purified from unused fraction III and its decay acceleration, cofactor, and C3 binding capacity were characterized in vitro. Biodistribution was assessed by high-resolution dynamic PET imaging. Finally, the efficacy of the purified FH preparation was tested in the mouse model of C3 glomerulopathy (Cfh-/- mice). Results: Our purification method resulted in a high yield of highly purified (92,07%), pathogen-safe FH. FH concentrate is intact and fully functional as demonstrated by in vitro functional assays. The biodistribution revealed lower renal and liver clearance of human FH in Cfh-/- mice than in wt mice. Treatment of Cfh-/- mice documented its efficacy in limiting C3 activation and promoting the clearance of C3 glomerular deposits. Conclusion: We developed an efficient and economical system for purifying intact and functional FH, starting from waste material of industrial plasma fractionation. The FH concentrate could therefore constitute possible treatments options of patients with C3 glomerulopathy, particularly for those with FH deficiency, but also for patients with other diseases associated with alternative pathway activation.


Asunto(s)
Complemento C3 , Factor H de Complemento , Ratones Noqueados , Factor H de Complemento/metabolismo , Factor H de Complemento/genética , Animales , Humanos , Ratones , Modelos Animales de Enfermedad , Prueba de Estudio Conceptual , Ratones Endogámicos C57BL
10.
Front Immunol ; 15: 1368852, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38933264

RESUMEN

The classical pathway of the complement system is activated by the binding of C1q in the C1 complex to the target activator, including immune complexes. Factor H is regarded as the key downregulatory protein of the complement alternative pathway. However, both C1q and factor H bind to target surfaces via charge distribution patterns. For a few targets, C1q and factor H compete for binding to common or overlapping sites. Factor H, therefore, can effectively regulate the classical pathway activation through such targets, in addition to its previously characterized role in the alternative pathway. Both C1q and factor H are known to recognize foreign or altered-self materials, e.g., bacteria, viruses, and apoptotic/necrotic cells. Clots, formed by the coagulation system, are an example of altered self. Factor H is present abundantly in platelets and is a well-known substrate for FXIIIa. Here, we investigated whether clots activate the complement classical pathway and whether this is regulated by factor H. We show here that both C1q and factor H bind to the fibrin formed in microtiter plates and the fibrin clots formed under in vitro physiological conditions. Both C1q and factor H become covalently bound to fibrin clots, and this is mediated via FXIIIa. We also show that fibrin clots activate the classical pathway of complement, as demonstrated by C4 consumption and membrane attack complex detection assays. Thus, factor H downregulates the activation of the classical pathway induced by fibrin clots. These results elucidate the intricate molecular mechanisms through which the complement and coagulation pathways intersect and have regulatory consequences.


Asunto(s)
Coagulación Sanguínea , Complemento C1q , Factor H de Complemento , Vía Clásica del Complemento , Fibrina , Humanos , Factor H de Complemento/metabolismo , Factor H de Complemento/inmunología , Fibrina/metabolismo , Complemento C1q/metabolismo , Complemento C1q/inmunología , Vía Clásica del Complemento/inmunología , Unión Proteica , Activación de Complemento/inmunología , Plaquetas/inmunología , Plaquetas/metabolismo
12.
Blood Adv ; 8(15): 4181-4193, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-38865712

RESUMEN

ABSTRACT: Excessively activated or dysregulated complement activation may contribute to the pathogenesis of a wide range of human diseases, thus leading to a surge in complement inhibitors. Herein, we developed a human-derived and antibody-like C3b-targeted fusion protein (CRIg-FH-Fc) x2, termed CG001, that could potently block all 3 complement pathways. Complement receptor of the immunoglobulin superfamily (CRIg) and factor H (FH) bind to distinct sites in C3b and synergistically inhibit complement activation. CRIg occupancy in C3b prevents the recruitment of C3 and C5 substrates, whereas FH occupancy in C3b accelerates the decay of C3/C5 convertases and promotes the factor I-mediated degradation and inactivation of C3b. CG001 also showed therapeutic effects in alternative pathways-induced hemolytic mouse and classical pathways-induced mesangial proliferative glomerulonephritis rat models. In the pharmacological/toxicological evaluation in rats and cynomolgus monkeys, CG001 displayed an antibody-like pharmacokinetic profile, a convincing complement inhibitory effect, and no observable toxic effects. Therefore, CG001 holds substantial potential for human clinical studies.


Asunto(s)
Complemento C3b , Animales , Humanos , Ratas , Ratones , Complemento C3b/metabolismo , Inactivadores del Complemento/farmacología , Inactivadores del Complemento/uso terapéutico , Macaca fascicularis , Activación de Complemento/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico , Factor H de Complemento/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos
13.
Front Immunol ; 15: 1383123, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38799460

RESUMEN

Most drugs that target the complement system are designed to inhibit the complement pathway at either the proximal or terminal levels. The use of a natural complement regulator such as factor H (FH) could provide a superior treatment option by restoring the balance of an overactive complement system while preserving its normal physiological functions. Until now, the systemic treatment of complement-associated disorders with FH has been deemed unfeasible, primarily due to high production costs, risks related to FH purified from donors' blood, and the challenging expression of recombinant FH in different host systems. We recently demonstrated that a moss-based expression system can produce high yields of properly folded, fully functional, recombinant FH. However, the half-life of the initial variant (CPV-101) was relatively short. Here we show that the same polypeptide with modified glycosylation (CPV-104) achieves a pharmacokinetic profile comparable to that of native FH derived from human serum. The treatment of FH-deficient mice with CPV-104 significantly improved important efficacy parameters such as the normalization of serum C3 levels and the rapid degradation of C3 deposits in the kidney compared to treatment with CPV-101. Furthermore, CPV-104 showed comparable functionality to serum-derived FH in vitro, as well as similar performance in ex vivo assays involving samples from patients with atypical hemolytic uremic syndrome, C3 glomerulopathy and paroxysomal nocturnal hematuria. CPV-104 - the human FH analog expressed in moss - will therefore allow the treatment of complement-associated human diseases by rebalancing instead of inhibiting the complement cascade.


Asunto(s)
Factor H de Complemento , Humanos , Factor H de Complemento/metabolismo , Factor H de Complemento/genética , Animales , Ratones , Semivida , Polisacáridos/metabolismo , Bryopsida/metabolismo , Bryopsida/genética , Glicosilación , Proteínas Recombinantes , Ratones Noqueados , Ratones Endogámicos C57BL , Masculino
14.
Front Immunol ; 15: 1352022, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38698856

RESUMEN

The complement system is an innate immune mechanism against microbial infections. It involves a cascade of effector molecules that is activated via classical, lectin and alternative pathways. Consequently, many pathogens bind to or incorporate in their structures host negative regulators of the complement pathways as an evasion mechanism. Factor H (FH) is a negative regulator of the complement alternative pathway that protects "self" cells of the host from non-specific complement attack. FH has been shown to bind viruses including human influenza A viruses (IAVs). In addition to its involvement in the regulation of complement activation, FH has also been shown to perform a range of functions on its own including its direct interaction with pathogens. Here, we show that human FH can bind directly to IAVs of both human and avian origin, and the interaction is mediated via the IAV surface glycoprotein haemagglutinin (HA). HA bound to common pathogen binding footprints on the FH structure, complement control protein modules, CCP 5-7 and CCP 15-20. The FH binding to H1 and H3 showed that the interaction overlapped with the receptor binding site of both HAs, but the footprint was more extensive for the H3 HA than the H1 HA. The HA - FH interaction impeded the initial entry of H1N1 and H3N2 IAV strains but its impact on viral multicycle replication in human lung cells was strain-specific. The H3N2 virus binding to cells was significantly inhibited by preincubation with FH, whereas there was no alteration in replicative rate and progeny virus release for human H1N1, or avian H9N2 and H5N3 IAV strains. We have mapped the interaction between FH and IAV, the in vivo significance of which for the virus or host is yet to be elucidated.


Asunto(s)
Factor H de Complemento , Glicoproteínas Hemaglutininas del Virus de la Influenza , Virus de la Influenza A , Gripe Humana , Unión Proteica , Humanos , Factor H de Complemento/metabolismo , Factor H de Complemento/inmunología , Animales , Gripe Humana/inmunología , Gripe Humana/virología , Gripe Humana/metabolismo , Virus de la Influenza A/inmunología , Virus de la Influenza A/fisiología , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Sitios de Unión , Gripe Aviar/virología , Gripe Aviar/inmunología , Gripe Aviar/metabolismo , Aves/virología , Interacciones Huésped-Patógeno/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Subtipo H9N2 del Virus de la Influenza A/inmunología
15.
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
16.
Ophthalmic Res ; 67(1): 358-386, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38754401

RESUMEN

INTRODUCTION: Anti-vascular endothelial growth factor (anti-VEGF) agents have a variable effect on patients with age-related macular degeneration (AMD) that has been attributed to several causes, including genetic factors. We evaluated the effects of Complement Factor H (CFH) rs1061170/Y402H polymorphism on the response to anti-VEGF therapy among AMD patients. METHODS: PubMed, Scopus, EMBASE, Web of Science, and Google Scholar were used for a literature search. Pooled odds ratios (ORs) and their 95% confidence intervals (CIs) were estimated to assess the effects of CFH Y402H polymorphism on the response to anti-VEGF therapy in AMD. I2 was used to present the amount of heterogeneity. We used STATA version 14.0 software. RESULTS: Twenty-five papers reporting data for 4,681 patients were included in this study. Better response to anti-VEGF therapy was seen in T over C (OR = 1.25, 95% CI = 1.04-1.50), TT over CC (OR = 1.60, 95% CI = 1.06-2.4), and TT + TC over CC (OR = 1.68, 95% CI = 1.23-2.28) genotypes. There was no significant difference in the three other genetic models (TT vs. TC, TT vs. TC + CC, TC vs. TT + CC). In Asians, no significant difference was observed in all six genetic models. Ranibizumab and bevacizumab had similar efficacy; however, conbercept was more effective in homozygous genotypes. The literature indicated that TT and TC genotypes and T allele were associated with a better functional response, while the CC genotype and C alleles had a better anatomical response. The combination of risk alleles in ARMS2 A69S (rs10490924), VEGF-A (rs699947), and VEGF-A (rs833069) with Y420H is a predictor of non-respondents. CONCLUSION: In patients with AMD, the CFH Y402H is a predictor of the response to anti-VEGF agents and should be considered in the treatment plan.


Asunto(s)
Inhibidores de la Angiogénesis , Factor H de Complemento , Degeneración Macular , Polimorfismo de Nucleótido Simple , Factor A de Crecimiento Endotelial Vascular , Humanos , Factor H de Complemento/genética , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Inhibidores de la Angiogénesis/uso terapéutico , Degeneración Macular/genética , Degeneración Macular/tratamiento farmacológico , Genotipo
17.
BMC Nephrol ; 25(1): 164, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38745129

RESUMEN

BACKGROUND: Atypical haemolytic uremic syndrome (aHUS) is an uncommon form of thrombotic microangiopathy (TMA). However, it remains difficult to diagnose the disease early, given its non-specific and overlapping presentation to other conditions such as thrombotic thrombocytopenic purpura and typical HUS. It is also important to identify the underlying causes and to distinguish between primary (due to a genetic abnormality leading to a dysregulated alternative complement pathway) and secondary (often attributed by severe infection or inflammation) forms of the disease, as there is now effective treatment such as monoclonal antibodies against C5 for primary aHUS. However, primary aHUS with severe inflammation are often mistaken as a secondary HUS. We presented an unusual case of adult-onset Still's disease (AOSD) with macrophage activation syndrome (MAS), which is in fact associated with anti-complement factor H (anti-CFH) antibodies related aHUS. Although the aHUS may be triggered by the severe inflammation from the AOSD, the presence of anti-CFH antibodies suggests an underlying genetic defect in the alternative complement pathway, predisposing to primary aHUS. One should note that anti-CFH antibodies associated aHUS may not always associate with genetic predisposition to complement dysregulation and can be an autoimmune form of aHUS, highlighting the importance of genetic testing. CASE PRESENTATION: A 42 years old man was admitted with suspected adult-onset Still's disease. Intravenous methylprednisolone was started but patient was complicated with acute encephalopathy and low platelet. ADAMTS13 test returned to be normal and concurrent aHUS was eventually suspected, 26 days after the initial thrombocytopenia was presented. Plasma exchange was started and patient eventually had 2 doses of eculizumab after funding was approved. Concurrent tocilizumab was also used to treat the adult-onset Still's disease with MAS. The patient was eventually stabilised and long-term tocilizumab maintenance treatment was planned instead of eculizumab following haematology review. Although the aHUS may be a secondary event to MAS according to haematology opinion and the genetic test came back negative for the five major aHUS gene, high titre of anti-CFH antibodies was detected (1242 AU/ml). CONCLUSION: Our case highlighted the importance of prompt anti-CFH antibodies test and genetic testing for aHUS in patients with severe AOSD and features of TMA. Our case also emphasized testing for structural variants within the CFH and CFH-related proteins gene region, as part of the routine genetic analysis in patients with anti-CFH antibodies associated aHUS to improve diagnostic approaches.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Factor H de Complemento , Enfermedad de Still del Adulto , Humanos , Enfermedad de Still del Adulto/complicaciones , Enfermedad de Still del Adulto/diagnóstico , Enfermedad de Still del Adulto/tratamiento farmacológico , Síndrome Hemolítico Urémico Atípico/complicaciones , Síndrome Hemolítico Urémico Atípico/inmunología , Factor H de Complemento/inmunología , Adulto , Masculino , Autoanticuerpos/sangre , Síndrome de Activación Macrofágica/diagnóstico , Síndrome de Activación Macrofágica/complicaciones , Síndrome de Activación Macrofágica/inmunología
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.
J Immunol ; 212(10): 1589-1601, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38558134

RESUMEN

Tumor-targeting Abs can be used to initiate an antitumor immune program, which appears essential to achieve a long-term durable clinical response to cancer. We previously identified an anti-complement factor H (CFH) autoantibody associated with patients with early-stage non-small cell lung cancer. We cloned from their peripheral B cells an mAb, GT103, that specifically recognizes CFH on tumor cells. Although the underlying mechanisms are not well defined, GT103 targets a conformationally distinct CFH epitope that is created when CFH is associated with tumor cells, kills tumor cells in vitro, and has potent antitumor activity in vivo. In the effort to better understand how an Ab targeting a tumor epitope can promote an effective antitumor immune response, we used the syngeneic CMT167 lung tumor C57BL/6 mouse model, and we found that murinized GT103 (mGT103) activates complement and enhances antitumor immunity through multiple pathways. It creates a favorable tumor microenvironment by decreasing immunosuppressive regulatory T cells and myeloid-derived suppressor cells, enhances Ag-specific effector T cells, and has an additive antitumor effect with anti-PD-L1 mAb. Furthermore, the immune landscape of tumors from early-stage patients expressing the anti-CFH autoantibody is associated with an immunologically active tumor microenvironment. More broadly, our results using an mAb cloned from autoantibody-expressing B cells provides novel, to our knowledge, mechanistic insights into how a tumor-specific, complement-activating Ab can generate an immune program to kill tumor cells and inhibit tumor growth.


Asunto(s)
Activación de Complemento , Ratones Endogámicos C57BL , Animales , Ratones , Humanos , Activación de Complemento/inmunología , Línea Celular Tumoral , Factor H de Complemento/inmunología , Microambiente Tumoral/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Autoanticuerpos/inmunología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Femenino , Linfocitos T Reguladores/inmunología
20.
Nephrology (Carlton) ; 29(8): 519-527, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38604995

RESUMEN

INTRODUCTION: Atypical haemolytic uremic syndrome (aHUS) is a rare form of thrombotic microangiopathy (TMA) associated with complement dysregulation; aHUS may be associated with other 'triggers' or 'clinical conditions'. This study aimed to characterize this patient population using data from the Global aHUS Registry, the largest collection of real-world data on patients with aHUS. METHODS: Patients enrolled in the Global aHUS Registry between April 2012 and June 2021 and with recorded aHUS-associated triggers or clinical conditions prior/up to aHUS onset were analysed. aHUS was diagnosed by the treating physician. Data were classified by age at onset of aHUS (< or ≥18 years) and additionally by the presence/absence of identified pathogenic complement genetic variant(s) and/or anti-complement factor H (CFH) antibodies. Genetically/immunologically untested patients were excluded. RESULTS: 1947 patients were enrolled in the Global aHUS Registry by June 2021, and 349 (17.9%) met inclusion criteria. 307/349 patients (88.0%) had a single associated trigger or clinical condition and were included in the primary analysis. Malignancy was most common (58/307, 18.9%), followed by pregnancy and acute infections (both 53/307, 17.3%). Patients with an associated trigger or clinical condition were generally more likely to be adults at aHUS onset. CONCLUSION: Our analysis suggests that aHUS-associated triggers or clinical conditions may be organized into clinically relevant categories, and their presence does not exclude the concurrent presence of pathogenic complement genetic variants and/or anti-CFH antibodies. Considering a diagnosis of aHUS with associated triggers or clinical conditions in patients presenting with TMA may allow faster and more appropriate treatment.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Sistema de Registros , Humanos , Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/epidemiología , Síndrome Hemolítico Urémico Atípico/diagnóstico , Síndrome Hemolítico Urémico Atípico/inmunología , Femenino , Masculino , Adulto , Persona de Mediana Edad , Adulto Joven , Adolescente , Embarazo , Factor H de Complemento/genética , Factor H de Complemento/inmunología , Niño , Neoplasias/epidemiología , Edad de Inicio , Preescolar , Factores de Riesgo , Anciano
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA