Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 17(7): e1009381, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34197564

RESUMEN

Clearance of viral infections, such as SARS-CoV-2 and influenza A virus (IAV), must be fine-tuned to eliminate the pathogen without causing immunopathology. As such, an aggressive initial innate immune response favors the host in contrast to a detrimental prolonged inflammation. The complement pathway bridges innate and adaptive immune system and contributes to the response by directly clearing pathogens or infected cells, as well as recruiting proinflammatory immune cells and regulating inflammation. However, the impact of modulating complement activation in viral infections is still unclear. In this work, we targeted the complement decay-accelerating factor (DAF/CD55), a surface protein that protects cells from non-specific complement attack, and analyzed its role in IAV infections. We found that DAF modulates IAV infection in vivo, via an interplay with the antigenic viral proteins hemagglutinin (HA) and neuraminidase (NA), in a strain specific manner. Our results reveal that, contrary to what could be expected, DAF potentiates complement activation, increasing the recruitment of neutrophils, monocytes and T cells. We also show that viral NA acts on the heavily sialylated DAF and propose that the NA-dependent DAF removal of sialic acids exacerbates complement activation, leading to lung immunopathology. Remarkably, this mechanism has no impact on viral loads, but rather on the host resilience to infection, and may have direct implications in zoonotic influenza transmissions.


Asunto(s)
Antígenos CD55/fisiología , Subtipo H1N1 del Virus de la Influenza A/aislamiento & purificación , Pulmón/inmunología , Viremia/inmunología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Antígenos CD55/química , Antígenos CD55/deficiencia , Quimiotaxis de Leucocito , Activación de Complemento , Glicoproteínas Hemaglutininas del Virus de la Influenza/fisiología , Adaptación al Huésped , Especificidad del Huésped , Interacciones Huésped-Patógeno , Subtipo H1N1 del Virus de la Influenza A/enzimología , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Subtipo H1N1 del Virus de la Influenza A/fisiología , Interferón gamma/análisis , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ácido N-Acetilneuramínico , Neuraminidasa/fisiología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Carga Viral , Proteínas Virales/fisiología , Virulencia , Replicación Viral , Pérdida de Peso
2.
Nat Immunol ; 22(2): 128-139, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33398182

RESUMEN

Complement hyperactivation, angiopathic thrombosis and protein-losing enteropathy (CHAPLE disease) is a lethal disease caused by genetic loss of the complement regulatory protein CD55, leading to overactivation of complement and innate immunity together with immunodeficiency due to immunoglobulin wasting in the intestine. We report in vivo human data accumulated using the complement C5 inhibitor eculizumab for the medical treatment of patients with CHAPLE disease. We observed cessation of gastrointestinal pathology together with restoration of normal immunity and metabolism. We found that patients rapidly renormalized immunoglobulin concentrations and other serum proteins as revealed by aptamer profiling, re-established a healthy gut microbiome, discontinued immunoglobulin replacement and other treatments and exhibited catch-up growth. Thus, we show that blockade of C5 by eculizumab effectively re-establishes regulation of the innate immune complement system to substantially reduce the pathophysiological manifestations of CD55 deficiency in humans.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Activación de Complemento/efectos de los fármacos , Complemento C5/antagonistas & inhibidores , Inactivadores del Complemento/uso terapéutico , Metabolismo Energético/efectos de los fármacos , Hipoproteinemia/tratamiento farmacológico , Inmunidad Innata/efectos de los fármacos , Enteropatías Perdedoras de Proteínas/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacocinética , Biomarcadores/sangre , Antígenos CD55/deficiencia , Antígenos CD55/genética , Complemento C5/metabolismo , Inactivadores del Complemento/efectos adversos , Inactivadores del Complemento/farmacocinética , Predisposición Genética a la Enfermedad , Humanos , Hipoproteinemia/genética , Hipoproteinemia/inmunología , Hipoproteinemia/metabolismo , Mutación , Fenotipo , Enteropatías Perdedoras de Proteínas/genética , Enteropatías Perdedoras de Proteínas/inmunología , Enteropatías Perdedoras de Proteínas/metabolismo , Resultado del Tratamiento
3.
Transgenic Res ; 30(1): 11-21, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33387103

RESUMEN

Decay accelerating factor (DAF), a key complement activation control protein, is a 70 kDa membrane bound glycoprotein which controls extent of formation of the C3 and C5 convertases by accelerating their decay. Using clustered regularly-interspaced short palindromic repeats, (CRISPR)/associated protein 9 (Cas9) genome editing we generated a novel DAF deficient (Daf-/-) rat model. The present study describes the renal and extrarenal phenotype of this model and assesses renal response to complement-dependent injury induced by administration of a complement-fixing antibody (anti-Fx1A) against the glomerular epithelial cell (podocyte). Rats generated were healthy, viable and able to reproduce normally. Complete absence of DAF was documented in renal as well as extra-renal tissues at both protein and mRNA level compared to Daf+/+ rats. Renal histology in Daf-/- rats showed no differences regarding glomerular or tubulointerstitial pathology compared to Daf+/+ rats. Moreover, there was no difference in urine protein excretion (ratio of urine albumin to creatinine) or in serum creatinine and urea levels. In Daf-/- rats, proteinuria was significantly increased following binding of anti-Fx1A antibody to podocytes while increased C3b deposition was observed. The DAF knock-out rat model developed validates the role of this complement cascade regulator in immune-mediated podocyte injury. Given the increasing role of dysregulated complement activation in various forms of kidney disease and the fact that the rat is the preferred animal for renal pathophysiology studies, the rat DAF deficient model may serve as a useful tool to study the role of this complement activation regulator in complement-dependent forms of kidney injury.


Asunto(s)
Lesión Renal Aguda/genética , Antígenos CD55/genética , Activación de Complemento/genética , Podocitos/metabolismo , Lesión Renal Aguda/patología , Albuminuria , Animales , Anticuerpos Antiidiotipos/farmacología , Antígenos CD55/deficiencia , Antígenos CD55/inmunología , Sistemas CRISPR-Cas/genética , Activación de Complemento/inmunología , Convertasas de Complemento C3-C5/genética , Complemento C5/genética , Técnicas de Inactivación de Genes , Complejo Antigénico de Nefritis de Heymann/genética , Complejo Antigénico de Nefritis de Heymann/inmunología , Humanos , Podocitos/patología , Ratas
5.
J Exp Med ; 217(9)2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32717081

RESUMEN

Kidney glomerulosclerosis commonly progresses to end-stage kidney failure, but pathogenic mechanisms are still poorly understood. Here, we show that podocyte expression of decay-accelerating factor (DAF/CD55), a complement C3 convertase regulator, crucially controls disease in murine models of adriamycin (ADR)-induced focal and segmental glomerulosclerosis (FSGS) and streptozotocin (STZ)-induced diabetic glomerulosclerosis. ADR induces enzymatic cleavage of DAF from podocyte surfaces, leading to complement activation. C3 deficiency or prevention of C3a receptor (C3aR) signaling abrogates disease despite DAF deficiency, confirming complement dependence. Mechanistic studies show that C3a/C3aR ligations on podocytes initiate an autocrine IL-1ß/IL-1R1 signaling loop that reduces nephrin expression, causing actin cytoskeleton rearrangement. Uncoupling IL-1ß/IL-1R1 signaling prevents disease, providing a causal link. Glomeruli of patients with FSGS lack DAF and stain positive for C3d, and urinary C3a positively correlates with the degree of proteinuria. Together, our data indicate that the development and progression of glomerulosclerosis involve loss of podocyte DAF, triggering local, complement-dependent, IL-1ß-induced podocyte injury, potentially identifying new therapeutic targets.


Asunto(s)
Antígenos CD55/metabolismo , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Podocitos/metabolismo , Podocitos/patología , Citoesqueleto de Actina/metabolismo , Anciano , Animales , Antígenos CD55/deficiencia , Línea Celular Transformada , Activación de Complemento/inmunología , Complemento C3b/metabolismo , Diabetes Mellitus Experimental/patología , Susceptibilidad a Enfermedades , Regulación hacia Abajo , Doxorrubicina/efectos adversos , Femenino , Glomeruloesclerosis Focal y Segmentaria/inducido químicamente , Glomeruloesclerosis Focal y Segmentaria/inmunología , Humanos , Interleucina-1beta/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Especificidad de Órganos , Fosfolipasa D/metabolismo , Podocitos/ultraestructura , Receptores de Complemento/metabolismo , Transducción de Señal
6.
Br J Haematol ; 189(5): 954-966, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32103498

RESUMEN

A retrospective analysis of presentation clinical, laboratory and immunophenotypic features of 1 081 patients with paroxysmal nocturnal haemoglobinuria (PNH) clones [glycosylphosphatidylinositol (GPI)-deficient blood cells] identified at our hospital by flow cytometry over the past 25 years was undertaken. Three distinct clusters of patients were identified and significant correlations between presentation disease type and PNH clone sizes were evident. Smaller PNH clones predominate in cytopenic and myelodysplastic subtypes; large PNH clones were associated with haemolytic, thrombotic and haemolytic/thrombotic subtypes. Rare cases with an associated chronic myeloproliferative disorder had either large or small PNH clones. Cytopenia was a frequent finding, highlighting bone marrow failure as the major underlying feature associated with the detection of PNH clones in the peripheral blood. Red cell PNH clones showed significant correlations between the presence of type II (partial GPI deficiency) red cells and thrombotic disease. Haemolytic PNH was associated with type III (complete GPI deficiency) red cell populations of >20%. Those with both haemolytic and thrombotic features had major type II and type III red cell populations. Distinct patterns of presentation age decade were evident for clinical subtypes with a peak incidence of haemolytic PNH in the 30-49 year age group and a biphasic age distribution for the cytopenia group.


Asunto(s)
Glicosilfosfatidilinositoles/deficiencia , Hemoglobinuria Paroxística/sangre , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anemia Aplásica/etiología , Anemia Hemolítica/etiología , Antígenos CD55/deficiencia , Antígenos CD59/deficiencia , Niño , Preescolar , Evolución Clonal , Células Clonales/patología , Progresión de la Enfermedad , Femenino , Citometría de Flujo , Hemoglobinuria Paroxística/complicaciones , Hemoglobinuria Paroxística/genética , Hemoglobinuria Paroxística/patología , Humanos , Inmunofenotipificación , Lactante , Linfocitos/patología , Masculino , Persona de Mediana Edad , Trastornos Mieloproliferativos/etiología , Neutrófilos/patología , Receptores de Transferrina/sangre , Estudios Retrospectivos , Trombosis/etiología , Adulto Joven
7.
J Pediatr Gastroenterol Nutr ; 68(3): 325-333, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30418410

RESUMEN

OBJECTIVES: Loss of the complement inhibitor CD55 leads to a syndrome of early-onset protein-losing enteropathy (PLE), associated with intestinal lymphangiectasia and susceptibility to large-vein thrombosis. The in vitro and short-term treatment benefits of eculizumab (C5-inhibitor) therapy for CD55-deficiency have been previously demonstrated. Here we present the 18-months treatment outcomes for 3 CD55-deficiency patients with sustained therapeutic response. METHODS: Three CD55-deficiency patients received off-label eculizumab treatment. Clinical and laboratory treatment outcomes included frequency and consistency of bowl movements, weight, patient/parent reports of overall well-being, and serum albumin and total protein levels. Membrane attack complex deposition on leukocytes was tested by flow cytometry, before and during eculizumab treatment. RESULTS: Marked clinical improvement was noted in all 3 patients with resolution of PLE manifestations, that is, diarrhea, edema, malabsorption, overall well-being, growth, and quality of life. In correlation with the clinical observations, we observed progress in all laboratory outcome parameters, including increase in albumin and total protein levels, and up to 80% reduction in membrane attack complex deposition on leukocytes (P < 0.001). The progress persisted over 18 months of treatment without any severe adverse events. CONCLUSIONS: CD55-deficiency patients present with early-onset diarrhea, edema, severe hypoalbuminemia, abdominal pain, and malnutrition. Targeted therapy with the terminal complement inhibitor eculizumab has positive clinical and laboratory outcomes in PLE related to CD55 loss-of-function mutations, previously a life-threatening condition. Our results demonstrate the potential of genetic diagnosis to guide tailored treatment, and underscore the significant role of the complement system in the intestine.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Antígenos CD55/deficiencia , Inactivadores del Complemento/administración & dosificación , Enteropatías Perdedoras de Proteínas/tratamiento farmacológico , Adulto , Niño , Preescolar , Ensayos de Uso Compasivo , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Estudios de Seguimiento , Humanos , Infusiones Intravenosas , Linfangiectasia Intestinal/complicaciones , Linfangiectasia Intestinal/patología , Uso Fuera de lo Indicado , Estudios Prospectivos , Enteropatías Perdedoras de Proteínas/etiología , Calidad de Vida , Inducción de Remisión
8.
J Immunol Methods ; 456: 15-22, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29447841

RESUMEN

BACKGROUND: To prevent unwanted complement activation and subsequent damage, complement activation must be tightly regulated on healthy host cells. Dysregulation of the complement system contributes to the pathology of diseases like Paroxysmal Nocturnal Hemoglobinuria and atypical Hemolytic Uremic Syndrome. To investigate complement regulator deficiencies, primary patient cells may be used, but access to patient cells may be limited and cells are heterogeneous between different patients. To inhibit regulator function on healthy host cells, blocking antibodies can be used, though it may be difficult to exclude antibody-mediated effects. To circumvent these issues, we created single and combined complement regulator human knockout cells to be able to in vitro investigate complement activation and regulation on human cells. METHODS: CRISPR/Cas9 was used to knockout (KO) complement regulatory proteins CD46, CD55 and/or CD59 in human HAP1 cells. Single cell derived cell lines were profiled by Sanger sequencing and flow cytometry. To confirm the lack of complement regulatory function, the cells were exposed to complement in normal human serum and subsequently C3 and C4 deposition on the cell surface were detected by using flow cytometry. RESULTS: We created single KO cell lines that completely lacked CD46, CD55 or CD59. We additionally generated double CD46/CD55, CD46/CD59 and CD55/CD59 KOs and triple CD46/CD55/CD59 KOs. Upon classical pathway activation, deletion of CD46 resulted in increased C3 and C4 deposition, while deleting CD55 mainly resulted to increased C3 deposition, confirming their reported function in complement regulation. Upon alternative pathway activation, C3 deposition was only observed on the triple CD46/CD55/CD59 KO cells and not on any of the other cell lines, suggesting that human cells are resistant to spontaneous complement activation and suggesting a role for CD59 in C3 regulation. CONCLUSIONS: The generation of complement regulator KO cell lines provides a relevant tool for future in vitro investigations of complement activation and regulation on human cells. Furthermore, these cell lines may also be helpful to evaluate therapeutic complement inhibitors and may shed light on novel roles of complement regulatory proteins as we here observed for CD59.


Asunto(s)
Antígenos CD55/deficiencia , Antígenos CD59/deficiencia , Sistemas CRISPR-Cas/genética , Proteínas del Sistema Complemento/genética , Proteínas del Sistema Complemento/inmunología , Edición Génica , Técnicas de Inactivación de Genes , Proteína Cofactora de Membrana/deficiencia , Línea Celular , Voluntarios Sanos , Humanos , Masculino
9.
Int J Lab Hematol ; 40(2): 128-135, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28963754

RESUMEN

INTRODUCTION: Hereditary haemolytic anaemias (HHA) encompass a heterogeneous group of anaemias characterized by decreased red blood cell survival. The aim of this study was to evaluate the status of red blood cell (RBC) surface molecules known or previously proposed to participate in preventing premature RBC clearance, analysing erythrocytes from patients with two types of HHA: hereditary spherocytosis (HS) and microcytosis. MATERIAL/METHODS: Relative binding of five monoclonal antibodies (mAbs), anti-CD55, anti-CD59, anti-CD44, anti-CD47 and anti-CD58, was evaluated in erythrocytes of patients with HS and hereditary microcytosis, using flow cytometry. The amount of CD55 protein was assessed by semi-quantitative Western blots densitometry analysis. RESULTS: The majority of both HS and microcytic patients demonstrated significant reduction of anti-CD55 binding by erythrocytes (average 23% and 19%, respectively, P < .001), with no concomitant anti-CD59-binding deficiency. Anti-CD44, anti-CD47 and anti-CD58 binding was within the healthy control range or was slightly decreased. CONCLUSIONS: This study provides evidence supporting the presence of erythrocytes deficient in CD55 presentation in HS and hereditary microcytosis. Moreover, deficiency of CD55 antigen presentation on RBC does not correlate with the amount of CD55 in RBC membrane. Further studies using molecular techniques will clarify the exact participation of CD55 deficiency in premature RBC clearance in HHA.


Asunto(s)
Anemia Hemolítica Congénita/sangre , Antígenos CD55/análisis , Eritrocitos/metabolismo , Anticuerpos Monoclonales/inmunología , Antígenos CD55/deficiencia , Antígenos CD55/inmunología , Membrana Eritrocítica/metabolismo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...