Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
Clin Immunol ; 219: 108555, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32771488

RESUMEN

Respiratory failure and acute kidney injury (AKI) are associated with high mortality in SARS-CoV-2-associated Coronavirus disease 2019 (COVID-19). These manifestations are linked to a hypercoaguable, pro-inflammatory state with persistent, systemic complement activation. Three critical COVID-19 patients recalcitrant to multiple interventions had skin biopsies documenting deposition of the terminal complement component C5b-9, the lectin complement pathway enzyme MASP2, and C4d in microvascular endothelium. Administration of anti-C5 monoclonal antibody eculizumab led to a marked decline in D-dimers and neutrophil counts in all three cases, and normalization of liver functions and creatinine in two. One patient with severe heart failure and AKI had a complete remission. The other two individuals had partial remissions, one with resolution of his AKI but ultimately succumbing to respiratory failure, and another with a significant decline in FiO2 requirements, but persistent renal failure. In conclusion, anti-complement therapy may be beneficial in at least some patients with critical COVID-19.


Asunto(s)
Lesión Renal Aguda/inmunología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Betacoronavirus/patogenicidad , Inactivadores del Complemento/uso terapéutico , Infecciones por Coronavirus/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Neumonía Viral/inmunología , Síndrome Respiratorio Agudo Grave/inmunología , Lesión Renal Aguda/complicaciones , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/virología , Adulto , Betacoronavirus/inmunología , Biomarcadores/metabolismo , COVID-19 , Activación de Complemento/efectos de los fármacos , Complemento C4b/antagonistas & inhibidores , Complemento C5/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Infecciones por Coronavirus/complicaciones , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Síndrome de Liberación de Citoquinas/complicaciones , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/virología , Femenino , Productos de Degradación de Fibrina-Fibrinógeno/metabolismo , Humanos , Inmunidad Humoral/efectos de los fármacos , Masculino , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/genética , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/inmunología , Persona de Mediana Edad , Neutrófilos/inmunología , Neutrófilos/patología , Pandemias , Fragmentos de Péptidos/antagonistas & inhibidores , Neumonía Viral/complicaciones , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/complicaciones , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Síndrome Respiratorio Agudo Grave/virología
2.
Osteoarthritis Cartilage ; 28(5): 685-697, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31981738

RESUMEN

OBJECTIVE: Innate immune response and particularly terminal complement complex (TCC) deposition are thought to be involved in the pathogenesis of posttraumatic osteoarthritis. However, the possible role of TCC in regulated cell death as well as chondrocyte hypertrophy and senescence has not been unraveled so far and was first addressed using an ex vivo human cartilage trauma-model. DESIGN: Cartilage explants were subjected to blunt impact (0.59 J) and exposed to human serum (HS) and cartilage homogenate (HG) with or without different potential therapeutics: RIPK1-inhibitor Necrostatin-1 (Nec), caspase-inhibitor zVAD, antioxidant N-acetyl cysteine (NAC) and TCC-inhibitors aurintricarboxylic acid (ATA) and clusterin (CLU). Cell death and hypertrophy/senescence-associated markers were evaluated on mRNA and protein level. RESULTS: Addition of HS resulted in significantly enhanced TCC deposition on chondrocytes and decrease of cell viability after trauma. This effect was potentiated by HG and was associated with expression of RIPK3, MLKL and CASP8. Cytotoxicity of HS could be prevented by heat-inactivation or specific inhibitors, whereby combination of Nec and zVAD as well as ATA exhibited highest cell protection. Moreover, HS+HG exposition enhanced the gene expression of CXCL1, IL-8, RUNX2 and VEGFA as well as secretion of IL-6 after cartilage trauma. CONCLUSIONS: Our findings imply crucial involvement of the complement system and primarily TCC in regulated cell death and phenotypic changes of chondrocytes after cartilage trauma. Inhibition of TCC formation or downstream signaling largely modified serum-induced pathophysiologic effects and might therefore represent a therapeutic target to maintain the survival and chondrogenic character of cartilage cells.


Asunto(s)
Muerte Celular/genética , Condrocitos/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/genética , Hipertrofia/genética , Osteoartritis/genética , Heridas no Penetrantes/genética , Acetilcisteína/farmacología , Anciano , Anciano de 80 o más Años , Ácido Aurintricarboxílico/farmacología , Cartílago Articular/citología , Muerte Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , Condrocitos/efectos de los fármacos , Condrocitos/patología , Clusterina/farmacología , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/efectos de los fármacos , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Depuradores de Radicales Libres/farmacología , Humanos , Imidazoles/farmacología , Inmunidad Innata/genética , Indoles/farmacología , Masculino , Persona de Mediana Edad , Oligopéptidos/farmacología , Osteoartritis/etiología , Osteoartritis/metabolismo , Osteoartritis/patología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Heridas no Penetrantes/complicaciones , Heridas no Penetrantes/metabolismo
3.
Acta Pharmacol Sin ; 41(11): 1433-1445, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32404983

RESUMEN

In general, anti-inflammatory treatment is considered for multiple liver diseases despite the etiology. But current drugs for alleviating liver inflammation have defects, making it necessary to develop more potent and safer drugs for liver injury. In this study, we screened a series of (dihydro-)stilbene or (dihydro-)phenanthrene derivatives extracted from Pholidota chinensis for their potential biological activities. Among 31 compounds, the dihydro-stilbene gigantol exerted most potent protective effects on human hepatocytes against lithocholic acid toxicity, and exhibited solid antioxidative and anti-inflammatory effect in vitro. In mice with CCl4-induced acute liver injury, pre-administration of gigantol (10, 20, 40 mg· kg-1· d-1, po, for 7 days) dose-dependently decreased serum transaminase levels and improved pathological changes in liver tissues. The elevated lipid peroxidation and inflammatory responses in the livers were also significantly alleviated by gigantol. The pharmacokinetic studies showed that gigantol was highly concentrated in the mouse livers, which consisted with its efficacy in preventing liver injury. Using a label-free quantitative proteomic analysis we revealed that gigantol mainly regulated the immune system process in liver tissues of CCl4-treated mice, and the complement and coagulation cascades was the predominant pathway; gigantol markedly inhibited the expression of complement component C9, which was a key component for the formation of terminal complement complex (TCC) C5b-9. These results were validated by immunohistochemistry (IHC) or real time-PCR. Confocal microscopy analysis showed that gigantol significantly inhibited the vascular deposition of TCC in the liver. In conclusion, we demonstrate for the first time that oral administration of gigantol potently relieves liver oxidative stress and inflammation, possibly via a novel mechanism of inhibiting the C5b-9 formation in the liver.


Asunto(s)
Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , Bibencilos/uso terapéutico , Guayacol/análogos & derivados , Inflamación/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Administración Oral , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacocinética , Antioxidantes/administración & dosificación , Antioxidantes/farmacocinética , Bibencilos/administración & dosificación , Bibencilos/farmacocinética , Tetracloruro de Carbono , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Guayacol/administración & dosificación , Guayacol/farmacocinética , Guayacol/uso terapéutico , Hepatocitos/efectos de los fármacos , Humanos , Inflamación/patología , Peroxidación de Lípido/efectos de los fármacos , Ácido Litocólico , Hígado/patología , Masculino , Ratones Endogámicos ICR , Fenantrenos/farmacología , Fenantrenos/uso terapéutico , Proteoma/metabolismo , Ratas Sprague-Dawley , Estilbenos/farmacología , Estilbenos/uso terapéutico
4.
J Neurosci ; 38(10): 2519-2532, 2018 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-29437855

RESUMEN

The complement system is implicated in promoting acute secondary injury after traumatic brain injury (TBI), but its role in chronic post-traumatic neuropathology remains unclear. Using various injury-site targeted complement inhibitors that block different complement pathways and activation products, we investigated how complement is involved in neurodegeneration and chronic neuroinflammation after TBI in a clinically relevant setting of complement inhibition. The current paradigm is that complement propagates post-TBI neuropathology predominantly through the terminal membrane attack complex (MAC), but the focus has been on acute outcomes. Following controlled cortical impact in adult male mice, we demonstrate that although inhibition of the MAC (with CR2-CD59) reduces acute deficits, inhibition of C3 activation is required to prevent chronic inflammation and ongoing neuronal loss. Activation of C3 triggered a sustained degenerative mechanism of microglial and astrocyte activation, reduced dendritic and synaptic density, and inhibited neuroblast migration several weeks after TBI. Moreover, inhibiting all complement pathways (with CR2-Crry), or only the alternative complement pathway (with CR2-fH), provided similar and significant improvements in chronic histological, cognitive, and functional recovery, indicating a key role for the alternative pathway in propagating chronic post-TBI pathology. Although we confirm a role for the MAC in acute neuronal loss after TBI, this study shows that upstream products of complement activation generated predominantly via the alternative pathway propagate chronic neuroinflammation, thus challenging the current concept that the MAC represents a therapeutic target for treating TBI. A humanized version of CR2fH has been shown to be safe and non-immunogenic in clinical trials.SIGNIFICANCE STATEMENT Complement, and specifically the terminal membrane attack complex, has been implicated in secondary injury and neuronal loss after TBI. However, we demonstrate here that upstream complement activation products, generated predominantly via the alternative pathway, are responsible for propagating chronic inflammation and injury following CCI. Chronic inflammatory microgliosis is triggered by sustained complement activation after CCI, and is associated with chronic loss of neurons, dendrites and synapses, a process that continues to occur even 30 d after initial impact. Acute and injury-site targeted inhibition of the alternative pathway significantly improves chronic outcomes, and together these findings modify the conceptual paradigm for targeting the complement system to treat TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/patología , Proteínas del Sistema Complemento , Inflamación/etiología , Inflamación/patología , Animales , Astrocitos/patología , Corteza Cerebral/lesiones , Activación de Complemento , Complemento C3/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Vía Alternativa del Complemento , Dendritas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/patología , Neuronas/patología , Proteínas Recombinantes de Fusión/farmacología , Recuperación de la Función/efectos de los fármacos , Sinapsis/patología
5.
Kidney Int ; 96(3): 761-768, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31345584

RESUMEN

Terminal complement inhibition therapy with eculizumab (a humanized monoclonal antibody to C5) has revolutionized the treatment of patients with thrombotic microangiopathy (TMA). Successful responders are often placed on long-standing therapy to prevent disease recurrence in the native kidney or allograft. The tissue deposition of eculizumab in patients with C3 glomerulopathy has been described but no studies have yet investigated tissue deposition of eculizumab in cases where it was indicated for thrombotic microangiopathy which, unlike C3 glomerulopathy, does not usually show immune-type electron dense deposits. To evaluate this, we reviewed biopsies from 13 patients who received eculizumab for TMA treatment or prevention of recurrence. We found IgG2, IgG4, and kappa positivity within arterioles corresponding to eculizumab deposits, with similar distribution to C5b-9, in all but one patient. In that patient eculizumab therapy had been discontinued 24 months prior to biopsy. Deposits in arterioles could be seen as early as one day after infusion and after a single dose of eculizumab, and were detected up to 162 days after therapy discontinuation. This may play a role in controlling local complement activation-associated vascular changes in these patients. Thus, IgG subclass staining by immunofluorescence is important to avoid misdiagnoses of immune-complex or monoclonal immunoglobulin deposition disease in patients with TMA who received eculizumab.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Arteriolas/patología , Inactivadores del Complemento/farmacología , Riñón/patología , Microangiopatías Trombóticas/tratamiento farmacológico , Adulto , Anciano , Anticuerpos Monoclonales Humanizados/uso terapéutico , Biopsia , Activación de Complemento/efectos de los fármacos , Inactivadores del Complemento/uso terapéutico , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Femenino , Humanos , Riñón/irrigación sanguínea , Masculino , Persona de Mediana Edad , Recurrencia , Estudios Retrospectivos , Prevención Secundaria/métodos , Microangiopatías Trombóticas/patología
6.
J Immunol ; 197(4): 1276-86, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27421478

RESUMEN

Complement activation occurs during enterohemorrhagic Escherichia coli (EHEC) infection and may exacerbate renal manifestations. In this study, we show glomerular C5b-9 deposits in the renal biopsy of a child with EHEC-associated hemolytic uremic syndrome. The role of the terminal complement complex, and its blockade as a therapeutic modality, was investigated in a mouse model of E. coli O157:H7 infection. BALB/c mice were treated with monoclonal anti-C5 i.p. on day 3 or 6 after intragastric inoculation and monitored for clinical signs of disease and weight loss for 14 d. All infected untreated mice (15 of 15) or those treated with an irrelevant Ab (8 of 8) developed severe illness. In contrast, only few infected mice treated with anti-C5 on day 3 developed symptoms (three of eight, p < 0.01 compared with mice treated with the irrelevant Ab on day 3) whereas most mice treated with anti-C5 on day 6 developed symptoms (six of eight). C6-deficient C57BL/6 mice were also inoculated with E. coli O157:H7 and only 1 of 14 developed disease, whereas 10 of 16 wild-type mice developed weight loss and severe disease (p < 0.01). Complement activation via the terminal pathway is thus involved in the development of disease in murine EHEC infection. Early blockade of the terminal complement pathway, before the development of symptoms, was largely protective, whereas late blockade was not. Likewise, lack of C6, and thereby deficient terminal complement complex, was protective in murine E. coli O157:H7 infection.


Asunto(s)
Complemento C6/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Infecciones por Escherichia coli/inmunología , Síndrome Hemolítico-Urémico/inmunología , Animales , Preescolar , Complemento C6/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Modelos Animales de Enfermedad , Escherichia coli Enterohemorrágica , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
7.
Biol Pharm Bull ; 41(10): 1600-1605, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30270330

RESUMEN

Baculovirus vectors (BVs) enable safe and efficient gene delivery to mammalian cells and are useful in a wide range of applications, including gene therapy and in vivo analysis of gene functions. We previously developed BVs expressing malaria sporozoite surface proteins for targeting liver cells or hepatocytes. However, BVs are known to be very vulnerable to complement attack and efforts to overcome their inactivation based on complement are important. In this study, BVs expressing complement regulatory proteins (CRPs) on the surfaces of virions were developed to inhibit complement reactions. Decay accelerating factor (DAF; CD55)-type BVs exhibited significantly higher complement resistance than control BVs without any CRPs in HepG2 cells transduction, although the transduction efficacy of DAF-type BV was low. In contrast, CD46-DAF-CD59 fusion type BVs showed significantly higher transduction efficacy and complement resistance than both control and DAF-type BVs. DAF-type and CD46-DAF-CD59 type BVs repressed formation of the membrane attack complex, a terminal product of complement reaction cascades, induced by BVs. These results suggest that the CD46-DAF-CD59 fusion construct confers complement protection ability superior to that of the DAF construct in gene delivery under complement active serum.


Asunto(s)
Baculoviridae/metabolismo , Proteínas del Sistema Complemento/metabolismo , Vectores Genéticos , Transducción Genética , Animales , Antígenos CD55 , Antígenos CD59 , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Terapia Genética/métodos , Células Hep G2 , Humanos , Proteína Cofactora de Membrana , Proteínas de la Membrana/metabolismo , Virión/metabolismo
8.
J Immunol ; 195(1): 257-64, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26026058

RESUMEN

Chronic inflammation of the arterial wall is a key element in the development of atherosclerosis, and cholesterol crystals (CC) that accumulate in plaques are associated with initiation and progression of the disease. We recently revealed a link between the complement system and CC-induced inflammasome caspase-1 activation, showing that the complement system is a key trigger in CC-induced inflammation. HDL exhibits cardioprotective and anti-inflammatory properties thought to explain its inverse correlation to cardiovascular risk. In this study, we sought to determine the effect of reconstituted HDL (rHDL) on CC-induced inflammation in a human whole blood model. rHDL bound to CC and inhibited the CC-induced complement activation as measured by soluble terminal C5b-9 formation and C3c deposition on the CC surface. rHDL attenuated the amount of CC-induced complement receptor 3 (CD11b/CD18) expression on monocytes and granulocytes, as well as reactive oxygen species generation. Moreover, addition of CC to whole blood resulted in release of proinflammatory cytokines that were inhibited by rHDL. Our results support and extend the notion that CC are potent triggers of inflammation, and that rHDL may have a beneficial role in controlling the CC-induced inflammatory responses by inhibiting complement deposition on the crystals.


Asunto(s)
Colesterol/efectos adversos , Activación de Complemento/efectos de los fármacos , Lipoproteínas HDL/farmacología , Células Sanguíneas/citología , Células Sanguíneas/efectos de los fármacos , Células Sanguíneas/inmunología , Antígeno CD11b/inmunología , Antígenos CD18/inmunología , Complemento C3c/antagonistas & inhibidores , Complemento C3c/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Cristalización , Humanos , Inflamación/inmunología , Inflamación/patología , Inflamación/prevención & control , Cultivo Primario de Células , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/inmunología
9.
J Immunol ; 192(5): 2339-48, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24489093

RESUMEN

Traumatic brain injury (TBI) is the leading cause of disability and death in young adults. The secondary neuroinflammation and neuronal damage that follows the primary mechanical injury is an important cause of disability in affected people. The membrane attack complex (MAC) of the complement system is detected in the traumatized brain early after TBI; however, its role in the pathology and neurologic outcome of TBI has not yet been investigated. We generated a C6 antisense oligonucleotide that blocks MAC formation by inhibiting C6, and we compared its therapeutic effect to that of Ornithodoros moubata complement inhibitor (OmCI), a known inhibitor of C5 activation that blocks generation of the anaphylatoxin C5a and C5b, an essential component of MAC. Severe closed head injury in mice induced abundant MAC deposition in the brain. Treatment with C6 antisense reduced C6 synthesis (85%) and serum levels (90%), and inhibited MAC deposition in the injured brain (91-96%). Treatment also reduced accumulation of microglia/macrophages (50-88%), neuronal apoptosis, axonal loss and weight loss (54-93%), and enhanced neurologic performance (84-92%) compared with placebo-treated controls after injury. These data provide the first evidence, to our knowledge, that inhibition of MAC formation in otherwise complement-sufficient animals reduces neuropathology and promotes neurologic recovery after TBI. Given the importance of maintaining a functional complement opsonization system to fight infections, a critical complication in TBI patients, inhibition of the MAC should be considered to reduce posttraumatic neurologic damage. This work identifies a novel therapeutic target for TBI and will guide the development of new therapy for patients.


Asunto(s)
Proteínas de Artrópodos/farmacología , Axones/inmunología , Lesiones Encefálicas/tratamiento farmacológico , Proteínas Portadoras/farmacología , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Macrófagos/inmunología , Oligodesoxirribonucleótidos Antisentido/farmacología , Animales , Axones/patología , Lesiones Encefálicas/inmunología , Lesiones Encefálicas/patología , Complemento C5a/antagonistas & inhibidores , Complemento C5a/inmunología , Complemento C5b/antagonistas & inhibidores , Complemento C5b/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Femenino , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Microglía/inmunología , Microglía/patología , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/inmunología
10.
J Intern Med ; 277(3): 294-305, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25495259

RESUMEN

First identified in human serum in the late 19th century as a 'complement' to antibodies in mediating bacterial lysis, the complement system emerged more than a billion years ago probably as the first humoral immune system. The contemporary complement system consists of nearly 60 proteins in three activation pathways (classical, alternative and lectin) and a terminal cytolytic pathway common to all. Modern molecular biology and genetics have not only led to further elucidation of the structure of complement system components, but have also revealed function-altering rare variants and common polymorphisms, particularly in regulators of the alternative pathway, that predispose to human disease by creating 'hyperinflammatory complement phenotypes'. To treat these 'complementopathies', a monoclonal antibody against the initiator of the membrane attack complex, C5, has received approval for use. Additional therapeutic reagents are on the horizon.


Asunto(s)
Trastornos de las Proteínas Sanguíneas/genética , Proteínas del Sistema Complemento/genética , Anticuerpos Monoclonales Humanizados/uso terapéutico , Trastornos de las Proteínas Sanguíneas/inmunología , Trastornos de las Proteínas Sanguíneas/terapia , Activación de Complemento/genética , Activación de Complemento/inmunología , Activación de Complemento/fisiología , Factor H de Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/fisiología , Síndrome Hemolítico-Urémico/inmunología , Humanos , Degeneración Macular/inmunología , Mutación/genética , Polimorfismo Genético/genética
11.
Biomacromolecules ; 16(11): 3594-602, 2015 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-26444518

RESUMEN

Complement activation plays an important role in innate immune defense by triggering formation of the membrane attack complex (MAC), which is a biomacromolecular assembly that exhibits membrane-lytic activity against foreign invaders including various pathogens and biomaterials. Understanding the details of MAC structure and function has been the subject of extensive work involving bulk liposome and erythrocyte assays. However, it is difficult to characterize the mechanism of action of MAC inhibitor drug candidates using the conventional assays. To address this issue, we employ a biomimetic supported lipid bilayer platform to investigate how two MAC inhibitors, vitronectin and clusterin, interfere with MAC assembly in a sequential addition format, as monitored by the quartz crystal microbalance-dissipation (QCM-D) technique. Two experimental strategies based on modular assembly were selected, precincubation of inhibitor and C5b-7 complex before addition to the lipid bilayer or initial addition of inhibitor followed by the C5b-7 complex. The findings indicate that vitronectin inhibits membrane association of C5b-7 via a direct interaction with C5b-7 and via competitive membrane association onto the supported lipid bilayer. On the other hand, clusterin directly interacts with C5b-7 such that C5b-7 is still able to bind to the lipid bilayer, and clusterin affects the subsequent binding of other complement proteins involved in the MAC assembly. Taken together, the findings in this study outline a biomimetic approach based on supported lipid bilayers to explore the interactions between complement proteins and inhibitors, thereby offering insight into MAC assembly and regulation.


Asunto(s)
Clusterina/química , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Membrana Dobles de Lípidos/química , Vitronectina/química , Biomimética , Activación de Complemento , Complemento C5/antagonistas & inhibidores , Complemento C5/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Eritrocitos/metabolismo , Humanos , Liposomas/metabolismo
12.
Gene Ther ; 21(5): 507-13, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24670995

RESUMEN

Age-related macular degeneration (AMD) is the leading cause of blindness among the elderly. Approximately 50% of AMD patients have a polymorphism in the negative regulator of complement known as Factor H. Individuals homozygous for a Y402H polymorphism in Factor H have elevated levels of membrane attack complex (MAC) in their choroid and retinal pigment epithelium relative to individuals homozygous for the wild-type allele. An inability to form MAC due to a polymorphism in C9 is protective against the formation of choroidal neovascularization (CNV) in AMD patients. Hence, blocking MAC in AMD patients may be protective against CNV. Here we investigate the potential of human proline/arginine-rich end leucine-rich repeat protein (PRELP) as an inhibitor of complement-mediated damage when delivered via the subretinal route using an AAV2/8 vector. In a fluorescence-activated cell sorting (FACS) lysis assay, PRELP inhibited normal human serum-mediated lysis of Hepa-1c1c7 cells by 18.7%. Unexpectedly, PRELP enhanced the formation of tubes by human umbilical vein endothelial cells (HUVECs) by approximately 240%, but, when delivered via an AAV vector to the retina of mice, PRELP inhibited laser-induced CNV by 60%. PRELP reduced deposition of MAC in vivo by 25.5%. Our results have implications for the development of complement inhibitors as a therapy for AMD.


Asunto(s)
Neovascularización Coroidal/prevención & control , Proteínas Inactivadoras de Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Proteínas de la Matriz Extracelular/genética , Glicoproteínas/genética , Degeneración Macular/genética , Animales , Ceguera/genética , Coroides/irrigación sanguínea , Coroides/patología , Neovascularización Coroidal/genética , Activación de Complemento , Factor H de Complemento/genética , Proteínas Inactivadoras de Complemento/biosíntesis , Complejo de Ataque a Membrana del Sistema Complemento/biosíntesis , Dependovirus/genética , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/biosíntesis , Terapia Genética , Vectores Genéticos , Glicoproteínas/biosíntesis , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Degeneración Macular/prevención & control , Ratones , Ratones Endogámicos C57BL , Polimorfismo de Nucleótido Simple , Retina/patología , Epitelio Pigmentado de la Retina/patología
14.
Biol Blood Marrow Transplant ; 20(4): 518-25, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24370861

RESUMEN

We recently observed that dysregulation of the complement system may be involved in the pathogenesis of hematopoietic stem cell transplantation-associated thrombotic microangiopathy (HSCT-TMA). These findings suggest that the complement inhibitor eculizumab could be a therapeutic option for this severe HSCT complication with high mortality. However, the efficacy of eculizumab in children with HSCT-TMA and its dosing requirements are not known. We treated 6 children with severe HSCT-TMA using eculizumab and adjusted the dose to achieve a therapeutic level >99 µg/mL. HSCT-TMA resolved over time in 4 of 6 children after achieving therapeutic eculizumab levels and complete complement blockade, as measured by low total hemolytic complement activity (CH50). To achieve therapeutic drug levels and a clinical response, children with HSCT-TMA required higher doses or more frequent eculizumab infusions than currently recommended for children with atypical hemolytic uremic syndrome. Two critically ill patients failed to reach therapeutic eculizumab levels, even after dose escalation, and subsequently died. Our data indicate that eculizumab may be a therapeutic option for HSCT-TMA, but HSCT patients appear to require higher medication dosing than recommended for other conditions. We also observed that a CH50 level ≤ 4 complement activity enzyme units correlated with therapeutic eculizumab levels and clinical response, and therefore CH50 may be useful to guide eculizumab dosing in HSCT patients as drug level monitoring is not readily available.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Inactivadores del Complemento/uso terapéutico , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Microangiopatías Trombóticas/tratamiento farmacológico , Niño , Preescolar , Ensayo de Actividad Hemolítica de Complemento , Esquema de Medicación , Cálculo de Dosificación de Drogas , Monitoreo de Drogas , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/mortalidad , Neoplasias Hematológicas/patología , Humanos , Índice de Severidad de la Enfermedad , Análisis de Supervivencia , Microangiopatías Trombóticas/etiología , Microangiopatías Trombóticas/inmunología , Microangiopatías Trombóticas/mortalidad , Trasplante Autólogo , Trasplante Homólogo
15.
J Virol ; 87(10): 5858-67, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23487461

RESUMEN

Hepatitis C virus (HCV) proteins inhibit complement component expression, which may attenuate immunity against infection. In this study, we examined whether HCV regulates the membrane attack complex (MAC) via complement component C9. MAC is composed of C5b to C9 (C5b-9) and mediates cell lysis of invaded pathogens. Liver biopsy specimens from chronically HCV-infected patients exhibited a lower level of C9 mRNA expression than liver biopsy specimens from unrelated disease or healthy control human liver RNA. Hepatocytes infected with cell culture-grown HCV or expressing HCV core protein also displayed significant repression of C9 mRNA and protein levels. Promoter analysis suggested that the T cell factor-4 (TCF-4E) transcription factor is responsible for HCV core-mediated C9 promoter regulation. Sera from chronically HCV-infected patients displayed a lower level of C5b-9 and a reduced antimicrobial effect on model organisms compared to unrelated patient sera or sera from healthy volunteers. Together, these results for C9 regulation by HCV core protein coupled with functional impairment of the membrane attack complex underscore HCV-mediated attenuation of immune mechanisms.


Asunto(s)
Complemento C9/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Hepacivirus/inmunología , Hepacivirus/patogenicidad , Biopsia , Perfilación de la Expresión Génica , Hepatitis C Crónica/patología , Humanos , Evasión Inmune , Hígado/patología
16.
J Biol Chem ; 287(11): 8092-100, 2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22267731

RESUMEN

PRELP is a 58-kDa proteoglycan found in a variety of extracellular matrices, including cartilage and at several basement membranes. In rheumatoid arthritis (RA), the cartilage tissue is destroyed and fragmented molecules, including PRELP, are released into the synovial fluid where they may interact with components of the complement system. In a previous study, PRELP was found to interact with the complement inhibitor C4b-binding protein, which was suggested to locally down-regulate complement activation in joints during RA. Here we show that PRELP directly inhibits all pathways of complement by binding C9 and thereby prevents the formation of the membrane attack complex (MAC). PRELP does not interfere with the interaction between C9 and already formed C5b-8, but inhibits C9 polymerization thereby preventing formation of the lytic pore. The alternative pathway is moreover inhibited already at the level of C3-convertase formation due to an interaction between PRELP and C3. This suggests that PRELP may down-regulate complement attack at basement membranes and on damaged cartilage and therefore limit pathological complement activation in inflammatory disease such as RA. The net outcome of PRELP-mediated complement inhibition will highly depend on the local concentration of other complement modulating molecules as well as on the local concentration of available complement proteins.


Asunto(s)
Artritis Reumatoide/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Membrana Basal/química , Membrana Basal/metabolismo , Membrana Basal/patología , Activación de Complemento/genética , Convertasas de Complemento C3-C5/química , Convertasas de Complemento C3-C5/genética , Convertasas de Complemento C3-C5/metabolismo , Proteína de Unión al Complemento C4b/química , Proteína de Unión al Complemento C4b/genética , Proteína de Unión al Complemento C4b/metabolismo , Complemento C9/química , Complemento C9/genética , Complemento C9/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/química , Complejo de Ataque a Membrana del Sistema Complemento/genética , Proteínas del Sistema Complemento/química , Proteínas del Sistema Complemento/genética , Proteínas del Sistema Complemento/metabolismo , Regulación hacia Abajo/genética , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/genética , Femenino , Glicoproteínas/química , Glicoproteínas/genética , Células HEK293 , Humanos , Masculino
17.
Arthritis Rheum ; 64(8): 2719-23, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22354668

RESUMEN

OBJECTIVE: Catastrophic antiphospholipid syndrome (CAPS) is characterized by histopathologic evidence of small vessel thrombosis, dysfunction of multiple organs occurring over a short period of time, and laboratory confirmation of the presence of antiphospholipid antibodies (aPL). Treatment of CAPS focuses on anticoagulation therapy and on removal of aPL that promote thrombosis by activating endothelial cells, monocytes, and platelets. Studies in animal models support the hypothesis that a more targeted intervention, such as complement inhibition, may be an effective means to prevent aPL-induced thrombosis. Herein we describe use of an inhibitor of complement activation to treat CAPS that was refractory to conventional therapy. METHODS: Our patient was a young man who had recurrent CAPS characterized by multiple arterial thromboses in large and small vessels despite maximal anticoagulation, immunosuppression, and plasma exchange therapy. We treated him with eculizumab, an anti-C5 monoclonal antibody that blocks activation of terminal complement. RESULTS: Administration of eculizumab, at doses that blocked complement activity, aborted acute progressive thrombotic events, reversed thrombocytopenia, and was associated with no further clinical episodes of thrombosis during >3 years of therapy. CONCLUSION: This first report of the use and clinical efficacy of eculizumab, an inhibitor of complement activation, in the treatment of CAPS demonstrates both the importance of complement (specifically, terminal complement components) in the pathogenesis of CAPS and the therapeutic benefit of complement inactivation.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Síndrome Antifosfolípido/tratamiento farmacológico , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Índice de Severidad de la Enfermedad , Adulto , Anticoagulantes/uso terapéutico , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Intercambio Plasmático , Recurrencia , Inducción de Remisión , Trombocitopenia/tratamiento farmacológico , Trombosis/tratamiento farmacológico , Insuficiencia del Tratamiento , Resultado del Tratamiento
18.
J Immunol ; 186(12): 6657-60, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21572031

RESUMEN

Cerebral malaria is the most severe complication of Plasmodium falciparum infection and accounts for a large number of malaria fatalities worldwide. Recent studies demonstrated that C5(-/-) mice are resistant to experimental cerebral malaria (ECM) and suggested that protection was due to loss of C5a-induced inflammation. Surprisingly, we observed that C5aR(-/-) mice were fully susceptible to disease, indicating that C5a is not required for ECM. C3aR(-/-) and C3aR(-/-) × C5aR(-/-) mice were equally susceptible to ECM as were wild-type mice, indicating that neither complement anaphylatoxin receptor is critical for ECM development. In contrast, C9 deposition in the brains of mice with ECM suggested an important role for the terminal complement pathway. Treatment with anti-C9 Ab significantly increased survival time and reduced mortality in ECM. Our data indicate that protection from ECM in C5(-/-) mice is mediated through inhibition of membrane attack complex formation and not through C5a-induced inflammation.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/fisiología , Malaria Cerebral/etiología , Animales , Encéfalo/inmunología , Complemento C5a/fisiología , Complemento C9/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Susceptibilidad a Enfermedades , Inflamación , Malaria Cerebral/inmunología , Ratones , Ratones Noqueados , Receptor de Anafilatoxina C5a , Receptores de Complemento , Tasa de Supervivencia
19.
Mol Microbiol ; 82(5): 1129-49, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22050461

RESUMEN

Complement evasion is an important survival strategy of Neisseria meningitidis (Nm) during colonization and infection. Previously, we have shown that Nm Opc binds to serum vitronectin to inhibit complement-mediated killing. In this study, we demonstrate meningococcal interactions with vitronectin via a novel adhesin, Msf (meningococcal surface fibril, previously NhhA or Hsf). As with Opc, Msf binds preferentially to activated vitronectin (aVn), engaging at its N-terminal region but the C-terminal heparin binding domain may also participate. However, unlike Opc, the latter binding is not heparin-mediated. By binding to aVn, Msf or Opc can impart serum resistance, which is further increased in coexpressers, a phenomenon dependent on serum aVn concentrations. The survival fitness of aVn-binding derivatives was evident from mixed population studies, in which msf/opc mutants were preferentially depleted. In addition, using vitronectin peptides to block Msf-aVn interactions, aVn-induced inhibition of lytic C5b-9 formation and of serum killing could be reversed. As Msf-encoding gene is ubiquitous in the meningococcal strains examined and is expressed in vivo, serum resistance via Msf may be of significance to meningococcal pathogenesis. The data imply that vitronectin binding may be an important strategy for the in vivo survival of Nm for which the bacterium has evolved redundant mechanisms.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Actividad Bactericida de la Sangre , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Interacciones Huésped-Patógeno , Neisseria meningitidis/inmunología , Neisseria meningitidis/patogenicidad , Vitronectina/metabolismo , Evasión Inmune , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Neisseria meningitidis/metabolismo , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas
20.
Am Heart J ; 164(1): 43-51, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22795281

RESUMEN

BACKGROUND: Reasons for pexelizumab lack of benefit in ST-elevation myocardial infarction patients undergoing primary percutaneous coronary intervention remain unclear. In a substudy of the APEX-AMI trial, we explored the hypothesis that early complement activation preceding drug administration explained the failure. METHODS: A panel of terminal complement complex proteins and fragments and biomarkers of inflammation, apoptosis, and high-risk features were assessed in serum obtained before and 24 hours after administration of placebo or pexelizumab and primary percutaneous coronary intervention (n = 356) and in human umbilical vein endothelial cell cultures coincubated with serum (n = 45). RESULTS: In the placebo group, C5a and sC5b-9 levels increased by 37% (7.9-14.2 ηg/mL, P = .007) and 96% (442-845 ηg/mL, P < .0001), respectively, during the first 24 hours. Pexelizumab prevented the increase in C5a (P = .01 vs placebo), but not that of sC5b-9 (502-1,157 ηg/mL, not significant vs placebo). Levels of C-reactive protein, interleukin (IL) 6, IL-1ß, Regulated on Activation, Normal T Cell Expressed and Secreted (RANTES) or Chemokine C-C motif ligand 5 (CCL5), and N-terminal probrain natriuretic peptide increased significantly in both groups; those of IL-10, IL-12, IL-1ra, and Interferon gamma-induced protein 10 (IP-10) or C-X-C motif chemokine 10 (CXCL10) decreased. Pexelizumab halved the increase in IL-6 (+92% vs 156%, P = .01) without effects on other markers, including C-reactive protein and N-terminal probrain natriuretic peptide. In cell culture, pexelizumab inhibited C5a, sC5b-9, and membrane-bound C5b-9 by 92%, 75%, and 78%, respectively (all P < .0001), without influencing cytokine levels and cell apoptosis. CONCLUSIONS: The blockage of both C5a and terminal complement in cell culture, but of C5a only in vivo with minimal effects on inflammation and risk biomarkers, supports the hypothesis that late administration of pexelizumab after the ischemia/reperfusion insult precluded adequate myocardial protection, resulting in a negative trial.


Asunto(s)
Angioplastia Coronaria con Balón , Anticuerpos Monoclonales Humanizados/uso terapéutico , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Infarto del Miocardio/terapia , Anticuerpos de Cadena Única/uso terapéutico , Anciano , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Insuficiencia del Tratamiento
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda