Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Res Vet Sci ; 136: 377-384, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33799167

RESUMEN

BACKGROUND: Osteoarthritis is currently one of the most common chronic diseases. As life expectancy increases, its prevalence and incidence are expected to rise. At present, more and more evidences prove the correlation between the complement system and osteoarthritis (OA). This study aims to investigate complement C5's influence on the effect of MK801 on osteoarthritis synovial fibroblasts (OA-SFs). METHODS: We used IL-1b to induce OA-SFs derived from mice to obtain OA-SFs. And we performed RT-PCR and Western Blot assays to evaluate the expression levels of associated mRNA and protein. The alteration of MAC expression on OA-SFs cell membrane was evaluated by immunofluorescence assay. The expression of related inflammatory factors of OA-SFs was evaluated by ELISA experiment. RESULTS: MK801 could significantly inhibit the expression of osteoarthritis (OA) marker factors, such as: membrane attack complex (MAC), tumor necrosis factor-α (TNF-α) and matrix metalloproteinase-13 (MMP13). Meanwhile, MK801 can significantly inhibit the expression of complement C5 (C5) in OA-SFs. Immunofluorescence assay showed that MAC expression on OA-SFs cell membrane was significantly inhibited by MK801. The nucleo-plasmic separation experiment demonstrated that MK801 could significantly inhibit the activation of Nuclear factor-κB (NF-κB) signaling pathway in OA-SFs. Futhermore, koncking down the expression of C5 reversed the inhibition MK801 on the expression of OA-SFs inflammatory factors. CONCLUSIONS: These results illustrated two points: first, MK801 inhibited the generation of MAC and the release of inflammation factors in OA-SFs through C5; second: MK801 inhibited the activation of NF-κB signaling pathway in OA-SFs.


Asunto(s)
Complemento C5/metabolismo , Maleato de Dizocilpina/uso terapéutico , Fibroblastos/efectos de los fármacos , Osteoartritis/tratamiento farmacológico , Animales , Western Blotting , Células Cultivadas , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Fibroblastos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Osteoartritis/metabolismo , Osteoartritis/patología , ARN Mensajero , Transducción de Señal , Líquido Sinovial/efectos de los fármacos , Líquido Sinovial/inmunología , Membrana Sinovial/citología , Factor de Necrosis Tumoral alfa/metabolismo
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.
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
4.
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
5.
Sci Rep ; 8(1): 11286, 2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30050126

RESUMEN

Borrelia (B.) bavariensis, B. burgdorferi, B. afzelii, B. garinii, B. spielmanii, and B. mayonii are the causative agents in Lyme disease. Lyme disease spirochetes reside in infected Ixodes ticks and are transferred to mammalian hosts during tick feeding. Once transmitted, spirochetes must overcome the first line of defense of the innate immune system either by binding complement regulators or by terminating the formation of the membrane attack complex (MAC). In B. bavariensis, the proteins BGA66 and BGA71 inhibit complement activation by interacting with the late complement components C7, C8, and C9, as well as with the formed MAC. In this study, we have determined the crystal structure of the potent MAC inhibitor BGA71 at 2.9 Ǻ resolution. The structure revealed a cysteine cross-linked homodimer. Based on the crystal structure of BGA71 and the structure-based sequence alignment with CspA from B. burgdorferi, we have proposed a potential binding site for C7 and C9, both of which are constituents of the formed MAC. Our results shed light on the molecular mechanism of immune evasion developed by the human pathogenic Borrelia species to overcome innate immunity. These results will aid in the understanding of Lyme disease pathogenesis and pave the way for the development of new strategies to prevent Lyme disease.


Asunto(s)
Borrelia/enzimología , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Factores Inmunológicos/química , Factores Inmunológicos/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Multimerización de Proteína
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.
J Mal Vasc ; 40(6): 384-90, 2015 Dec.
Artículo en Francés | MEDLINE | ID: mdl-26205796

RESUMEN

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare acquired disorder of hematopoietic stem cells. Somatic mutation in the phosphatidylinositol glycan class A (PIG-A), X-linked gene, is responsible for a deficiency in glycosphosphatidylinositol-anchored proteins (GPI-AP). The lack of one of the GPI-AP complement regulatory proteins (CD55, CD59) leads to hemolysis. The disease is diagnosed with hemolytic anemia, marrow failure and thrombosis. Thromboembolic complication occurs in 30% of patient after 10 years of follow-up and is the first event in one out of 10 patients. The two most common sites are hepatic and cerebral veins. These locations are correlated with high risk of death. Currently, these data are balanced with the use of a monoclonal antibody (Eculizumab), which has significantly improved the prognosis with a survival similar to general population after 36 months of follow-up. Anticoagulant treatment is recommended after a thromboembolic event but has no place in primary prophylaxis.


Asunto(s)
Hemoglobinuria Paroxística/sangre , Trombofilia/etiología , Trombosis/etiología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticoagulantes/uso terapéutico , Trasplante de Médula Ósea , Antígenos CD55/fisiología , Antígenos CD59/fisiología , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , GMP Cíclico/metabolismo , Manejo de la Enfermedad , Endotelio Vascular/patología , Femenino , Estudios de Seguimiento , Glicosilfosfatidilinositoles/metabolismo , Hemoglobinuria Paroxística/complicaciones , Hemoglobinuria Paroxística/genética , Hemoglobinuria Paroxística/terapia , Humanos , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Óxido Nítrico/metabolismo , Guías de Práctica Clínica como Asunto , Trombofilia/tratamiento farmacológico , Trombosis/tratamiento farmacológico , Trombosis/prevención & control
8.
J Exp Med ; 211(9): 1793-805, 2014 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-25113972

RESUMEN

Liver resection is commonly performed under ischemic conditions, resulting in two types of insult to the remnant liver: ischemia reperfusion injury (IRI) and loss of liver mass. Complement inhibition is recognized as a potential therapeutic modality for IRI, but early complement activation products are also essential for liver regeneration. We describe a novel site-targeted murine complement inhibitor, CR2-CD59, which specifically inhibits the terminal membrane attack complex (MAC), and we use this protein to investigate the complement-dependent balance between liver injury and regeneration in a clinical setting of pharmacological inhibition. CR2-CD59 did not impact in vivo generation of C3 and C5 activation products but was as effective as the C3 activation inhibitor CR2-Crry at ameliorating hepatic IRI, indicating that the MAC is the principle mediator of hepatic IRI. Furthermore, unlike C3 or C5 inhibition, CR2-CD59 was not only protective but significantly enhanced hepatocyte proliferation after partial hepatectomy, including when combined with ischemia and reperfusion. Remarkably, CR2-CD59 also enhanced regeneration after 90% hepatectomy and improved long-term survival from 0 to 70%. CR2-CD59 functioned by increasing hepatic TNF and IL-6 levels with associated STAT3 and Akt activation, and by preventing mitochondrial depolarization and allowing recovery of ATP stores.


Asunto(s)
Activación de Complemento , Regeneración Hepática/inmunología , Regeneración Hepática/fisiología , Hígado/lesiones , Daño por Reperfusión/prevención & control , Adenosina Trifosfato/metabolismo , Animales , Antígenos CD59/administración & dosificación , Antígenos CD59/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hepatectomía/efectos adversos , Cinética , Hígado/inmunología , Hígado/fisiopatología , Fallo Hepático Agudo/etiología , Fallo Hepático Agudo/inmunología , Fallo Hepático Agudo/terapia , Trasplante de Hígado/efectos adversos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Hepáticas/metabolismo , Receptores de Complemento 3d/administración & dosificación , Receptores de Complemento 3d/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/metabolismo , Daño por Reperfusión/inmunología , Daño por Reperfusión/fisiopatología , Transducción de Señal , Distribución Tisular
9.
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
11.
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
12.
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
13.
Immunol Res ; 56(2-3): 477-91, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23615835

RESUMEN

The complement system is a component of the innate immune system. Its main function was initially believed to be limited to the recognition and elimination of pathogens through direct killing or stimulation of phagocytosis. However, in recent years, the immunoregulatory functions of the complement system were demonstrated and it was determined that the complement proteins play an important role in modulating adaptive immunity and in bridging innate and adaptive responses. When the delicate mechanisms that regulate this sophisticated enzymatic system are unbalanced, the complement system may cause damage, mediating tissue inflammation. Dysregulation of the complement system has been involved in the pathogenesis and clinical manifestations of several autoimmune diseases, such as systemic lupus erythematosus, vasculitides, Sjögren's syndrome, antiphospholipid syndrome, systemic sclerosis, dermatomyositis, and rheumatoid arthritis. Complement deficiencies have been associated with an increased risk to develop autoimmune disorders. Because of its functions, the complement system is an attractive therapeutic target for a wide range of diseases. Up to date, several compounds interfering with the complement cascade have been studied in experimental models for autoimmune diseases. The main therapeutic strategies are inhibition of complement activation components, inhibition of complement receptors, and inhibition of membrane attack complex. At present, none of the available agents was proven to be both safe and effective for treatment of autoimmune diseases in humans. Nonetheless, data from preclinical studies and initial clinical trials suggest that the modulation of the complement system could constitute a viable strategy for the treatment of autoimmune conditions in the decades to come.


Asunto(s)
Antiinflamatorios/uso terapéutico , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Proteínas del Sistema Complemento/inmunología , Inmunoterapia/métodos , Inmunidad Adaptativa , Animales , Antiinflamatorios/farmacología , Autoinmunidad , Activación de Complemento/efectos de los fármacos , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Inmunidad Innata , Receptores de Complemento/antagonistas & inhibidores , Riesgo
14.
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
15.
Acta Haematol ; 126(2): 103-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21597283

RESUMEN

The clinical and biological spectrum of paroxysmal nocturnal hemoglobinuria (PNH) is variable, ranging from classical hemolytic forms to PNH associated with aplastic anemia or other bone marrow (BM) failure syndromes. We report a previously undescribed case of PNH occurring after autologous stem cell transplantation (ASCT) in a patient affected by relapsing non-Hodgkin's lymphoma. The intensive chemotherapy and the ASCT resulted in a contraction of the effective hematopoietic stem cell (HSC) pool and a derangement of the immune system. The delayed engraftment and the BM hypoplasia represented a favorable environment for the expansion of the pathological clone. This case is paradigmatic even for the unexpected trend of the PNH clone during treatment with the terminal complement inhibitor eculizumab; in fact, the clone reduced until undergoing unexpected extinction, i.e. the recovery of normal hematopoiesis. Eculizumab seems not to play a direct role in HSC kinetics; the clinical remission probably occurred because the environmental conditions that led to the expansion of the PNH clone were transient and disappeared.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Hemoglobinuria Paroxística/tratamiento farmacológico , Trasplante de Células Madre de Sangre Periférica/efectos adversos , Anticuerpos Monoclonales Humanizados , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Supervivencia de Injerto/efectos de los fármacos , Hemoglobinuria Paroxística/etiología , Humanos , Linfoma no Hodgkin/terapia , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/terapia , Trasplante Autólogo
16.
Retina ; 29(6 Suppl): S45-8, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19553801

RESUMEN

Vascular endothelial growth factor (VEGF) is an important mediator of angiogenesis in age-related macular degeneration (AMD) and is a validated therapeutic target. However, combination of anti-VEGF agents with complementary inhibition of other mediators of angiogenesis, such as platelet-derived growth factor and integrin alpha5beta1, may result in enhanced visual acuity. Other concomitant treatments, such as inhibitors of inflammation, may generate an even stronger barrier to the progression of AMD than that now observed in individuals receiving anti-VEGF therapies alone. Experimental studies are providing support for the general principles of combination treatment in AMD.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neovascularización Coroidal/tratamiento farmacológico , Quimioterapia Combinada , Degeneración Macular/tratamiento farmacológico , Anticuerpos Monoclonales/uso terapéutico , Complemento C5a/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Humanos , Integrina alfa5beta1/inmunología , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
17.
Vet Immunol Immunopathol ; 120(3-4): 246-53, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17825428

RESUMEN

The channel catfish, Ictalurus punctatus, complement membrane attack complex inhibitor CD59 gene was cloned and analyzed. Total RNA from tissues was isolated and cDNA libraries were constructed by the rapid amplification cDNA end (RACE) method. The gene-specific primers in conjunction with the RACE primers were used to PCR amplify 5'- and 3'-ends of the CD59 transcript. The complete channel catfish CD59 cDNA comprised 1109 bp including a 132-bp 5'-untranslated, a 360-bp open reading frame, and a 617-bp 3'-untranslated region. The open reading frame encodes a putative protein of 119-amino acid residues with calculated molecular mass (without potential glycosylation) of 13.2 kDa. However, the CD59 protein has a potential N-glycosylation site at the Asn35 residue. The degree of conservation of the channel catfish amino acid sequence to mammalian counterparts is 24-32%, while to those of other fish species is 44-54%. One remarkable feature is that the number and position of cysteine residues were conserved in the mature protein among species examined, suggesting that although the primary amino acid sequences are divergent, the three-dimensional structure of CD59 via disulfide linkages may be conserved through the evolutionary process. The putative protein could be further divided into three domains: a 21-amino acid signal peptide at the N-terminus, a 72-amino acid mature protein, and a 26-amino acid glycosylphosphatidylinositol (GPI) anchoring signal peptide at the carboxyl terminus. CD59 was expressed in all channel catfish tissues studied, suggesting that like mammals, channel catfish CD59 is constitutively expressed.


Asunto(s)
Antígenos CD59/genética , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Regulación de la Expresión Génica , Ictaluridae/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Antígenos CD59/química , Clonación Molecular , Datos de Secuencia Molecular , Filogenia
18.
J Immunol ; 179(1): 172-8, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17579035

RESUMEN

Passive Heymann nephritis (PHN), a model of human membranous nephritis, is induced in susceptible rat strains by injection of heterologous antisera to rat renal tubular Ag extract. PHN is currently considered the archetypal complement-dependent form of nephritis, with the proteinuria resulting from sublytic glomerular epithelial cell injury induced by the complement membrane attack complex (MAC) of C5b-9. This study examined whether C6 and MAC are essential to the development of proteinuria in PHN by comparing the effect of injection of anti-Fx1A antisera into PVG rats deficient in C6 (PVG/C6(-)) and normal PVG rats (PVG/c). PVG/c and PVG/C6(-) rats developed similar levels of proteinuria at 3, 7, 14, and 28 days following injection of antisera. Isolated whole glomeruli showed similar deposition of rat Ig and C3 staining in PVG/c and PVG/C6(-) rats. C9 deposition was abundant in PVG/c but was not detected in PVG/C6(-) glomeruli, indicating C5b-9/MAC had not formed in PVG/C6(-) rats. There was also no difference in the glomerular cellular infiltrate of T cells and macrophages nor the size of glomerular basement membrane deposits measured on electron micrographs. To examine whether T cells effect injury, rats were depleted of CD8+ T cells which did not affect proteinuria in the early heterologous phase but prevented the increase in proteinuria associated with the later autologous phase. These studies showed proteinuria in PHN occurs without MAC and that other mechanisms, such as immune complex size, early complement components, CD4+ and CD8+ T cells, disrupt glomerular integrity and lead to proteinuria.


Asunto(s)
Complemento C6/deficiencia , Complemento C6/genética , Glomerulonefritis Membranosa/genética , Glomerulonefritis Membranosa/inmunología , Animales , Complejo Antígeno-Anticuerpo/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Movimiento Celular/genética , Movimiento Celular/inmunología , Complemento C3/metabolismo , Complemento C9/deficiencia , Complemento C9/metabolismo , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/biosíntesis , Modelos Animales de Enfermedad , Glomerulonefritis Membranosa/patología , Inmunoglobulinas/metabolismo , Corteza Renal/inmunología , Corteza Renal/patología , Corteza Renal/ultraestructura , Linfopenia/genética , Linfopenia/inmunología , Linfopenia/patología , Macrófagos/patología , Masculino , Proteinuria/genética , Proteinuria/inmunología , Proteinuria/patología , Ratas , Ratas Mutantes , Subgrupos de Linfocitos T/patología
19.
J Immunol ; 174(10): 6227-32, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15879120

RESUMEN

Complement component C5 binds to components C6 and C7 in reversible reactions that are distinct from the essentially nonreversible associations that form during assembly of the complement membrane attack complex (MAC). We previously reported that the approximately 150-aa residue C345C domain (also known as NTR) of C5 mediates these reversible reactions, and that the corresponding recombinant module (rC5-C345C) binds directly to the tandem pair of approximately 75-residue factor I modules from C7 (C7-FIMs). We suggested from these and other observations that binding of the C345C module of C5 to the FIMs of C7, but not C6, is also essential for MAC assembly itself. The present report describes a novel method for assembling a complex that appears to closely resemble the MAC on the sensor chip of a surface plasmon resonance instrument using the complement-reactive lysis mechanism. This method provides the ability to monitor individually the incorporation of C7, C8, and C9 into the complex. Using this method, we found that C7 binds to surface-bound C5b,6 with a K(d) of approximately 3 pM, and that micromolar concentrations of either rC5-C345C or rC7-FIMs inhibit this early step in MAC formation. We also found that similar concentrations of either module inhibited complement-mediated erythrocyte lysis by both the reactive lysis and classical pathway mechanisms. These results demonstrate that the interaction between the C345C domain of C5 and the FIMs of C7, which mediates reversible binding of C5 to C7 in solution, also plays an essential role in MAC formation and complement lytic activity.


Asunto(s)
Complemento C5/fisiología , Complemento C7/antagonistas & inhibidores , Complemento C7/fisiología , Factor I de Complemento/fisiología , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Cisteína , Proteínas Recombinantes/farmacología , Secuencias de Aminoácidos , Animales , Unión Competitiva , Complemento C5/metabolismo , Complemento C5b , Complemento C7/metabolismo , Proteínas Inactivadoras de Complemento/antagonistas & inhibidores , Proteínas Inactivadoras de Complemento/metabolismo , Proteínas Inactivadoras de Complemento/fisiología , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Vía Clásica del Complemento/inmunología , Hemólisis/inmunología , Humanos , Cinética , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ovinos , Resonancia por Plasmón de Superficie/instrumentación , Resonancia por Plasmón de Superficie/métodos , Propiedades de Superficie
20.
Immunobiology ; 209(8): 629-35, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15638131

RESUMEN

Inhibiting complement anaphlytoxin C5a during sepsis may prevent sepsis mortality. Although human anti-C5 antibodies exist, their therapeutic use in microbial sepsis has been avoided because of the hypothesis that inhibiting C5b will prevent formation of the bactericidal membrane attack complex (MAC) and worsen clinical outcome. We wished to test the hypothesis that inhibition of C5 would improve outcomes in sepsis. Sepsis was induced in rats by laparotomy and cecal ligation and puncture (CLP) by an IACUC-approved protocol. Sham animals underwent laparotomy without CLP. Following CLP rats were randomized to receive a single IV dose of purified IgG ant-C5 antibody (Ab) or control IgG Ab. Anti-C5 Ab treated rats (n = 20) had significantly lower mortality vs. controls (n = 21), 20% vs. 52% (P = 0.019, log-rank). Analysis of bacterial load by culture of spleen and liver homogenates showed a reduction in colony forming units in anti-C5 Ab treated rats vs. control IgG (P = 0.003 and 0.009, respectively). Anti-C5 treatment reduced lung injury as measured by total MPO content of lung tissue (P = 0.024). Finally, rats genetically deficient in C6 production, unable to form MAC but capable of producing C5a and C5b, were protected from CLP-induced sepsis mortality. Our results show that in anti-C5 antibody therapy prevents CLP sepsis-induced mortality and improves lung injury. Inhibition of the complement MAC does not increase bacterial load or mortality, therefore, the use of anti-C5 therapy may be beneficial rather than detrimental in sepsis.


Asunto(s)
Complemento C5/antagonistas & inhibidores , Complemento C6/genética , Sepsis/tratamiento farmacológico , Sepsis/mortalidad , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Anticuerpos/uso terapéutico , Complemento C5/inmunología , Complemento C6/deficiencia , Complejo de Ataque a Membrana del Sistema Complemento/análisis , Complejo de Ataque a Membrana del Sistema Complemento/antagonistas & inhibidores , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Regulación hacia Abajo , Inmunoglobulina G/inmunología , Inmunoglobulina G/farmacología , Inmunoglobulina G/uso terapéutico , Interleucina-6/sangre , Interleucina-6/metabolismo , Hígado/microbiología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Masculino , Mutación/genética , Ratas , Ratas Endogámicas , Sepsis/genética , Bazo/microbiología , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA