Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Transl Med ; 21(1): 864, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-38017492

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive cancers with a very low survival rate at 5 years. The use of chemotherapeutic agents results in only modest prolongation of survival and is generally associated with the occurrence of toxicity effects. Antibody-based immunotherapy has been proposed for the treatment of PDAC, but its efficacy has so far proved limited. The proteoglycan glypican-1 (GPC1) may be a useful immunotherapeutic target because it is highly expressed on the surface of PDAC cells, whereas it is not expressed or is expressed at very low levels in benign neoplastic lesions, chronic pancreatitis, and normal adult tissues. Here, we developed and characterized a specific mouse IgM antibody (AT101) targeting GPC1. METHODS: We developed a mouse monoclonal antibody of the IgM class directed against an epitope of GPC1 in close proximity to the cell membrane. For this purpose, a 46 amino acid long peptide of the C-terminal region was used to immunize mice by an in-vivo electroporation protocol followed by serum titer and hybridoma formation. RESULTS: The ability of AT101 to bind the GPC1 protein was demonstrated by ELISA, and by flow cytometry and immunofluorescence analysis in the GPC1-expressing "PDAC-like" BXPC3 cell line. In-vivo experiments in the BXPC3 xenograft model showed that AT101 was able to bind GPC1 on the cell surface and accumulate in the BXPC3 tumor masses. Ex-vivo analyses of BXPC3 tumor masses showed that AT101 was able to recruit immunological effectors (complement system components, NK cells, macrophages) to the tumor site and damage PDAC tumor tissue. In-vivo treatment with AT101 reduced tumor growth and prolonged survival of mice with BXPC3 tumor (p < 0.0001). CONCLUSIONS: These results indicate that AT101, an IgM specific for an epitope of GPC1 close to PDAC cell surface, is a promising immunotherapeutic agent for GPC1-expressing PDAC, being able to selectively activate the complement system and recruit effector cells in the tumor microenvironment, thus allowing to reduce tumor mass growth and improve survival in treated mice.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Adulto , Humanos , Ratones , Animales , Glipicanos/metabolismo , Glipicanos/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Inmunoterapia , Epítopos , Inmunoglobulina M , Línea Celular Tumoral , Microambiente Tumoral
2.
Haematologica ; 108(7): 1861-1872, 2023 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-36172817

RESUMEN

ß2-glycoprotein I (ß2-GPI) is a serum protein widely recognized as the main target of antibodies present in patients with antiphospholipid syndrome (APS). ß2-GPI binds to activated endothelial cells, platelets and leukocytes, key players in thrombus formation. We developed a new targeted thrombolytic agent consisting of nanobubbles (NB) coated with recombinant tissue plasminogen activator (rtPA) and a recombinant antibody specific for cell-bound ß2-GPI. The therapeutic efficacy of targeted NB was evaluated in vitro, using platelet-rich blood clots, and in vivo in three different animal models: i) thrombosis developed in a rat model of APS; ii) ferric chloride-induced mesenteric thrombosis in rats, and iii) thrombotic microangiopathy in a mouse model of atypical hemolytic uremic syndrome (C3-gain-of-function mice). Targeted NB bound preferentially to platelets and leukocytes within thrombi and to endothelial cells through ß2-GPI expressed on activated cells. In vitro, rtPA-targeted NB (rtPA-tNB) induced greater lysis of platelet-rich blood clots than untargeted NB. In a rat model of APS, administration of rtPA-tNB caused rapid dissolution of thrombi and, unlike soluble rtPA that induced transient thrombolysis, prevented new thrombus formation. In a rat model of ferric chloride triggered thrombosis, rtPA-tNB, but not untargeted NB and free rtPA, induced rapid and persistent recanalization of occluded vessels. Finally, treatment of C3-gain-of-function mice with rtPA-tNB, that target ß2-GPI deposited in kidney glomeruli, decreased fibrin deposition, and improved urinalysis data with a greater efficiency than untargeted NB. Our findings suggest that targeting cell-bound ß2-GPI may represent an efficient and thrombus-specific thrombolytic strategy in both APS-related and APS-unrelated thrombotic conditions.


Asunto(s)
Síndrome Antifosfolípido , Tromboembolia , Trombosis , Animales , Ratones , Ratas , Fibrinolíticos/farmacología , Fibrinolíticos/uso terapéutico , Activador de Tejido Plasminógeno/farmacología , Activador de Tejido Plasminógeno/uso terapéutico , beta 2 Glicoproteína I , Células Endoteliales , Trombosis/tratamiento farmacológico , Trombosis/etiología
3.
J Nanobiotechnology ; 21(1): 376, 2023 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-37838659

RESUMEN

BACKGROUND: Nanoparticles represent one of the most important innovations in the medical field. Among nanocarriers, polymeric nanoparticles (PNPs) attracted much attention due to their biodegradability, biocompatibility, and capacity to increase efficacy and safety of encapsulated drugs. Another important improvement in the use of nanoparticles as delivery systems is the conjugation of a targeting agent that enables the nanoparticles to accumulate in a specific tissue. Despite these advantages, the clinical translation of therapeutic approaches based on nanoparticles is prevented by their interactions with blood proteins. In fact, the so-formed protein corona (PC) drastically alters the biological identity of the particles. Adsorbed activated proteins of the complement cascade play a pivotal role in the clearance of nanoparticles, making them more easily recognized by macrophages, leading to their rapid elimination from the bloodstream and limiting their efficacy. Since the mouse is the most used preclinical model for human disease, this work compared human and mouse PC formed on untargeted PNPs (uPNPs) and targeted PNPs (tPNPs), paying particular attention to complement activation. RESULTS: Mouse and human serum proteins adsorbed differently to PNPs. The differences in the binding of mouse complement proteins are minimal, whereas human complement components strongly distinguish the two particles. This is probably due to the human origin of the Fc portion of the antibody used as targeting agent on tPNPs. tPNPs and uPNPs mainly activate complement via the classical and alternative pathways, respectively, but this pattern did not affect their binding and internalization in macrophages and only a limited consumption of the activity of the human complement system was documented. CONCLUSIONS: The results clearly indicate the presence of complement proteins on PNPs surface but partially derived from an unspecific deposition rather than an effective complement activation. The presence of a targeting antibody favors the activation of the classical pathway, but its absence allows an increased activation of the alternative pathway. This results in similar opsonization of both PNPs and similar phagocytosis by macrophages, without an impairment of the activity of circulating complement system and, consequently, not enhancing the susceptibility to infection.


Asunto(s)
Nanopartículas , Corona de Proteínas , Humanos , Ratones , Animales , Opsonización , Proteínas del Sistema Complemento/metabolismo , Anticuerpos , Polímeros
4.
J Biol Chem ; 297(2): 100890, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34197876

RESUMEN

ß2-glycoprotein I (ß2GPI) is an abundant multidomain plasma protein that plays various roles in the clotting and complement cascades. It is also the main target of antiphospholipid antibodies (aPL) in the acquired coagulopathy known as antiphospholipid syndrome (APS). Previous studies have shown that ß2GPI adopts two interconvertible biochemical conformations, oxidized and reduced, depending on the integrity of the disulfide bonds. However, the precise contribution of the disulfide bonds to ß2GPI structure and function is unknown. Here, we substituted cysteine residues with serine to investigate how the disulfide bonds C32-C60 in domain I (DI) and C288-C326 in domain V (DV) regulate ß2GPI's structure and function. Results of our biophysical and biochemical studies support the hypothesis that the C32-C60 disulfide bond plays a structural role, whereas the disulfide bond C288-C326 is allosteric. We demonstrate that absence of the C288-C326 bond, unlike absence of the C32-C60 bond, diminishes membrane binding without affecting the thermodynamic stability and overall structure of the protein, which remains elongated in solution. We also document that, while absence of the C32-C60 bond directly impairs recognition of ß2GPI by pathogenic anti-DI antibodies, absence of the C288-C326 disulfide bond is sufficient to abolish complex formation in the presence of anionic phospholipids. We conclude that the disulfide bond C288-C326 operates as a molecular switch capable of regulating ß2GPI's physiological functions in a redox-dependent manner. We propose that in APS patients with anti-DI antibodies, selective rupture of the C288-C326 disulfide bond may be a valid strategy to lower the pathogenic potential of aPL.


Asunto(s)
Anticuerpos Antifosfolípidos/inmunología , Síndrome Antifosfolípido/inmunología , Autoanticuerpos/inmunología , Proteínas Recombinantes/metabolismo , beta 2 Glicoproteína I/metabolismo , Regulación Alostérica , Anticuerpos Antifosfolípidos/sangre , Síndrome Antifosfolípido/patología , Autoanticuerpos/sangre , Línea Celular , Cristalografía por Rayos X/métodos , Humanos , Oxidación-Reducción , Dominios Proteicos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , beta 2 Glicoproteína I/química , beta 2 Glicoproteína I/inmunología , beta 2 Glicoproteína I/aislamiento & purificación
5.
J Immunol ; 205(5): 1385-1392, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32759297

RESUMEN

ß2-Glycoprotein I (ß2-GPI) is an abundant plasma glycoprotein with unknown physiological function and is currently recognized as the main target of antiphospholipid Abs responsible for complement activation and vascular thrombosis in patients with antiphospholipid syndrome (APS). In this study, we provide evidence that mannose-binding lectin (MBL) binds to ß2-GPI in Ca++ and a dose-dependent manner and that this interaction activates complement and promotes complement-dependent thrombin generation. Surprisingly, a significant binding was observed between MBL and isolated domains II and IV of ß2-GPI, whereas the carbohydrate chains, domain I and domain V, were not involved in the interaction, documenting a noncanonical binding mode between MBL and ß2-GPI. Importantly, this interaction may occur on endothelial cells because binding of MBL to ß2-GPI was detected on the surface of HUVECs, and colocalization of MBL with ß2-GPI was observed on the endothelium of a biopsy specimen of a femoral artery from an APS patient. Because ß2-GPI-mediated MBL-dependent thrombin generation was increased after priming the endothelium with TNF-α, our data suggests that this mechanism could play an important yet unrecognized role under physiological conditions and may be upregulated in pathological situations. Moreover, the complement activation and the procoagulant effects of the ß2-GPI/MBL complex may contribute to amplify similar activities of anti-ß2-GPI Abs in APS and possibly act independently of Abs, raising the issue of developing appropriate therapies to avoid recurrences and disability in patients at risk for these clinical conditions.


Asunto(s)
Activación de Complemento/inmunología , Lectina de Unión a Manosa/metabolismo , Trombina/metabolismo , beta 2 Glicoproteína I/metabolismo , Síndrome Antifosfolípido/inmunología , Síndrome Antifosfolípido/metabolismo , Calcio/metabolismo , Línea Celular , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Endotelio/inmunología , Endotelio/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lectina de Unión a Manosa/inmunología , Unión Proteica/inmunología , Trombina/inmunología , Trombosis/inmunología , Trombosis/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , beta 2 Glicoproteína I/inmunología
6.
Int J Mol Sci ; 23(17)2022 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-36077433

RESUMEN

Hepatocellular carcinoma (HCC) is the second most lethal tumor, with a 5-year survival rate of 18%. Early stage HCC is potentially treatable by therapies with curative intent, whereas chemoembolization/radioembolization and systemic therapies are the only therapeutic options for intermediate or advanced HCC. Drug resistance is a critical obstacle in the treatment of HCC that could be overcome by the use of targeted nanoparticle-based therapies directed towards specific tumor-associated antigens (TAAs) to improve drug delivery. Glypican 3 (GPC3) is a member of the glypican family, heparan sulfate proteoglycans bound to the cell surface via a glycosylphosphatidylinositol anchor. The high levels of GPC3 detected in HCC and the absence or very low levels in normal and non-malignant liver make GPC3 a promising TAA candidate for targeted nanoparticle-based therapies. The use of nanoparticles conjugated with anti-GPC3 agents may improve drug delivery, leading to a reduction in severe side effects caused by chemotherapy and increased drug release at the tumor site. In this review, we describe the main clinical features of HCC and the common treatment approaches. We propose the proteoglycan GPC3 as a useful TAA for targeted therapies. Finally, we describe nanotechnology approaches for anti-GPC3 drug delivery systems based on NPs for HCC treatment.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Resistencia a Medicamentos , Glipicanos/metabolismo , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Nanotecnología , Terapias en Investigación
7.
Int J Mol Sci ; 23(18)2022 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-36142190

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) accounts for 90% of all pancreatic cancers, with a 5-year survival rate of 7% and 80% of patients diagnosed with advanced or metastatic malignancies. Despite recent advances in diagnostic testing, surgical techniques, and systemic therapies, there remain limited options for the effective treatment of PDAC. There is an urgent need to develop targeted therapies that are able to differentiate between cancerous and non-cancerous cells to reduce side effects and better inhibit tumor growth. Antibody-targeted strategies are a potentially effective option for introducing innovative therapies. Antibody-based immunotherapies and antibody-conjugated nanoparticle-based targeted therapies with antibodies targeting specific tumor-associated antigens (TAA) can be proposed. In this context, glypican-1 (GPC1), which is highly expressed in PDAC and not expressed or expressed at very low levels in non-malignant lesions and healthy pancreatic tissues, is a useful TAA that can be achieved by a specific antibody-based immunotherapy and antibody-conjugated nanoparticle-based targeted therapy. In this review, we describe the main clinical features of PDAC. We propose the proteoglycan GPC1 as a useful TAA for PDAC-targeted therapies. We also provide a digression on the main developed approaches of antibody-based immunotherapy and antibody-conjugated nanoparticle-based targeted therapy, which can be used to target GPC1.


Asunto(s)
Carcinoma Ductal Pancreático , Inmunoconjugados , Neoplasias Pancreáticas , Carcinoma Ductal Pancreático/tratamiento farmacológico , Glipicanos , Humanos , Inmunoconjugados/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Proteoglicanos , Neoplasias Pancreáticas
8.
J Biol Chem ; 295(31): 10794-10806, 2020 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-32518155

RESUMEN

ß2-Glycoprotein I (ß2GPI) is an abundant plasma protein displaying phospholipid-binding properties. Because it binds phospholipids, it is a target of antiphospholipid antibodies (aPLs) in antiphospholipid syndrome (APS), a life-threatening autoimmune thrombotic disease. Indeed, aPLs prefer membrane-bound ß2GPI to that in solution. ß2GPI exists in two almost equally populated redox states: oxidized, in which all the disulfide bonds are formed, and reduced, in which one or more disulfide bonds are broken. Furthermore, ß2GPI can adopt multiple conformations (i.e. J-elongated, S-twisted, and O-circular). While strong evidence indicates that the J-form is the structure bound to aPLs, which conformation exists and predominates in solution remains controversial, and so is the conformational pathway leading to the bound state. Here, we report that human recombinant ß2GPI purified under native conditions is oxidized. Moreover, under physiological pH and salt concentrations, this oxidized form adopts a J-elongated, flexible conformation, not circular or twisted, in which the N-terminal domain I (DI) and the C-terminal domain V (DV) are exposed to the solvent. Consistent with this model, binding kinetics and mutagenesis experiments revealed that in solution the J-form interacts with negatively charged liposomes and with MBB2, a monoclonal anti-DI antibody that recapitulates most of the features of pathogenic aPLs. We conclude that the preferential binding of aPLs to phospholipid-bound ß2GPI arises from the ability of its preexisting J-form to accumulate on the membranes, thereby offering an ideal environment for aPL binding. We propose that targeting the J-form of ß2GPI provides a strategy to block pathogenic aPLs in APS.


Asunto(s)
Anticuerpos Antifosfolípidos/química , Síndrome Antifosfolípido , beta 2 Glicoproteína I/química , Animales , Anticuerpos Antifosfolípidos/metabolismo , Cricetinae , Células HEK293 , Humanos , Cinética , Mutagénesis , Dominios Proteicos , beta 2 Glicoproteína I/metabolismo
9.
J Autoimmun ; 124: 102728, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34592707

RESUMEN

Extremely rare reactions characterized by thrombosis and thrombocytopenia have been described in subjects that received ChAdOx1 nCoV-19 vaccination 5-16 days earlier. Although patients with vaccine-induced thrombotic thrombocytopenia (VITT) have high levels of antibodies to platelet factor 4 (PF4)-polyanion complexes, the exact mechanism of the development of thrombosis is still unknown. Here we reported serum studies as well as proteomics and genomics analyses demonstrating a massive complement activation potentially linked to the presence of anti-PF4 antibodies in a patient with severe VITT. At admission, complement activity of the classical and lectin pathways were absent (0% for both) with normal levels of the alternative pathway (73%) in association with elevated levels of the complement activation marker sC5b-9 (630 ng/mL [n.v. 139-462 ng/mL]) and anti-PF4 IgG (1.918 OD [n.v. 0.136-0.300 OD]). The immunoblotting analysis of C2 showed the complete disappearance of its normal band at 110 kDa. Intravenous immunoglobulin treatment allowed to recover complement activity of the classical pathway (91%) and lectin pathway (115%), to reduce levels of sC5b-9 (135 ng/mL) and anti-PF4 IgG (0.681 OD) and to normalize the C2 pattern at immunoblotting. Proteomics and genomics analyses in addition to serum studies showed that the absence of complement activity during VITT was not linked to alterations of the C2 gene but rather to a strong complement activation leading to C2 consumption. Our data in a single patient suggest monitoring complement parameters in other VITT patients considering also the possibility to target complement activation with specific drugs.


Asunto(s)
Vacunas contra la COVID-19/efectos adversos , COVID-19/prevención & control , Complemento C2 , Complejo de Ataque a Membrana del Sistema Complemento , Vía Clásica del Complemento , Lectina de Unión a Manosa de la Vía del Complemento , Púrpura Trombocitopénica Trombótica , SARS-CoV-2 , Adulto , Autoanticuerpos/sangre , Vacunas contra la COVID-19/administración & dosificación , ChAdOx1 nCoV-19 , Complemento C2/genética , Complemento C2/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Vía Clásica del Complemento/efectos de los fármacos , Vía Clásica del Complemento/genética , Lectina de Unión a Manosa de la Vía del Complemento/efectos de los fármacos , Lectina de Unión a Manosa de la Vía del Complemento/genética , Femenino , Humanos , Factor Plaquetario 4/sangre , Púrpura Trombocitopénica Trombótica/sangre , Púrpura Trombocitopénica Trombótica/inducido químicamente , Púrpura Trombocitopénica Trombótica/genética
10.
Medicina (Kaunas) ; 56(3)2020 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-32164349

RESUMEN

Complement deficiencies are rare and often underdiagnosed primary immunodeficiencies that may be associated with invasive bacterial diseases. Serious infections with encapsulated organisms (mainly Streptococcus pneumoniae, but also Neisseria meningitides and Haemophilus influenzae type B) are frequent in patients with a deficiency of the second component of complement (C2), but no data are available on long-term follow-up. This study aimed to evaluate the long-term clinical outcome and the importance of an early diagnosis and subsequent infection prophylaxis in C2 deficiency. Here, we report the 21-year follow-up of a whole family which was tested for complement parameters, genetic analysis and biochemical measurements, due to recurrent pneumococcal meningitis in the elder brother. The two sons were diagnosed with homozygous type 1 C2 deficiency, while their parents were heterozygous with normal complement parameters. For the two brothers, a recommended vaccination program and antibiotic prophylaxis were prescribed. During the long-term follow-up, no severe/invasive infections were observed in either patient. At the age of 16, the younger brother developed progressive hypogammaglobulinemia of all three classes, IgA, IgM and IgG. A next generation sequencing panel excluded the presence of gene defects related to primary antibody deficiencies. Our data show that early diagnosis, use of vaccinations and antibiotic prophylaxis may allow a normal life in hereditary C2 deficiency, which can be characterized using functional and genetic methods. Moreover, a periodical check of immunoglobulin serum levels could be useful to detect a possible hypogammaglobulinemia.


Asunto(s)
Cuidados Posteriores/métodos , Complemento C2/análisis , Familia , Enfermedades Genéticas Congénitas/diagnóstico , Adolescente , Adulto , Niño , Preescolar , Complemento C2/deficiencia , Femenino , Enfermedades Genéticas Congénitas/genética , Hospitales Universitarios/organización & administración , Hospitales Universitarios/estadística & datos numéricos , Humanos , Italia , Masculino
11.
J Autoimmun ; 103: 102288, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31213399

RESUMEN

Despite the advances in the treatment of rheumatoid arthritis (RA) achieved in the last few years, several patients are diagnosed late, do not respond to or have to stop therapy because of inefficacy and/or toxicity, leaving still a huge unmet need. Tissue-specific strategies have the potential to address some of these issues. The aim of the study is the development of a safe nanotechnology approach for tissue-specific delivery of drugs and diagnostic probes. CD34 + endothelial precursors were addressed in inflamed synovium using targeted biodegradable nanoparticles (tBNPs). These nanostructures were made of poly-lactic acid, poly-caprolactone, and PEG and then coated with a synovial homing peptide. Immunofluorescence analysis clearly demonstrated their capacity to selectively address CD34 + endothelial cells in synovial tissue obtained from human, mouse, and rat. Biodistribution studies in two different animal models of rheumatoid arthritis (antigen-induced arthritis/AIA and collagen-induced arthritis/CIA) confirmed the selective accumulation in inflamed joints but also evidenced the capacity of tBNP to detect early phases of the disease and the preferential liver elimination. The therapeutic effect of methotrexate (MTX)-loaded tBNPs were studied in comparison with conventional MTX doses. MTX-loaded tBNPs prevented and treated CIA and AIA at a lower dose and reduced administration frequency than MTX. Moreover, MTX-loaded tBNP showed a novel mechanism of action, in which the particles target and kill CD34 + endothelial progenitors, preventing neo-angiogenesis and, consequently, synovial inflammation. tBNPs represent a stable and safe platform to develop highly-sensitive imaging and therapeutic approaches in RA targeting specifically synovial neo-angiogenesis to reduce local inflammation.


Asunto(s)
Artritis Reumatoide/terapia , Células Endoteliales/inmunología , Inflamación/terapia , Metotrexato/uso terapéutico , Nanopartículas/uso terapéutico , Membrana Sinovial/inmunología , Membrana Sinovial/patología , Animales , Antígenos CD34/metabolismo , Modelos Animales de Enfermedad , Humanos , Nanopartículas/química , Neovascularización Patológica , Poliésteres/química , Ratas , Ratas Wistar
12.
Haematologica ; 104(4): 819-826, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30442725

RESUMEN

Clinical studies have reported different diagnostic/predictive values of antibodies to domain 1 or 4/5 of ß2glycoproteinI in terms of risk of thrombosis and pregnancy complications in patients with antiphospholipid syndrome. To obtain direct evidence for the pathogenic role of anti-domain 1 or anti-domain 4/5 antibodies, we analyzed the in vivo pro-coagulant effect of two groups of 5 sera IgG each reacting selectively with domain 1 or domain 5 in lipopolysaccharide (LPS)-treated rats. Antibody-induced thrombus formation in mesenteric vessels was followed by intravital microscopy, and vascular deposition of ß2glycoproteinI, human IgG and C3 was analyzed by immunofluorescence. Five serum IgG with undetectable anti-ß2glycoproteinI antibodies served as controls. All the anti-domain 1-positive IgG exhibited potent pro-coagulant activity while the anti-domain 5-positive and the negative control IgG failed to promote blood clot and vessel occlusion. A stronger granular deposit of IgG/C3 was found on the mesenteric endothelium of rats treated with anti-domain 1 antibodies, as opposed to a mild linear IgG staining and absence of C3 observed in rats receiving anti-domain 5 antibodies. Purified anti-domain 5 IgG, unlike anti-domain 1 IgG, did not recognize cardiolipin-bound ß2glycoproteinI while being able to interact with fluid-phase ß2glycoproteinI. These findings may explain the failure of anti-domain 5 antibodies to exhibit a thrombogenic effect in vivo, and the interaction of these antibodies with circulating ß2glycoproteinI suggests their potential competitive role with the pro-coagulant activity of anti-domain 1 antibodies. These data aim at better defining "really at risk" patients for more appropriate treatments to avoid recurrences and disability.


Asunto(s)
Síndrome Antifosfolípido , Autoanticuerpos , Inmunoglobulina G , Isquemia Mesentérica , beta 2 Glicoproteína I , Animales , Síndrome Antifosfolípido/sangre , Síndrome Antifosfolípido/inmunología , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Complemento C3/inmunología , Complemento C3/metabolismo , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Lipopolisacáridos/toxicidad , Masculino , Isquemia Mesentérica/sangre , Isquemia Mesentérica/inducido químicamente , Isquemia Mesentérica/inmunología , Dominios Proteicos , Ratas , Ratas Wistar , beta 2 Glicoproteína I/sangre , beta 2 Glicoproteína I/inmunología
14.
J Shoulder Elbow Surg ; 27(10): 1837-1844, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30139682

RESUMEN

BACKGROUND: The early diagnosis of suspected periprosthetic low-grade infections in shoulder arthroplasties is important for the outcome of the revision surgical procedures. The aim of this study was to investigate new biomarkers of infection in revision shoulder arthroplasties, taking into account the implant design, patient age, and comorbidities. METHODS: The study included 33 patients with shoulder arthroplasties undergoing revision surgical procedures. Microbiological diagnostic testing was performed in all cases. C-reactive protein serum levels and white blood cell counts were evaluated, and the periprosthetic tissue was stained immunohistologically for the terminal complement pathway components (C3, C5, and C9) and for CD68 and α-defensin. RESULTS: Microbiological diagnostic testing detected a periprosthetic infection in 10 reverse shoulder arthroplasties and in 4 anatomic shoulder arthroplasties, while the remaining 19 shoulder arthroplasties were classified as aseptic. We observed more Staphylococcus epidermidis infections in reverse shoulder arthroplasties and more Staphylococcus aureus infections in anatomic shoulder arthroplasties. The revision rate correlated with pre-existing comorbidities and number of previous surgical procedures. The C-reactive protein values and the incidence of specific periprosthetic radiolucent lines were significantly increased in septic revision cases. We found increased staining for all tested complement factors (C3, C5, and C9) but not for α-defensin and CD68 in septic tissue. The most interesting finding was that C9 separated septic from aseptic tissue with a predictive specificity of 100% and a sensitivity of 88.89%. CONCLUSION: We observed a strong correlation between C9 expressions in septic revision tissue. We propose that the terminal complement pathway, especially C9 deposition, may be a potential biomarker to identify septic complications using tissue biopsy specimens.


Asunto(s)
Artroplastía de Reemplazo de Hombro/efectos adversos , Proteína C-Reactiva/metabolismo , Infecciones Relacionadas con Prótesis/diagnóstico , Infecciones Relacionadas con Prótesis/metabolismo , Infecciones Estafilocócicas/diagnóstico , Infecciones Estafilocócicas/metabolismo , Anciano , Anciano de 80 o más Años , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Artroplastía de Reemplazo de Hombro/métodos , Biomarcadores/metabolismo , Complemento C3/metabolismo , Complemento C5/metabolismo , Complemento C9/metabolismo , Vía Alternativa del Complemento , Humanos , Recuento de Leucocitos , Persona de Mediana Edad , Prótesis e Implantes/efectos adversos , Infecciones Relacionadas con Prótesis/microbiología , Reoperación/efectos adversos , Sensibilidad y Especificidad , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus , Staphylococcus epidermidis , alfa-Defensinas/metabolismo
15.
Nanotechnology ; 28(5): 055102, 2017 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-28032617

RESUMEN

Herein we provided the first proof of principle for in vivo fluorescence optical imaging application using monoolein-based cubosomes in a healthy mouse animal model. This formulation, administered at a non-cytotoxic concentration, was capable of providing both exogenous contrast for NIR fluorescence imaging with very high efficiency and chemospecific information upon lifetime analysis. Time-resolved measurements of fluorescence after the intravenous injection of cubosomes revealed that the dye rapidly accumulated mainly in the liver, while lifetimes profiles obtained in vivo allowed for discriminating between free dye or dye embedded within the cubosome nanostructure after injection.


Asunto(s)
Carbocianinas/química , Colorantes Fluorescentes/química , Liposomas/farmacocinética , Nanopartículas/química , Imagen Óptica/métodos , Espectroscopía Infrarroja Corta/métodos , Animales , Carbocianinas/farmacocinética , Carbocianinas/farmacología , Supervivencia Celular/efectos de los fármacos , Composición de Medicamentos/métodos , Eritrocitos/efectos de los fármacos , Femenino , Colorantes Fluorescentes/farmacocinética , Colorantes Fluorescentes/farmacología , Glicéridos/química , Humanos , Inyecciones Intravenosas , Liposomas/síntesis química , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/ultraestructura , Ratones , Ratones Endogámicos BALB C , Células 3T3 NIH , Nanopartículas/administración & dosificación , Tamaño de la Partícula , Imagen de Lapso de Tiempo
16.
J Immunol ; 195(12): 5602-7, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26561549

RESUMEN

The abortion-prone mating combination CBA/J × DBA/2 has been recognized as a model of preeclampsia, and complement activation has been implicated in the high rate of pregnancy loss observed in CBA/J mice. We have analyzed the implantation sites collected from DBA/2-mated CBA/J mice for the deposition of the complement recognition molecules using CBA/J mated with BALB/c mice as a control group. MBL-A was observed in the implantation sites of CBA/J × DBA/2 combination in the absence of MBL-C and was undetectable in BALB/c-mated CBA/J mice. Conversely, C1q was present in both mating combinations. Searching for other complement components localized at the implantation sites of CBA/J × DBA/2, we found C4 and C3, but we failed to reveal C1r. These data suggest that complement is activated through the lectin pathway and proceeds to completion of the activation sequence as revealed by C9 deposition. MBL-A was detected as early as 3.5 d of pregnancy, and MBL-A deficiency prevented pregnancy loss in the abortion-prone mating combination. The contribution of the terminal complex to miscarriage was supported by the finding that pregnancy failure was largely inhibited by the administration of neutralizing Ab to C5. Treatment of DBA/2-mated CBA/J mice with Polyman2 that binds to MBL-A with high affinity proved to be highly effective in controlling the activation of the lectin pathway and in preventing fetal loss.


Asunto(s)
Lectina de Unión a Manosa de la Vía del Complemento , Preeclampsia/tratamiento farmacológico , Animales , Anticuerpos Bloqueadores/administración & dosificación , Complemento C5/inmunología , Complemento C5/metabolismo , Lectina de Unión a Manosa de la Vía del Complemento/efectos de los fármacos , Modelos Animales de Enfermedad , Implantación del Embrión/efectos de los fármacos , Femenino , Humanos , Masculino , Lectina de Unión a Manosa/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos CBA , Ratones Endogámicos DBA , Preeclampsia/inmunología , Embarazo
17.
Blood ; 123(22): 3478-87, 2014 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-24642748

RESUMEN

A single-chain fragment variable (scFv) recognizing ß2-glycoprotein 1 (ß2GPI) from humans and other species was isolated from a human phage display library and engineered to contain an IgG1 hinge-CH2-CH3 domain. The scFv-Fc directed against ß2GPI domain I-induced thrombosis and fetal loss, thus mimicking the effect of antibodies from patients with antiphospholipid syndrome (APS). Complement is involved in the biological effect of anti-ß2GPI scFv-Fc, as demonstrated by its ability to promote in vitro and in vivo complement deposition and the failure to induce vascular thrombosis in C6-deficient rats and fetal loss in C5-depleted mice. A critical role for complement was also supported by the inability of the CH2-deleted scFv-Fc to cause vessel occlusion and pregnancy failure. This antibody prevented the pathological effects of anti-ß2GPI antibodies from APS patients and displaced ß2GPI-bound patient antibodies. The CH2-deleted antibody represents an innovative approach potentially useful to treat APS patients refractory to standard therapy.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Síndrome Antifosfolípido/tratamiento farmacológico , Síndrome Antifosfolípido/inmunología , Autoantígenos/inmunología , Proteínas del Sistema Complemento/inmunología , beta 2 Glicoproteína I/inmunología , Aborto Espontáneo/inmunología , Animales , Anticuerpos Monoclonales/genética , Activación de Complemento/efectos de los fármacos , Activación de Complemento/inmunología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inmunoglobulina G/inmunología , Masculino , Ratones , Unión Proteica/inmunología , Ratas , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/uso terapéutico , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/aislamiento & purificación , Anticuerpos de Cadena Única/uso terapéutico , Trombosis/inmunología , Trofoblastos , beta 2 Glicoproteína I/metabolismo
18.
Blood ; 124(11): 1715-26, 2014 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-25037630

RESUMEN

Atypical hemolytic-uremic syndrome (aHUS) is associated with genetic complement abnormalities/anti-complement factor H antibodies, which paved the way to treatment with eculizumab. We studied 44 aHUS patients and their relatives to (1) test new assays of complement activation, (2) verify whether such abnormality occurs also in unaffected mutation carriers, and (3) search for a tool for eculizumab titration. An abnormal circulating complement profile (low C3, high C5a, or SC5b-9) was found in 47% to 64% of patients, irrespective of disease phase. Acute aHUS serum, but not serum from remission, caused wider C3 and C5b-9 deposits than control serum on unstimulated human microvascular endothelial cells (HMEC-1). In adenosine 5'-diphosphate-activated HMEC-1, also sera from 84% and 100% of patients in remission, and from all unaffected mutation carriers, induced excessive C3 and C5b-9 deposits. At variance, in most patients with C3 glomerulopathies/immune complex-associated membranoproliferative glomerulonephritis, serum-induced endothelial C5b-9 deposits were normal. In 8 eculizumab-treated aHUS patients, C3/SC5b-9 circulating levels did not change posteculizumab, whereas serum-induced endothelial C5b-9 deposits normalized after treatment, paralleled or even preceded remission, and guided drug dosing and timing. These results point to efficient complement inhibition on endothelium for aHUS treatment. C5b-9 endothelial deposits might help monitor eculizumab effectiveness, avoid drug overexposure, and save money considering the extremely high cost of the drug.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Activación de Complemento/efectos de los fármacos , Síndrome Hemolítico-Urémico/sangre , Síndrome Hemolítico-Urémico/tratamiento farmacológico , Monitoreo Fisiológico , Adenosina Difosfato Ribosa/farmacología , Síndrome Hemolítico Urémico Atípico , Complemento C3/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Glomerulonefritis Membranoproliferativa/sangre , Glomerulonefritis Membranoproliferativa/tratamiento farmacológico , Glomerulonefritis Membranoproliferativa/patología , Síndrome Hemolítico-Urémico/patología , Humanos , Masculino , Inducción de Remisión , Factores de Tiempo
20.
Arthritis Rheum ; 64(8): 2559-67, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22334275

RESUMEN

OBJECTIVE: To show that a new recombinant protein (MT07) obtained by fusing a synovial-homing peptide to a neutralizing antibody to C5 can be selectively delivered to inflamed synovium and can effectively control joint inflammation in experimental models of arthritis. METHODS: Binding of MT07 to human, rat, and mouse synovial tissue was evaluated in vitro by immunofluorescence, and selective localization in the inflamed joints of rats was documented in vivo using time-domain optical imaging. The antiinflammatory effect of MT07 was tested in a rat model of antigen-induced arthritis (AIA) and in a mouse model of collagen antibody-induced arthritis (CAIA). RESULTS: MT07 was able to bind to samples of inflamed synovium from humans, mice, and rats while failing to recognize uninflamed synovium as well as inflamed mouse lung or rat kidney. In vivo analysis of the biodistribution of MT07 confirmed its preferential homing to inflamed joints, with negligible inhibition of circulating C5 levels. MT07 prevented and resolved established inflammation in a rat model of AIA, as demonstrated by changes in joint swelling, polymorphonuclear cell counts in synovial washes, release of interleukin-6 and tumor necrosis factor α, and tissue damage. A similar therapeutic effect was obtained testing MT07 in a CAIA model. CONCLUSION: Our findings show that the novel recombinant molecule MT07 has the unique ability to selectively target inflamed joints and to exert local control of the inflammatory process by neutralizing the complement system without interfering with circulating C5 levels. We believe that this approach can be extended to other antiinflammatory drugs currently used to treat patients with rheumatoid arthritis.


Asunto(s)
Anticuerpos Neutralizantes/uso terapéutico , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/metabolismo , Complemento C5/inmunología , Membrana Sinovial/metabolismo , Animales , Artritis Experimental/inducido químicamente , Colágeno/efectos adversos , Modelos Animales de Enfermedad , Endotelio/metabolismo , Adyuvante de Freund/efectos adversos , Humanos , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratas , Ratas Wistar , Proteínas Recombinantes/uso terapéutico , Albúmina Sérica Bovina/efectos adversos , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA