Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Infect Dis ; 229(3): 680-690, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-37878754

RESUMEN

Most patients with COVID-19 in the intensive care unit develop an acute respiratory distress syndrome characterized by severe hypoxemia, decreased lung compliance, and high vascular permeability. Activation of the complement system is a hallmark of moderate and severe COVID-19, with abundant deposition of complement proteins in inflamed tissue and on the endothelium during COVID-19. Using a transgenic mouse model of SARS-CoV-2 infection, we assessed the therapeutic utility of an inhibitory antibody (HG4) targeting MASP-2, a key enzyme in the lectin pathway. Treatment of infected mice with HG4 reduced the disease severity score and improved survival vs mice that received an isotype control antibody. Administration of HG4 significantly reduced the lung injury score, including alveolar inflammatory cell infiltration, alveolar edema, and alveolar hemorrhage. The ameliorating effect of MASP-2 inhibition on the severity of COVID-19 pathology is reflected by a significant reduction in the proinflammatory activation of brain microglia in HG4-treated mice.


Asunto(s)
COVID-19 , Síndrome de Dificultad Respiratoria , Humanos , Animales , Ratones , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/metabolismo , SARS-CoV-2/metabolismo , Activación de Complemento , Modelos Animales de Enfermedad , Proteínas del Sistema Complemento
2.
BMC Microbiol ; 24(1): 167, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755524

RESUMEN

BACKGROUND: The world faces a major infectious disease challenge. Interest in the discovery, design, or development of antimicrobial peptides (AMPs) as an alternative approach for the treatment of bacterial infections has increased. Insects are a good source of AMPs which are the main effector molecules of their innate immune system. Black Soldier Fly Larvae (BSFL) are being developed for large-scale rearing for food sustainability, waste reduction and as sustainable animal and fish feed. Bioinformatic studies have suggested that BSFL have the largest number of AMPs identified in insects. However, most AMPs identified in BSF have not yet undergone antimicrobial evaluation but are promising leads to treat critical infections. RESULTS: Jg7197.t1, Jg7902.t1 and Jg7904.t1 were expressed into the haemolymph of larvae following infection with Salmonella enterica serovar Typhimurium and were predicted to be AMPs using the computational tool ampir. The genes encoding these proteins were within 2 distinct clusters in chromosome 1 of the BSF genome. Following removal of signal peptides, predicted structures of the mature proteins were superimposed, highlighting a high degree of structural conservation. The 3 AMPs share primary sequences with proteins that contain a Kunitz-binding domain; characterised for inhibitory action against proteases, and antimicrobial activities. An in vitro antimicrobial screen indicated that heterologously expressed SUMO-Jg7197.t1 and SUMO-Jg7902.t1 did not show activity against 12 bacterial strains. While recombinant SUMO-Jg7904.t1 had antimicrobial activity against a range of Gram-negative and Gram-positive bacteria, including the serious pathogen Pseudomonas aeruginosa. CONCLUSIONS: We have cloned and purified putative AMPs from BSFL and performed initial in vitro experiments to evaluate their antimicrobial activity. In doing so, we have identified a putative novel defensin-like AMP, Jg7904.t1, encoded in a paralogous gene cluster, with antimicrobial activity against P. aeruginosa.


Asunto(s)
Antibacterianos , Defensinas , Dípteros , Larva , Animales , Defensinas/farmacología , Defensinas/genética , Defensinas/química , Defensinas/aislamiento & purificación , Antibacterianos/farmacología , Antibacterianos/química , Dípteros/genética , Larva/efectos de los fármacos , Larva/genética , Pruebas de Sensibilidad Microbiana , Secuencia de Aminoácidos , Proteínas de Insectos/genética , Proteínas de Insectos/farmacología , Proteínas de Insectos/química , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/genética , Péptidos Antimicrobianos/química , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/genética , Bacterias Gramnegativas/efectos de los fármacos
3.
BMC Microbiol ; 23(1): 378, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38036998

RESUMEN

BACKGROUND: There is a global need to develop new therapies to treat infectious diseases and tackle the rise in antimicrobial resistance. To date, the larvae of the Black Solider Fly, Hermetia illucens, have the largest repertoire of antimicrobial peptides derived from insects. Antimicrobial peptides are of particular interest in the exploration of alternative antimicrobials due to their potent action and reduced propensity to induce resistance compared with more traditional antibiotics. RESULTS: The predicted attacin from H. illucens, Hill_BB_C10074, was first identified in the transcriptome of H. illucens populations that had been fed a plant-oil based diet. In this study, recombinant Hill_BB_C10074 (500 µg/mL), was found to possess potent antimicrobial activity against the serious Gram-negative pathogen, Pseudomonas aeruginosa. Sequence and structural homology modelling predicted that Hill_BB_C10074 formed a homotrimeric complex that may form pores in the Gram-negative bacterial outer membrane. In vitro experiments defined the antimicrobial action of Hill_BB_C10074 against P. aeruginosa and transmission electron microscopy and electrochemical impedance spectroscopy confirmed the outer membrane disruptive power of Hill_BB_C10074 which was greater than the clinically relevant antibiotic, polymyxin B. CONCLUSIONS: Combining predictive tools with in vitro approaches, we have characterised Hill_BB_C10074 as an important insect antimicrobial peptide and promising candidate for the future development of clinical antimicrobials.


Asunto(s)
Antiinfecciosos , Dípteros , Animales , Pseudomonas aeruginosa , Péptidos Antimicrobianos , Dípteros/microbiología , Antiinfecciosos/farmacología , Antibacterianos/farmacología , Antibacterianos/química
4.
Microb Pathog ; 164: 105408, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35063609

RESUMEN

Infection caused by K. pneumoniae is associated with severe inflammation due to stimulation of the innate immune components including the complement system, which is the main player of the innate immune response. Excessive complement-mediated inflammation may cause severe lung injury. Here we clearly show that K. pneumoniae binds to different lectin pathway carbohydrate recognition molecules and activates the complement cascade via the LP. Administration of anti-CL-11 antibodies 6 h before the infection impairs LP functional activity but it shows no effect on the survival time of mice infected with K. pneumoniae. Similarly, no significant difference in bacterial load in blood and lung tissues was observed between mice that received anti-CL-11 and control group treated with an isotype antibody. Interestingly, treatment of mice with anti-CL-11 prior to infection significantly improved histopathological changes and lung injury score induced by K. pneumoniae. Moreover, administration of anti-CL-11 reduced leukocytes infiltration into lung tissues and decreased the levels of the inflammatory mediators TNF-α, IL-6, and IL-1ß in the infected mice. These findings indicate that inhibition of the LP could secure a significant level of protection against lung injury during the infection caused by K. pneumoniae.


Asunto(s)
Infecciones por Klebsiella , Neumonía , Animales , Inflamación/patología , Infecciones por Klebsiella/tratamiento farmacológico , Klebsiella pneumoniae , Pulmón/patología , Ratones , Neumonía/tratamiento farmacológico , Neumonía/patología
5.
J Appl Biomed ; 20(4): 124-129, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36708717

RESUMEN

OBJECTIVES: This study aims to evaluate the pharmacological role of indigo extract in accelerating the wound healing in a rat model. METHODS: Female Sprague-Dawley rats were anesthetized with ketamine (30 mg/kg, i.p.) and the full thickness of the marked skin was then cut carefully and wounds were left undressed. Indigo extract (5%) in PBS was applied topically twice daily until healing was complete. A control group of rats was treated with povidone-iodide (Betadine®). Rats treated with phosphate buffer saline were used as a negative control group. The rate of wound healing was assessed daily. Histopathological examination of skin sections were qualitatively assessed by independent evaluators. The inflammatory and apoptotic markers were assessed in skin tissue homogenates using ELISA. RESULTS: Histopathology data showed that applying indigo to skin wounds enhanced the healing process, resulting in a significant decrease in dermal inflammation in comparison to untreated rats. Topical application of indigo significantly increased antioxidant enzyme activities with reduced malondialdehyde (MDA) levels in wound tissues. The levels of matrix metalloproteases-2 and -9 were significantly lower with an accompanied increase in the level of TGF-ß1 in skin tissues from rats treated with indigo compared to the control group treated with PBS. CONCLUSIONS: The antioxidant and anti-inflammatory properties of indigo leaf extract accelerate the healing of skin injuries.


Asunto(s)
Carmin de Índigo , Enfermedades de la Piel , Ratas , Animales , Ratas Sprague-Dawley , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Extractos Vegetales/farmacología , Cicatrización de Heridas , Hojas de la Planta
6.
Infect Immun ; 89(5)2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33593889

RESUMEN

Enterococcus faecalis infections are considered a major public health concern worldwide. The complement system has a crucial role in the protection against different microbial pathogens, including E. faecalis Complement can be activated through three different pathways, including the classical, lectin, and alternative pathways. There is limited information on the role of the classical pathway (CP) in protection against infections caused by E. faecalis In the present study, we generated Fab fragments that successfully block the CP in mouse via inhibition of a key enzyme, C1s-A. Our results showed that anti-C1s-A Fab fragments block CP-mediated C3b and C4b deposition in vitro We further showed that administration of anti-C1s-A Fab fragments significantly impairs the CP functional activity in vivo Moreover, treatment of mice infected with E. faecalis using anti-C1s-A Fab fragments significantly impairs bacterial clearance as determined from the viable bacterial counts recovered from blood, kidneys, spleens, livers, and lungs of infected mice. Overall, this study highlights the essential role of the CP in host defense against E. faecalis.


Asunto(s)
Activación de Complemento/inmunología , Vía Clásica del Complemento , Proteínas del Sistema Complemento/inmunología , Enterococcus faecalis/inmunología , Infecciones por Bacterias Grampositivas/inmunología , Infecciones por Bacterias Grampositivas/microbiología , Interacciones Huésped-Patógeno , Animales , Carga Bacteriana , Susceptibilidad a Enfermedades , Humanos , Ratones , Especificidad de Órganos
7.
Microb Pathog ; 138: 103761, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31560974

RESUMEN

Pneumococcal surface protein A (PspA) is one of the major virulence factors expressed by almost all pneumococcal serotypes and was suggested to be a promising universal vaccine candidate for all pneumococcal sero-groups. Here, we expressed and purified the proline-rich region (PR) of PspA and tested it as a recombinant vaccine against infection caused by a clinical isolate (SP19) of Streptococcus pneumoniae serotype 19F. Our results showed that BALB/c mice immunized with recombinant proline-rich (rPR) region showed a significant higher antibody titre against rPR region compared to control non-immunized group. However, immunized mice or mice recived polyclonal antibodies against rPR region challenged via the intra-peritoneal route with a lethal dose of SP19 isolate showed no significant difference in survival compared to control non-immunized group. These results suggested that, immunization of BALB/c mice with rPR region of PspA is not protective against infection caused by serotype 19F in a mouse model.


Asunto(s)
Proteínas Bacterianas/inmunología , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/microbiología , Dominios Proteicos Ricos en Prolina , Streptococcus pneumoniae/clasificación , Streptococcus pneumoniae/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Especificidad de Anticuerpos/inmunología , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Modelos Animales de Enfermedad , Inmunización , Ratones , Infecciones Neumocócicas/prevención & control , Vacunas Neumococicas/inmunología , Dominios Proteicos Ricos en Prolina/inmunología , Conejos , Proteínas Recombinantes/inmunología
8.
J Infect Dis ; 219(9): 1439-1447, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30476337

RESUMEN

Emergence of multidrug-resistant (MDR) bacterial infections is a major problem in clinical medicine. Development of new strategies such as phage therapy may be a novel approach for treatment of life-threatening infections caused by MDR bacteria. A newly isolated phage, MMI-Ps1, with strong lytic activity was used for treatment of acute lung infection with Pseudomonas aeruginosa in a mouse model. Intranasal administration of a single dose of MMI-Ps1 immediately after infection provided a significant level of protection and increased the survival duration. Moreover, treatment of infected mice with phage as late as 12 hours after infection was still protective. Our in vitro results are the first to show the synergistic elimination of serum-resistant Pseudomonas strains by phage and complement. Phage therapy increases the efficacy of complement-mediated lysis of serum-resistant P. aeruginosa strains, indicating the importance of an intact complement system in clearing Pseudomonas infection during phage therapy.


Asunto(s)
Bacteriófagos , Proteínas del Sistema Complemento/uso terapéutico , Enfermedades Pulmonares/microbiología , Enfermedades Pulmonares/terapia , Terapia de Fagos , Infecciones por Pseudomonas/terapia , Enfermedad Aguda , Administración Intranasal , Animales , Bacteriólisis/efectos de los fármacos , Caudovirales , Recuento de Colonia Microbiana , Proteínas del Sistema Complemento/farmacología , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana Múltiple , Femenino , Técnicas In Vitro , Enfermedades Pulmonares/prevención & control , Ratones , Ratones Endogámicos BALB C , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/prevención & control , Pseudomonas aeruginosa/fisiología
9.
J Infect Dis ; 220(6): 1061-1070, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31058287

RESUMEN

BACKGROUND: Enterococcus faecalis is considered to be the most important species of enterococci responsible for blood stream infections in critically ill patients. In blood, the complement system is activated via the classical pathway (CP), the lectin pathway (LP), or the alternative pathway (AP), and it plays a critical role in opsonophagocytosis of bacteria including E faecalis. METHODS: In a mouse model of enterococcus peritonitis, BALB-C mice were challenged with a high dose of E faecalis 12 hours after intraperitoneal administration of anti-Factor H (FH) antibodies or isotype control. Four hours later, control mice developed higher bacterial burden in blood and organs compared with mice treated with anti-FH antibodies. RESULTS: We demonstrate that complement recognition molecules C1q, CL-11, and murine ficolin-A bind the enterococcus and drive the CP and the LP in human and mouse. We further describe that E faecalis evades the AP by recruitment of FH on its surface. Our results show a strong C3b deposition on E faecalis via both the CP and the LP but not through the AP. CONCLUSIONS: These findings indicate that E faecalis avoids the complement phagocytosis by the AP via sequestering complement FH from the host blood.


Asunto(s)
Factor H de Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Enterococcus faecalis/inmunología , Infecciones por Bacterias Grampositivas/inmunología , Peritonitis/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Complemento C3b/inmunología , Complemento C4b/inmunología , Lectina de Unión a Manosa de la Vía del Complemento/inmunología , Modelos Animales de Enfermedad , Humanos , Lectinas , Ratones , Ratones Endogámicos BALB C , Peritonitis/microbiología , Peritonitis/patología , Fagocitosis/inmunología , Ficolinas
10.
Infect Immun ; 87(1)2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30323030

RESUMEN

Complement is a critical component of antimicrobial immunity. Various complement regulatory proteins prevent host cells from being attacked. Many pathogens have acquired the ability to sequester complement regulators from host plasma to evade complement attack. We describe here how Streptococcus pneumoniae adopts a strategy to prevent the formation of the C3 convertase C4bC2a by the rapid conversion of surface bound C4b and iC4b into C4dg, which remains bound to the bacterial surface but no longer forms a convertase complex. Noncapsular virulence factors on the pneumococcus are thought to facilitate this process by sequestering C4b-binding protein (C4BP) from host plasma. When S. pneumoniae D39 was opsonized with human serum, the larger C4 activation products C4b and iC4b were undetectable, but the bacteria were liberally decorated with C4dg and C4BP. With targeted deletions of either PspA or PspC, C4BP deposition was markedly reduced, and there was a corresponding reduction in C4dg and an increase in the deposition of C4b and iC4b. The effect was greatest when PspA and PspC were both knocked out. Infection experiments in mice indicated that the deletion of PspA and/or PspC resulted in the loss of bacterial pathogenicity. Recombinant PspA and PspC both bound serum C4BP, and both led to increased C4b and reduced C4dg deposition on S. pneumoniae D39. We conclude that PspA and PspC help the pneumococcus to evade complement attack by binding C4BP and so inactivating C4b.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteína de Unión al Complemento C4b/metabolismo , Complemento C4b/antagonistas & inhibidores , Evasión Inmune , Streptococcus pneumoniae/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Infecciones Neumocócicas/microbiología , Unión Proteica , Streptococcus pneumoniae/patogenicidad
11.
Clin Immunol ; 197: 27-33, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30145330

RESUMEN

Acute lung injury (ALI) and/or acute respiratory distress syndrome (ARDS) are clinical conditions caused by trauma, lung infection or sepsis. ALI/ARDS is associated with massive recruitment of neutrophils into the lung with release of reactive oxygen species and excessive inflammatory response that damage alveolar tissue. Here we report the successful use of a potent recombinant chemotaxis inhibitory protein (rCHIPS) derived from Staphylococcus aureus in reducing the severity of ALI/ARDS. Treatment with rCHIPS reduces pulmonary inflammation and permeability in mice after intranasal administration of lipopolysaccharide (LPS). rCHIPS treatment significantly reduces lung myeloperoxidase (MPO) activity, pro-inflammatory cytokines, broncho-alveolar lavage (BAL) fluid protein content as well as histopathological changes. In addition, treatment with rCHIPS significantly diminishes neutrophils and leukocytes recruitment into lung tissue after LPS administration and hence protects mice from reactive oxygen species mediated lung injury. Our finding reveals potential therapeutic benefits of using rCHIPS for the treatment of ALI/ARDS.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Proteínas Bacterianas/farmacología , Citocinas/efectos de los fármacos , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Peroxidasa/efectos de los fármacos , Lesión Pulmonar Aguda/patología , Animales , Líquido del Lavado Bronquioalveolar/química , Citocinas/metabolismo , Femenino , Pulmón/metabolismo , Pulmón/patología , Ratones , Infiltración Neutrófila/efectos de los fármacos , Permeabilidad/efectos de los fármacos , Peroxidasa/metabolismo , Proteínas Recombinantes/farmacología
12.
FASEB J ; 28(9): 3996-4003, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24868011

RESUMEN

Mannan-binding lectin-associated serine protease 2 (MASP-2) has been described as the essential enzyme for the lectin pathway (LP) of complement activation. Since there is strong published evidence indicating that complement activation via the LP critically contributes to ischemia reperfusion (IR) injury, we assessed the effect of MASP-2 deficiency in an isogenic mouse model of renal transplantation. The experimental transplantation model used included nephrectomy of the remaining native kidney at d 5 post-transplantation. While wild-type (WT) kidneys grafted into WT recipients (n=7) developed acute renal failure (control group), WT grafts transplanted into MASP-2-deficient recipients (n=7) showed significantly better kidney function, less C3 deposition, and less IR injury. In the absence of donor or recipient complement C4 (n=7), the WT to WT phenotype was preserved, indicating that the MASP-2-mediated damage was independent of C4 activation. This C4-bypass MASP-2 activity was confirmed in mice deficient for both MASP-2 and C4 (n=7), where the protection from postoperative acute renal failure was no greater than in mice with MASP-2 deficiency alone. Our study highlights the role of LP activation in renal IR injury and indicates that injury occurs through MASP-2-dependent activation events independent of C4.


Asunto(s)
Complemento C4/fisiología , Enfermedades Renales/etiología , Trasplante de Riñón , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/fisiología , Complicaciones Posoperatorias , Daño por Reperfusión/etiología , Animales , Nitrógeno de la Urea Sanguínea , Complemento C3d/metabolismo , Femenino , Técnicas para Inmunoenzimas , Enfermedades Renales/metabolismo , Enfermedades Renales/cirugía , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nefrectomía , Daño por Reperfusión/metabolismo , Daño por Reperfusión/cirugía
13.
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
14.
J Am Soc Nephrol ; 24(1): 43-52, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23184055

RESUMEN

Complement factor H (CFH) is a negative regulator of the alternative pathway of complement, and properdin is the sole positive regulator. CFH-deficient mice (CFH(-/-)) develop uncontrolled C3 activation and spontaneous renal disease characterized by accumulation of C3 along the glomerular basement membrane, but the role of properdin in the pathophysiology is unknown. Here, we studied mice deficient in both CFH and properdin (CFH(-/-).P(-/-)). Although CFH(-/-) mice had plasma depleted of both C3 and C5, CFH(-/-).P(-/-) animals exhibited depletion of C3 predominantly, recapitulating the plasma complement profile observed in humans with properdin-independent C3 nephritic factors. Glomerular inflammation, thickening of the capillary wall, and glomerular C3 staining were significantly increased in CFH(-/-).P(-/-) compared with CFH(-/-) mice. We previously reported that exogenous CFH ameliorates C3 staining of the glomerular basement membrane and triggers the appearance of mesangial C3 deposits in CFH(-/-) mice; here, we show that these effects require properdin. In summary, during uncontrolled activation of C3 driven by complete CFH deficiency, properdin influences the intraglomerular localization of C3, suggesting that therapeutic inhibition of properdin would be detrimental in this setting.


Asunto(s)
Glomerulonefritis/etiología , Enfermedades Renales/complicaciones , Properdina/deficiencia , Animales , Complemento C3/metabolismo , Factor H de Complemento/deficiencia , Glomerulonefritis/metabolismo , Enfermedades por Deficiencia de Complemento Hereditario , Humanos , Glomérulos Renales/metabolismo , Ratones , Ratones Endogámicos C57BL
15.
Infect Immun ; 81(4): 1374-80, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23403557

RESUMEN

Several outer membrane proteins of Neisseria meningitidis are subject to phase variation due to alterations in simple sequence repeat tracts. The PorA protein is a major outer membrane protein and a target for protective host immune responses. Phase variation of PorA is mediated by a poly-G repeat tract present within the promoter, leading to alterations in protein expression levels. N. meningitidis strain 8047 was subjected to serial passage in the presence of P1.2, a PorA-specific bactericidal monoclonal antibody. Rapid development of resistance to bactericidal activity was associated with a switch in the PorA repeat tract from 11G to 10G. Phase variants with a 10G repeat tract exhibited a 2-fold reduction in surface expression of PorA protein. A mutS mutant of strain 8047, with an elevated phase variation rate, exhibited a higher rate of escape and an association of escape with 10G and 9G variants, the latter having a 13-fold reduction in surface expression of PorA. We conclude that graduated reductions in the surface expression of outer membrane proteins mediated by phase variation enable meningococci to escape killing in vitro by bactericidal antibodies. These findings indicate how phase variation could have a major impact on immune escape and host persistence of meningococci.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Variación Antigénica , Evasión Inmune , Neisseria meningitidis/genética , Neisseria meningitidis/inmunología , Porinas/genética , Porinas/inmunología , Adulto , Anticuerpos Monoclonales/inmunología , Perfilación de la Expresión Génica , Humanos , Viabilidad Microbiana , Neisseria meningitidis/patogenicidad , Pase Seriado
16.
Infect Immun ; 81(5): 1730-40, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23478320

RESUMEN

Aspergillus species are saprophytic molds causing life-threatening invasive fungal infections in the immunocompromised host. Innate immune recognition, in particular, the mechanisms of opsonization and complement activation, has been reported to be an integral part of the defense against fungi. We have shown that the complement component ficolin-A significantly binds to Aspergillus conidia and hyphae in a concentration-dependent manner and was inhibited by N-acetylglucosamine and N-acetylgalactosamine. Calcium-independent binding to Aspergillus fumigatus and A. terreus was observed, but binding to A. flavus and A. niger was calcium dependent. Ficolin-A binding to conidia was increased under low-pH conditions, and opsonization led to enhanced binding of conidia to A549 airway epithelial cells. In investigations of the lectin pathway of complement activation, ficolin-A-opsonized conidia did not lead to lectin pathway-specific C4 deposition. In contrast, the collectin mannose binding lectin C (MBL-C) but not MBL-A led to efficient lectin pathway activation on A. fumigatus in the absence of ficolin-A. In addition, ficolin-A opsonization led to a modulation of the proinflammatory cytokine interleukin-8. We conclude that ficolin-A may play an important role in the innate defense against Aspergillus by opsonizing conidia, immobilizing this fungus through enhanced adherence to epithelial cells and modulation of inflammation. However, it appears that other immune pattern recognition molecules, i.e., those of the collectin MBL-C, are involved in the Aspergillus-lectin complement pathway activation rather than ficolin-A.


Asunto(s)
Aspergillus/inmunología , Lectina de Unión a Manosa de la Vía del Complemento/inmunología , Inmunidad Innata/fisiología , Lectinas/fisiología , Animales , Humanos , Interleucina-8/metabolismo , Ratas , Ficolinas
17.
Front Immunol ; 14: 1192767, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37325666

RESUMEN

Acute respiratory distress syndrome (ARDS) is a life-threatening disorder with a high rate of mortality. Complement activation in ARDS initiates a robust inflammatory reaction that can cause progressive endothelial injury in the lung. Here, we tested whether inhibition of the lectin pathway of complement could reduce the pathology and improve the outcomes in a murine model of LPS-induced lung injury that closely mimics ARDS in human. In vitro, LPS binds to murine and human collectin 11, human MBL and murine MBL-A, but not to C1q, the recognition subcomponent of the classical pathway. This binding initiates deposition of the complement activation products C3b, C4b and C5b-9 on LPS via the lectin pathway. HG-4, a monoclonal antibody that targets MASP-2, a key enzyme in the lectin pathway, inhibited lectin pathway functional activity in vitro, with an IC50 of circa 10nM. Administration of HG4 (5mg/kg) in mice led to almost complete inhibition of the lectin pathway activation for 48hrs, and 50% inhibition at 60hrs post administration. Inhibition of the lectin pathway in mice prior to LPS-induced lung injury improved all pathological markers tested. HG4 reduces the protein concentration in bronchoalveolar lavage fluid (p<0.0001) and levels of myeloid peroxide (p<0.0001), LDH (p<0.0001), TNFα and IL6 (both p<0.0001). Lung injury was significantly reduced (p<0.001) and the survival time of the mice increased (p<0.01). From the previous findings we concluded that inhibition of the lectin pathway has the potential to prevent ARDS pathology.


Asunto(s)
Lesión Pulmonar , Síndrome de Dificultad Respiratoria , Animales , Humanos , Ratones , Lectinas , Lipopolisacáridos/toxicidad , Activación de Complemento , Síndrome de Dificultad Respiratoria/inducido químicamente , Complemento C3b/metabolismo
18.
Front Immunol ; 13: 841759, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35572551

RESUMEN

A high incidence of secondary Klebsiella pneumoniae and Staphylococcus aureus infection were observed in patients with severe COVID-19. The cause of this predisposition to infection is unclear. Our data demonstrate consumption of complement in acute COVID-19 patients reflected by low levels of C3, C4, and loss of haemolytic activity. Given that the elimination of Gram-negative bacteria depends in part on complement-mediated lysis, we hypothesised that secondary hypocomplementaemia is rendering the antibody-dependent classical pathway activation inactive and compromises serum bactericidal activity (SBA). 217 patients with severe COVID-19 were studied. 142 patients suffered secondary bacterial infections. Klebsiella species were the most common Gram-negative organism, found in 58 patients, while S. aureus was the dominant Gram-positive organism found in 22 patients. Hypocomplementaemia was observed in patients with acute severe COVID-19 but not in convalescent survivors three months after discharge. Sera from patients with acute COVID-19 were unable to opsonise either K. pneumoniae or S. aureus and had impaired complement-mediated killing of Klebsiella. We conclude that hyperactivation of complement during acute COVID-19 leads to secondary hypocomplementaemia and predisposes to opportunistic infections.


Asunto(s)
COVID-19 , Infecciones Estafilocócicas , Proteínas del Sistema Complemento , Enfermedades por Deficiencia de Complemento Hereditario , Humanos , Klebsiella pneumoniae , Staphylococcus aureus
19.
Front Immunol ; 12: 714511, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34290717

RESUMEN

Early and persistent activation of complement is considered to play a key role in the pathogenesis of COVID-19. Complement activation products orchestrate a proinflammatory environment that might be critical for the induction and maintenance of a severe inflammatory response to SARS-CoV-2 by recruiting cells of the cellular immune system to the sites of infection and shifting their state of activation towards an inflammatory phenotype. It precedes pathophysiological milestone events like the cytokine storm, progressive endothelial injury triggering microangiopathy, and further complement activation, and causes an acute respiratory distress syndrome (ARDS). To date, the application of antiviral drugs and corticosteroids have shown efficacy in the early stages of SARS-CoV-2 infection, but failed to ameliorate disease severity in patients who progressed to severe COVID-19 pathology. This report demonstrates that lectin pathway (LP) recognition molecules of the complement system, such as MBL, FCN-2 and CL-11, bind to SARS-CoV-2 S- and N-proteins, with subsequent activation of LP-mediated C3b and C4b deposition. In addition, our results confirm and underline that the N-protein of SARS-CoV-2 binds directly to the LP- effector enzyme MASP-2 and activates complement. Inhibition of the LP using an inhibitory monoclonal antibody against MASP-2 effectively blocks LP-mediated complement activation. FACS analyses using transfected HEK-293 cells expressing SARS-CoV-2 S protein confirm a robust LP-dependent C3b deposition on the cell surface which is inhibited by the MASP-2 inhibitory antibody. In light of our present results, and the encouraging performance of our clinical candidate MASP-2 inhibitor Narsoplimab in recently published clinical trials, we suggest that the targeting of MASP-2 provides an unsurpassed window of therapeutic efficacy for the treatment of severe COVID-19.


Asunto(s)
COVID-19/sangre , Activación de Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Lectinas/sangre , Insuficiencia Renal Crónica/metabolismo , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Pueblo Asiatico , Biomarcadores/sangre , COVID-19/complicaciones , COVID-19/patología , COVID-19/fisiopatología , Estudios de Cohortes , Proteínas del Sistema Complemento/inmunología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/virología , Índice de Severidad de la Enfermedad , Población Blanca
20.
J Microbiol Immunol Infect ; 51(3): 312-320, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28291719

RESUMEN

BACKGROUND: Pseudomonas aeruginosa is a Gram-negative opportunistic bacterium, which considered as a common cause of nosocomial infection and life-threatening complications in immunocompromized and cystic fibrosis patients. Here, we evaluate the protective effect of recombinant vaccines composed of outer membrane proteins OprF and OprI alone or in combination with flagellin B against mucoid and nonmucoid pseudomonas infection. METHODS: BALB/C mice were immunized subcutaneous using OprF and OprI with or without flagellin B and antibody titers were determined. Serum bactericidal and opsonophagocytosis activities of immunized and control sera were estimated against mucoid and nonmucoid pseudomonas strains. Lung tissue sections from immunized and nonimmunized mice were analyzed and the levels of peripheral neutrophils infiltration into the lung and tissue inflammation were scored. RESULTS: Subcutaneous immunization using OprF and OprI with or without flagellin B elicited higher antibody titers against OprF, OprI, and flagellin B. The produced antibodies successfully opsonized both mucoid and nonmucoid strains with subsequent activation of the terminal pathway of complement that enhances killing of nonmucoid strains via complement-mediated lysis. Furthermore, opsonized mucoid and nonmucoid strains showed enhanced opsonophagocytosis via human peripheral neutrophils, a mechanism that kills P. aeruginosa when complement mediated lysis is not effective especially with mucoid strains. Immunized mice also showed a significant prolonged survival time, lower bacteremia, and reduced lung damage when compared with control nonimmunized mice. CONCLUSION: Our data showed that mice immunized with OprF/OprI or OprF/OprI and flagellin B are significantly protected from infection caused by mucoid and nonmucoid strains of P. aeruginosa.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Bacterianas/inmunología , Flagelina/inmunología , Inmunización , Lipoproteínas/inmunología , Infecciones por Pseudomonas/prevención & control , Vacunas contra la Infección por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Infecciones del Sistema Respiratorio/prevención & control , Animales , Anticuerpos Antibacterianos/sangre , Proteínas Bacterianas/genética , Clonación Molecular , Modelos Animales de Enfermedad , Femenino , Flagelina/genética , Regulación Bacteriana de la Expresión Génica , Lipoproteínas/genética , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Fagocitosis , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/microbiología , Vacunas contra la Infección por Pseudomonas/genética , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidad , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Infecciones del Sistema Respiratorio/microbiología , Vacunación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA