Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Innov Surg Sci ; 5(1-2): 43-51, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33506093

RESUMEN

OBJECTIVES: Cutaneous ischemia/reperfusion (CI/R) injury has shown to play a significant role in chronic wounds such as decubitus ulcers, diabetic foot ulcers, atherosclerotic lesions, and venous stasis wounds. CI/R also plays a role in free tissue transfer in reconstructive microsurgery and has been linked to clinical burn-depth progression after thermal injury. While the role of the complement system has been elucidated in multiple organ systems, evidence is lacking with respect to its role in the skin. Therefore, we evaluated the role of the complement system in CI/R injury. METHODS: Using a single pedicle skin flap mouse model of acute CI/R, we performed CI/R in wild-type (WT) mice and complement knock out (KO) mice, deficient in either C1q (C1q KO; classical pathway inhibition), mannose-binding lectin (MBL null; lectin pathway inhibition) or factor B (H2Bf KO; alternative pathway inhibition). Following 10 h ischemia and 7 days reperfusion, mice were sacrificed, flaps harvested and flap viability assessed via Image J software. The flap necrotic area was expressed as % total flap area. In another group, mice were sacrificed following CI/R with 10 h ischemia and 48 h reperfusion. Two cranial skin flap samples were taken for gene expression analysis of IL1b, IL6, TNFα, ICAM1, VCAM1, IL10, IL13 using real-time polymerase chain reaction (RT-PCR). RESULTS: Following CI/R, MBL null mice had a statistically significant smaller %necrotic flap area compared to WT mice (10.6 vs. 43.1%; p<0.05) suggesting protection from CI/R. A significantly reduced mean %necrotic flap area was not seen in either C1q KO or H2Bf KO mice relative to WT (22.9 and 31.3 vs. 43.1%; p=0.08 and p=0.244, respectively). There were no statistically significant differences between groups for markers of inflammation (TNFα, ICAM1, VCAM1, IL1b, IL6). In contrast, mRNA levels of IL10, a regulator of inflammation, were significantly increased in the MBL null group (p=0.047). CONCLUSIONS: We demonstrated for the first time a significant role of MBL and the lectin complement pathway in ischemia/reperfusion injury of the skin and a potential role for IL10 in attenuating CI/R injury, as IL10 levels were significantly increased in the tissue from the CI/R-protected MBL null group.

3.
Front Immunol ; 7: 473, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27857715

RESUMEN

Aspergillus fumigatus infections are associated with a high mortality rate for immunocompromised patients. The complement system is considered to be important in protection against this fungus, yet the course of activation is unclear. The aim of this study was to unravel the role of the classical, lectin, and alternative pathways under both immunocompetent and immunocompromised conditions to provide a relevant dual-perspective on the response against A. fumigatus. Conidia (spores) from a clinical isolate of A. fumigatus were combined with various human serum types (including serum deficient of various complement components and serum from umbilical cord blood). We also combined this with inhibitors against C1q, mannose-binding lectin (MBL), and ficolin-2 before complement activation products and phagocytosis were detected by flow cytometry. Our results showed that alternative pathway amplified complement on A. fumigatus, but required classical and/or lectin pathway for initiation. In normal human serum, this initiation came primarily from the classical pathway. However, with a dysfunctional classical pathway (C1q-deficient serum), lectin pathway activated complement and mediated opsonophagocytosis through MBL. To model the antibody-decline in a compromised immune system, we used serum from normal umbilical cords and found MBL to be the key complement initiator. In another set of experiments, serum from patients with different kinds of immunoglobulin insufficiencies showed that the MBL lectin pathway contribution was highest in the samples with the lowest IgG/IgM binding. In conclusion, lectin pathway appears to be the primary route of complement activation in the absence of anti-A. fumigatus antibodies, whereas in a balanced immune state classical pathway is the main activator. This suggests a crucial role for the lectin pathway in innate immune protection against A. fumigatus in immunocompromised patients.

4.
Kidney Int ; 90(4): 774-82, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27378476

RESUMEN

Hemolytic uremic syndrome caused by Shiga toxin-producing Escherichia coli (STEC HUS) is a worldwide endemic problem, and its pathophysiology is not fully elucidated. Here we tested whether the mannose-binding lectin (MBL2), an initiating factor of lectin complement pathway activation, plays a crucial role in STEC HUS. Using novel human MBL2-expressing mice (MBL2 KI) that lack murine Mbls (MBL2(+/+)Mbl1(-/-)Mbl2(-/-)), a novel STEC HUS model consisted of an intraperitoneal injection with Shiga toxin-2 (Stx-2) with or without anti-MBL2 antibody (3F8, intraperitoneal). Stx-2 induced weight loss, anemia, and thrombocytopenia and increased serum creatinine, free serum hemoglobin, and cystatin C levels, but a significantly decreased glomerular filtration rate compared with control/sham mice. Immunohistochemical staining revealed renal C3d deposition and fibrin deposition in glomeruli in Stx-2-injected mice. Treatment with 3F8 completely inhibited serum MBL2 levels and significantly attenuated Stx-2 induced-renal injury, free serum hemoglobin levels, renal C3d, and fibrin deposition and preserved the glomerular filtration rate. Thus, MBL2 inhibition significantly protected against complement activation and renal injury induced by Stx-2. This novel mouse model can be used to study the role of complement, particularly lectin pathway-mediated complement activation, in Stx-2-induced renal injury.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Complemento C3d/metabolismo , Infecciones por Escherichia coli/tratamiento farmacológico , Síndrome Hemolítico-Urémico/tratamiento farmacológico , Inmunoglobulina G/uso terapéutico , Lectina de Unión a Manosa/inmunología , Toxina Shiga II/toxicidad , Animales , Anticuerpos Monoclonales de Origen Murino , Activación de Complemento/efectos de los fármacos , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/sangre , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Técnicas de Sustitución del Gen , Tasa de Filtración Glomerular , Síndrome Hemolítico-Urémico/sangre , Síndrome Hemolítico-Urémico/inmunología , Síndrome Hemolítico-Urémico/microbiología , Humanos , Inmunohistoquímica , Riñón/inmunología , Masculino , Lectina de Unión a Manosa/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Toxina Shiga II/inmunología , Escherichia coli Shiga-Toxigénica/metabolismo
5.
J Immunol ; 196(12): 5064-74, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27183610

RESUMEN

Cholesterol crystals (CC) play an essential role in the formation of atherosclerotic plaques. CC activate the classical and the alternative complement pathways, but the role of the lectin pathway is unknown. We hypothesized that the pattern recognition molecules (PRMs) from the lectin pathway bind CC and function as an upstream innate inflammatory signal in the pathophysiology of atherosclerosis. We investigated the binding of the PRMs mannose-binding lectin (MBL), ficolin-1, ficolin-2, and ficolin-3, the associated serine proteases, and complement activation products to CC in vitro using recombinant proteins, specific inhibitors, as well as deficient and normal sera. Additionally, we examined the deposition of ficolin-2 and MBL in human carotid plaques by immunohistochemistry and fluorescence microscopy. The results showed that the lectin pathway was activated on CC by binding of ficolin-2 and MBL in vitro, resulting in activation and deposition of complement activation products. MBL bound to CC in a calcium-dependent manner whereas ficolin-2 binding was calcium-independent. No binding was observed for ficolin-1 or ficolin-3. MBL and ficolin-2 were present in human carotid plaques, and binding of MBL to CC was confirmed in vivo by immunohistochemistry, showing localization of MBL around CC clefts. Moreover, we demonstrated that IgM, but not IgG, bound to CC in vitro and that C1q binding was facilitated by IgM. In conclusion, our study demonstrates that PRMs from the lectin pathway recognize CC and provides evidence for an important role for this pathway in the inflammatory response induced by CC in the pathophysiology of atherosclerosis.


Asunto(s)
Aterosclerosis/inmunología , Aterosclerosis/fisiopatología , Activación de Complemento , Progresión de la Enfermedad , Lectinas/metabolismo , Lectina de Unión a Manosa/metabolismo , Calcio/metabolismo , Estenosis Carotídea/inmunología , Colesterol/química , Colesterol/inmunología , Colesterol/metabolismo , Colesterol/farmacología , Complemento C4/metabolismo , Cristalización , Técnica del Anticuerpo Fluorescente , Glicoproteínas/inmunología , Glicoproteínas/metabolismo , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Inmunoglobulina M/inmunología , Inmunoglobulina M/metabolismo , Inflamación , Lectinas/inmunología , Lectina de Unión a Manosa/inmunología , Microscopía Fluorescente , Receptores de Reconocimiento de Patrones/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Proteínas Recombinantes/metabolismo , Ficolinas
6.
Mol Ther ; 23(6): 1066-1076, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25807289

RESUMEN

Oncolytic viruses (OVs) have shown promising clinical activity when administered by direct intratumoral injection. However, natural barriers in the blood, including antibodies and complement, are likely to limit the ability to repeatedly administer OVs by the intravenous route. We demonstrate here that for a prototype of the clinical vaccinia virus based product Pexa-Vec, the neutralizing activity of antibodies elicited by smallpox vaccination, as well as the anamnestic response in hyperimmune virus treated cancer patients, is strictly dependent on the activation of complement. In immunized rats, complement depletion stabilized vaccinia virus in the blood and led to improved delivery to tumors. Complement depletion also enhanced tumor infection when virus was directly injected into tumors in immunized animals. The feasibility and safety of using a complement inhibitor, CP40, in combination with vaccinia virus was tested in cynomolgus macaques. CP40 pretreatment elicited an average 10-fold increase in infectious titer in the blood early after the infusion and prolonged the time during which infectious virus was detectable in the blood of animals with preexisting immunity. Capitalizing on the complement dependence of antivaccinia antibody with adjunct complement inhibitors may increase the infectious dose of oncolytic vaccinia virus delivered to tumors in virus in immune hosts.


Asunto(s)
Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Virus Vaccinia/inmunología , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Línea Celular Tumoral , Chlorocebus aethiops , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Estudios de Factibilidad , Femenino , Células HeLa , Humanos , Inyecciones Intralesiones , Macaca fascicularis/inmunología , Masculino , Neoplasias/sangre , Neoplasias/terapia , Pruebas de Neutralización , Piridonas/inmunología , Piridonas/farmacología , Ratas , Ratas Endogámicas F344 , Vacuna contra Viruela/sangre , Vacuna contra Viruela/inmunología , Vacunación , Células Vero
7.
Am J Pathol ; 185(2): 347-55, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25482922

RESUMEN

Myocardial infarction and coagulation disorders are leading causes of disability and death in the world. An important role of the lectin complement pathway in myocardial infarction and coagulation has been demonstrated in mice genetically deficient in lectin complement pathway proteins. However, these studies are limited to comparisons between wild-type and deficient mice and lack the ability to examine reversal/inhibition of injury after disease establishment. We developed a novel mouse that expresses functional human mannose-binding lectin (MBL) 2 under the control of Mbl1 promoter. Serum MBL2 concentrations averaged approximately 3 µg/mL in MBL2(+/+)Mbl1(-/-)Mbl2(-/-) [MBL2 knock in (KI)] mice. Serum MBL2 level in MBL2 KI mice significantly increased after 7 (8 µg/mL) or 14 (9 µg/mL) days of hyperglycemia compared to normoglycemic mice (P < 0.001). Monoclonal antibody 3F8 inhibited C3 deposition on mannan-coated plates in MBL2 KI, but not wild-type, mice. Myocardial ischemia/reperfusion in MBL2 KI mice revealed that 3F8 preserved cardiac function and decreased infarct size and fibrin deposition in a time-dependent manner. Furthermore, 3F8 prevented ferric chloride-induced occlusive arterial thrombogenesis in vivo. MBL2 KI mice represent a novel animal model that can be used to study the lectin complement pathway in acute and chronic models of human disease. Furthermore, these novel mice demonstrate the therapeutic window for MBL2 inhibition for effective treatment of disease and its complications.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/farmacología , Anticuerpos Neutralizantes/farmacología , Modelos Animales de Enfermedad , Lectina de Unión a Manosa/antagonistas & inhibidores , Infarto del Miocardio/tratamiento farmacológico , Trombosis/tratamiento farmacológico , Animales , Técnicas de Sustitución del Gen , Humanos , Lectina de Unión a Manosa/sangre , Lectina de Unión a Manosa/genética , Lectina de Unión a Manosa/metabolismo , Ratones , Ratones Noqueados , Infarto del Miocardio/sangre , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Regiones Promotoras Genéticas , Trombosis/sangre , Trombosis/genética , Trombosis/patología
8.
Thromb Haemost ; 113(3): 548-52, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25031089

RESUMEN

The complement system is an intrinsic part of the immune system and has important functions in both innate and adaptive immunity. On the other hand, inadvertent or misdirected complement activation is also involved in the pathogenesis of many diseases, contributing solely or significantly to tissue injury and disease development. Multiple approaches to develop pharmacological agents to inhibit complement are currently being pursued. We have developed a conceptually different approach of not inhibiting but depleting complement, based on the complement-depleting activities of cobra venom factor (CVF), a non-toxic cobra venom component with structural and functional homology to complement component C3. We developed a humanised version of CVF by creating human complement component C3 derivatives with complement-depleting activities of CVF (humanised CVF) as a promising therapeutic agent for diseases with complement pathogenesis. Here we review the beneficial therapeutic effect of humanised CVF in several murine models of vascular diseases such as reperfusion injury.


Asunto(s)
Activación de Complemento/efectos de los fármacos , Complemento C3/farmacología , Inactivadores del Complemento/farmacología , Proteínas del Sistema Complemento/metabolismo , Venenos Elapídicos/farmacología , Factores Inmunológicos/farmacología , Daño por Reperfusión/tratamiento farmacológico , Lesión Pulmonar Inducida por Ventilación Mecánica/tratamiento farmacológico , Animales , Proteínas del Sistema Complemento/inmunología , Modelos Animales de Enfermedad , Humanos , Proteínas Recombinantes de Fusión/farmacología , Daño por Reperfusión/inmunología , Lesión Pulmonar Inducida por Ventilación Mecánica/inmunología
9.
PLoS One ; 9(4): e95160, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24740152

RESUMEN

There is considerable evidence that influenza A virus (IAV) promotes adherence, colonization, and superinfection by S. pneumoniae (Spn) and contributes to the pathogenesis of otitis media (OM). The complement system is a critical innate immune defense against both pathogens. To assess the role of the complement system in the host defense and the pathogenesis of acute pneumococcal OM following IAV infection, we employed a well-established transtympanically-induced mouse model of acute pneumococcal OM. We found that antecedent IAV infection enhanced the severity of acute pneumococcal OM. Mice deficient in complement C1qa (C1qa-/-) or factor B (Bf -/-) exhibited delayed viral and bacterial clearance from the middle ear and developed significant mucosal damage in the eustachian tube and middle ear. This indicates that both the classical and alternative complement pathways are critical for the oto-immune defense against acute pneumococcal OM following influenza infection. We also found that Spn increased complement activation following IAV infection. This was characterized by sustained increased levels of anaphylatoxins C3a and C5a in serum and middle ear lavage samples. In contrast, mice deficient in the complement C5a receptor (C5aR) demonstrated enhanced bacterial clearance and reduced severity of OM. Our data support the concept that C5a-C5aR interactions play a significant role in the pathogenesis of acute pneumococcal OM following IAV infection. It is possible that targeting the C5a-C5aR axis might prove useful in attenuating acute pneumococcal OM in patients with influenza infection.


Asunto(s)
Trompa Auditiva/inmunología , Infecciones por Orthomyxoviridae/inmunología , Otitis Media/inmunología , Infecciones Neumocócicas/inmunología , Receptor de Anafilatoxina C5a/genética , Enfermedad Aguda , Animales , Coinfección , Activación de Complemento , Complemento C1q/deficiencia , Complemento C1q/genética , Complemento C3a/genética , Complemento C3a/inmunología , Complemento C5a/genética , Complemento C5a/inmunología , Factor B del Complemento/deficiencia , Factor B del Complemento/genética , Trompa Auditiva/microbiología , Trompa Auditiva/patología , Trompa Auditiva/virología , Femenino , Eliminación de Gen , Expresión Génica , Inmunidad Innata , Subtipo H1N1 del Virus de la Influenza A/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Otitis Media/genética , Otitis Media/microbiología , Otitis Media/patología , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/patología , Receptor de Anafilatoxina C5a/deficiencia , Índice de Severidad de la Enfermedad , Streptococcus pneumoniae/inmunología , Streptococcus pneumoniae/patogenicidad
10.
FASEB J ; 28(7): 3171-82, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24668752

RESUMEN

A defining feature in proliferative retinopathies is the formation of pathological neovessels. In these diseases, the balance between neovessel formation and regression determines blindness, making the modulation of neovessel growth highly desirable. The role of the immune system in these retinopathies is of increasing interest, but it is not completely understood. We investigated the role of the alternative complement pathway during the formation and resolution of aberrant neovascularization. We used alternative complement pathway-deficient (Fb(-/-)) mice and age- and strain-matched control mice to assess neovessel development and regression in an oxygen-induced retinopathy (OIR) mouse model. In the control mice, we found increased transcription of Fb after OIR treatment. In the Fb(-/-) mice, we prepared retinal flatmounts and identified an increased number of neovessels, peaking at postnatal day 17 (P17; P=0.001). Subjecting human umbilical vein endothelial cells (HUVECs) to low oxygen, mimicking a characteristic of neovessels, decreased the expression of the complement inhibitor Cd55. Finally, using laser capture microdissection (LCM) to isolate the neovessels after OIR, we found decreased expression of Cd55 (P=0.005). Together, our data implicate the alternative complement pathway in facilitating neovessel clearance by down-regulating the complement inhibitor Cd55 specifically on neovessels, allowing for their targeted removal while leaving the established vasculature intact.-Sweigard, J. H., Yanai, R., Gaissert, P., Saint-Geniez, M., Kataoka, K., Thanos, A., Stahl, G. L., Lambris, J. D., Connor, K. M. The alternative complement pathway regulates pathological angiogenesis in the retina.


Asunto(s)
Vía Alternativa del Complemento/fisiología , Neovascularización Patológica/patología , Neovascularización Retiniana/patología , Animales , Apoptosis/fisiología , Antígenos CD55/metabolismo , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo/fisiología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/metabolismo , Oxígeno/metabolismo , Retina , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Neovascularización Retiniana/metabolismo
11.
J Immunol ; 190(12): 6423-33, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23650618

RESUMEN

The complement system is involved in mediation of joint damage in rheumatoid arthritis, with evidence suggesting activation of both the classical and alternative pathway (AP). The AP is both necessary and sufficient to mediate collagen Ab-induced arthritis, an experimental animal model of immune complex-induced joint disease. The AP in mice is dependent on MASP-1/3 cleavage of pro-factor D (pro-FD) into mature factor D (FD). The objectives of the current study were to determine the cells synthesizing MASP-1/3 and pro-FD in synovial tissue. Collagen Ab-induced arthritis was studied in wild-type C57BL/6 mice, and the localization of mRNA and protein for FD and MASP-1/3 in synovial adipose tissue (SAT) and fibroblast-like synoviocytes (FLS) was determined using various techniques, including laser capture microdissection. SAT was the sole source of mRNA for pro-FD. Cultured differentiated 3T3 adipocytes, a surrogate for SAT, produced pro-FD but no mature FD. FLS were the main source of MASP-1/3 mRNA and protein. Using cartilage microparticles (CMPs) coated with anti-collagen mAb and serum from MASP-1/3(-/-) mice as a source of factor B, pro-FD in 3T3 supernatants was cleaved into mature FD by MASP-1/3 in FLS supernatants. The mature FD was eluted from the CMP, and was not present in the supernatants from the incubation with CMP, indicating that cleavage of pro-FD into mature FD by MASP-1 occurred on the CMP. These results demonstrate that pathogenic activation of the AP can occur in the joint through immune complexes adherent to cartilage and the local production of necessary AP proteins by adipocytes and FLS.


Asunto(s)
Adipocitos/metabolismo , Artritis Experimental/metabolismo , Activación de Complemento/fisiología , Vía Alternativa del Complemento/fisiología , Fibroblastos/metabolismo , Adipocitos/inmunología , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Western Blotting , Fibroblastos/inmunología , Inmunohistoquímica , Captura por Microdisección con Láser , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Membrana Sinovial/inmunología , Membrana Sinovial/metabolismo , Membrana Sinovial/patología
12.
PLoS One ; 8(4): e60838, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23573288

RESUMEN

Mannose-binding lectin (MBL) is a key soluble effector of the innate immune system that recognizes pathogen-specific surface glycans. Surprisingly, low-producing MBL genetic variants that may predispose children and immunocompromised individuals to infectious diseases are more common than would be expected in human populations. Since certain immune defense molecules, such as immunoglobulins, can be exploited by invasive pathogens, we hypothesized that MBL might also enhance infections in some circumstances. Consequently, the low and intermediate MBL levels commonly found in human populations might be the result of balancing selection. Using model infection systems with pseudotyped and authentic glycosylated viruses, we demonstrated that MBL indeed enhances infection of Ebola, Hendra, Nipah and West Nile viruses in low complement conditions. Mechanistic studies with Ebola virus (EBOV) glycoprotein pseudotyped lentiviruses confirmed that MBL binds to N-linked glycan epitopes on viral surfaces in a specific manner via the MBL carbohydrate recognition domain, which is necessary for enhanced infection. MBL mediates lipid-raft-dependent macropinocytosis of EBOV via a pathway that appears to require less actin or early endosomal processing compared with the filovirus canonical endocytic pathway. Using a validated RNA interference screen, we identified C1QBP (gC1qR) as a candidate surface receptor that mediates MBL-dependent enhancement of EBOV infection. We also identified dectin-2 (CLEC6A) as a potentially novel candidate attachment factor for EBOV. Our findings support the concept of an innate immune haplotype that represents critical interactions between MBL and complement component C4 genes and that may modify susceptibility or resistance to certain glycosylated pathogens. Therefore, higher levels of native or exogenous MBL could be deleterious in the setting of relative hypocomplementemia which can occur genetically or because of immunodepletion during active infections. Our findings confirm our hypothesis that the pressure of infectious diseases may have contributed in part to evolutionary selection of MBL mutant haplotypes.


Asunto(s)
Ebolavirus/fisiología , Infecciones por Filoviridae/metabolismo , Lectina de Unión a Manosa/metabolismo , Receptores Mitogénicos/metabolismo , Internalización del Virus , Animales , Chlorocebus aethiops , Proteínas del Sistema Complemento/metabolismo , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Glicoproteínas de Membrana/metabolismo , Pinocitosis , Células Vero , Proteínas del Envoltorio Viral/metabolismo
13.
Circulation ; 126(18): 2227-35, 2012 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-23032324

RESUMEN

BACKGROUND: Coagulation disorders and reperfusion of ischemic myocardium are major causes of morbidity and mortality. Lectin pathway initiation complexes are composed of multimolecular carbohydrate recognition subcomponents and 3 lectin pathway-specific serine proteases. We have recently shown that the lectin pathway-specific carbohydrate recognition subcomponent mannose-binding lectin plays an essential role in the pathophysiology of thrombosis and ischemia/reperfusion injury. Thus, we hypothesized that the endogenous mannose-binding lectin (MBL)/ficolin-associated protein-1 (MAP-1) that inhibits complement activation in vitro also could be an in vivo regulator by attenuating myocardial schema/reperfusion injury and thrombogenesis when used at pharmacological doses in wild-type mice. METHODS AND RESULTS: In 2 mouse models, MAP-1 preserves cardiac function, decreases infarct size, decreases C3 deposition, inhibits MBL deposition, and prevents thrombogenesis. Furthermore, we demonstrate that MAP-1 displaces MBL/ficolin-associated serine protease (MASP)-1, MASP-2, and MASP-3 from the MBL complex. CONCLUSIONS: Our results suggest that the natural, endogenous inhibitor MAP-1 effectively inhibits lectin pathway activation in vivo. MAP-1 at pharmacological doses represents a novel therapeutic approach for human diseases involving the lectin pathway and its associated MASPs.


Asunto(s)
Anticoagulantes/uso terapéutico , Trombosis de las Arterias Carótidas/tratamiento farmacológico , Lectina de Unión a Manosa de la Vía del Complemento/efectos de los fármacos , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/antagonistas & inhibidores , Infarto del Miocardio/tratamiento farmacológico , Daño por Reperfusión Miocárdica/prevención & control , Animales , Anticoagulantes/farmacología , Trombosis de las Arterias Carótidas/inducido químicamente , Complemento C3/análisis , Lectina de Unión a Manosa de la Vía del Complemento/fisiología , Depresión Química , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Lectinas/metabolismo , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/deficiencia , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/genética , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/farmacología , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/fisiología , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Cardiovasculares , Modelos Inmunológicos , Peso Molecular , Complejos Multiproteicos/efectos de los fármacos , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/diagnóstico por imagen , Daño por Reperfusión Miocárdica/patología , Unión Proteica , Proteínas Recombinantes de Fusión/metabolismo , Ultrasonografía , Ficolinas
14.
J Immunol ; 189(9): 4640-7, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23028050

RESUMEN

There is mounting evidence indicating an important role for complement in the pathogenesis of cerebral ischemia-reperfusion injury, or ischemic stroke. The role of the alternative complement pathway in ischemic stroke has not been investigated, and there is conflicting data on the role of the terminal pathway. In this study, we show that compared with wild-type mice, mice deficient in the alternative pathway protein factor B or mice treated with the alternative pathway inhibitor CR2-fH have improved outcomes after 60-min middle cerebral artery occlusion and 24-h reperfusion. Factor B-deficient or CR2-fH-treated mice were protected in terms of improved neurologic function and reduced cerebral infarct, demyelination, P-selectin expression, neutrophil infiltration, and microthrombi formation. Mice deficient in both the classical and lectin pathways (C1q/MBL deficient) were also protected from cerebral ischemia-reperfusion injury, and there was no detectable C3d deposition in the ipsilateral brain of these mice. These data demonstrate that the alternative pathway is not alone sufficient to initiate complement activation and indicate that the alternative pathway propagates cerebral injury via amplification of the cascade. Deficiency of C6, a component of the terminal cytolytic membrane attack complex, had no effect on outcome after ischemic stroke, indicating that the membrane attack complex is not involved in mediating injury in this model. We additionally show that the protective effect of factor B deficiency and CR2-fH treatment is sustained in the subacute stage of infarct development, adding to the clinical relevance of these findings.


Asunto(s)
Isquemia Encefálica/inmunología , Isquemia Encefálica/patología , Vía Alternativa del Complemento/inmunología , Mediadores de Inflamación/fisiología , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/patología , Animales , Isquemia Encefálica/genética , Antígenos CD59/genética , Complemento C1q/deficiencia , Complemento C1q/genética , Complemento C6/deficiencia , Complemento C6/genética , Factor B del Complemento/deficiencia , Factor B del Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento/deficiencia , Complejo de Ataque a Membrana del Sistema Complemento/genética , Vía Alternativa del Complemento/genética , Lectina de Unión a Manosa de la Vía del Complemento/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión/genética , Accidente Cerebrovascular/genética
15.
Immunobiology ; 217(11): 1026-33, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22964228

RESUMEN

Tissue injury and inflammation following ischemia and reperfusion of various organs have been recognized for many years. Many reviews have been written over the last several decades outlining the role of complement in ischemia/reperfusion injury. This short review provides a current state of the art knowledge on the complement pathways activated, complement components involved and a review of the clinical biologics/inhibitors used in the clinical setting of ischemia/reperfusion. This is not a complete review of the complement system in ischemia and reperfusion injury but will give the reader an updated view point of the field, potential clinical use of complement inhibitors, and the future studies needed to advance the field.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento/inmunología , Daño por Reperfusión/inmunología , Proteínas del Sistema Complemento/metabolismo , Humanos , Inflamación/inmunología
16.
Microbes Infect ; 14(14): 1308-18, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22975410

RESUMEN

There is considerable evidence that phase variation among transparent and opaque colony phenotypes of Streptococcus pneumoniae (Spn) plays an important role in the pneumococcal adherence and invasion. The current study was designed to investigate the interactions of the opacity phenotype variants of Spn with specific complement pathway activation in a mouse model of acute otitis media (AOM). Although the opaque colony phenotype was expected to be more resistant to complement mediated killing compared to the transparent Spn variant, we discovered that C3b deposition on the transparent Spn is, in large part, dependent on the alternative pathway activation. There were no significant differences in resistance to complement mediated opsonophagocytosis between the two variants in factor B deficient mice. In addition, an in vitro study demonstrated that significantly more C4b-binding protein (C4BP) (the classical pathway inhibitor) and factor H (FH) (the alternative pathway inhibitor) bound to the transparent strain compared with the opaque one. Our data suggest that the difference in the relative virulence of Spn opacity phenotypes is associated with its ability to evade complement-mediated opsonophagocytosis in a mouse model of pneumococcal AOM.


Asunto(s)
Vía Alternativa del Complemento/inmunología , Vía Clásica del Complemento/inmunología , Interacciones Huésped-Patógeno/inmunología , Otitis Media/inmunología , Otitis Media/microbiología , Infecciones Neumocócicas/inmunología , Streptococcus pneumoniae/inmunología , Enfermedad Aguda , Animales , Proteína de Unión al Complemento C4b , Factor H de Complemento , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Modelos Animales de Enfermedad , Oído Medio/química , Oído Medio/microbiología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Otitis/inmunología , Fenotipo
17.
J Immunol ; 189(5): 2606-13, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22851705

RESUMEN

Properdin is well known as an enhancer of the alternative complement amplification loop when C3 is activated, whereas its role as a recognition molecule of exogenous pathogen-associated molecular patterns and initiator of complement activation is less understood. We therefore studied the role of properdin in activation of complement in normal human serum by zymosan and various Escherichia coli strains. In ELISA, microtiter plates coated with zymosan induced efficient complement activation with deposition of C4b and terminal complement complex on the solid phase. Virtually no deposition of C4b or terminal complement complex was observed with mannose-binding lectin (MBL)-deficient serum. Reconstitution with purified MBL showed distinct activation in both readouts. In ELISA, normal human serum-induced deposition of properdin by zymosan was abolished by the C3-inhibiting peptide compstatin. Flow cytometry was used to further explore whether properdin acts as an initial recognition molecule reacting directly with zymosan and three E. coli strains. Experiments reported by other authors were made with EGTA Mg²âº buffer, permitting autoactivation of C3. We found inhibition by compstatin on these substrates, indicating that properdin deposition depended on initial C3b deposition followed by properdin in a second step. Properdin released from human polymorphonuclear cells stimulated with PMA did not bind to zymosan or E. coli, but when incubated in properdin-depleted serum this form of properdin bound efficiently to both substrates in a strictly C3-dependent manner, as the binding was abolished by compstatin. Collectively, these data indicate that properdin in serum as well as polymorphonuclear-released properdin is unable to bind and initiate direct alternative pathway activation on these substrates.


Asunto(s)
Vía Alternativa del Complemento/inmunología , Proteínas de Escherichia coli/fisiología , Escherichia coli/inmunología , Properdina/fisiología , Zimosan/fisiología , Adulto , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Humanos , Masculino , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Properdina/metabolismo , Unión Proteica/inmunología , Especificidad por Sustrato/inmunología
18.
Circulation ; 126(12): 1484-94, 2012 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-22879370

RESUMEN

BACKGROUND: The involvement of the complement system in brain injury has been scarcely investigated. Here, we document the pivotal role of mannose-binding lectin (MBL), one of the recognition molecules of the lectin complement pathway, in brain ischemic injury. METHODS AND RESULTS: Focal cerebral ischemia was induced in mice (by permanent or transient middle cerebral artery occlusion) and rats (by 3-vessel occlusion). We first observed that MBL is deposited on ischemic vessels up to 48 hours after injury and that functional MBL/MBL-associated serine protease 2 complexes are increased. Next, we demonstrated that (1) MBL(-/-) mice are protected from both transient and permanent ischemic injury; (2) Polyman2, the newly synthesized mannosylated molecule selected for its binding to MBL, improves neurological deficits and infarct volume when given up to 24 hours after ischemia in mice; (3) anti-MBL-A antibody improves neurological deficits and infarct volume when given up to 18 hours after ischemia, as assessed after 28 days in rats. CONCLUSIONS: Our data show an important role for MBL in the pathogenesis of brain ischemic injury and provide a strong support to the concept that MBL inhibition may be a relevant therapeutic target in humans, one with a wide therapeutic window of application.


Asunto(s)
Isquemia Encefálica/fisiopatología , Infarto de la Arteria Cerebral Media/fisiopatología , Lectina de Unión a Manosa/genética , Lectina de Unión a Manosa/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/genética , Edema Encefálico/fisiopatología , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/genética , Modelos Animales de Enfermedad , Humanos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/genética , Masculino , Mananos/metabolismo , Mananos/farmacología , Lectina de Unión a Manosa/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratas , Ratas Endogámicas
19.
PLoS Pathog ; 8(7): e1002793, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22792067

RESUMEN

The complement system plays a key role in host defense against pneumococcal infection. Three different pathways, the classical, alternative and lectin pathways, mediate complement activation. While there is limited information available on the roles of the classical and the alternative activation pathways of complement in fighting streptococcal infection, little is known about the role of the lectin pathway, mainly due to the lack of appropriate experimental models of lectin pathway deficiency. We have recently established a mouse strain deficient of the lectin pathway effector enzyme mannan-binding lectin associated serine protease-2 (MASP-2) and shown that this mouse strain is unable to form the lectin pathway specific C3 and C5 convertases. Here we report that MASP-2 deficient mice (which can still activate complement via the classical pathway and the alternative pathway) are highly susceptible to pneumococcal infection and fail to opsonize Streptococcus pneumoniae in the none-immune host. This defect in complement opsonisation severely compromises pathogen clearance in the lectin pathway deficient host. Using sera from mice and humans with defined complement deficiencies, we demonstrate that mouse ficolin A, human L-ficolin, and collectin 11 in both species, but not mannan-binding lectin (MBL), are the pattern recognition molecules that drive lectin pathway activation on the surface of S. pneumoniae. We further show that pneumococcal opsonisation via the lectin pathway can proceed in the absence of C4. This study corroborates the essential function of MASP-2 in the lectin pathway and highlights the importance of MBL-independent lectin pathway activation in the host defense against pneumococci.


Asunto(s)
Activación de Complemento , Inmunidad Innata , Lectinas/inmunología , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/metabolismo , Infecciones Neumocócicas/inmunología , Streptococcus pneumoniae/inmunología , Animales , Colectinas/metabolismo , Complemento C4/inmunología , Humanos , Lectinas/metabolismo , Lectina de Unión a Manosa/metabolismo , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/deficiencia , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Opsoninas , Receptores de Reconocimiento de Patrones/metabolismo , Ficolinas
20.
Anesth Analg ; 115(4): 759-71, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22798530

RESUMEN

Complement activation and the resulting inflammatory response is an important potential mechanism for multisystem organ injury in cardiac surgery. Novel therapeutic strategies using complement inhibitors may hold promise for improving outcomes for cardiac surgical patients by attenuating complement activation or its biologically active effector molecules. Recent clinical trials evaluating complement inhibitors have provided important data to further delineate the impact of complement activation and its inhibition on clinical outcomes. In this review we examine the role of complement activation and its inhibition as a therapeutic approach in cardiac surgery.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos/efectos adversos , Fármacos Cardiovasculares/administración & dosificación , Activación de Complemento/efectos de los fármacos , Activación de Complemento/inmunología , Sistemas de Liberación de Medicamentos , Animales , Sistemas de Liberación de Medicamentos/métodos , Humanos , Mediadores de Inflamación/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...