Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
1.
Transl Res ; 245: 1-17, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35158097

RESUMEN

The complement system plays crucial roles in homeostasis and host defense against microbes. Deficiency of early complement cascade components has been associated with increased susceptibility to systemic lupus erythematosus (SLE), whereas excessive complement consumption is a hallmark of this disease. Although enhanced classical pathway activation by immune complexes was initially thought to be the main contributor to lupus nephritis (LN) pathogenesis, an increasing body of evidence has suggested the alternative and the lectin pathways are also involved. Therapeutic agents targeting complement activation have been used in LN patients and clinical trials are ongoing. We review the mechanisms by which complement system dysregulation contributes to renal injury in SLE and summarize the latest evidence on the use of anticomplement agents to manage this condition.


Asunto(s)
Lupus Eritematoso Sistémico , Nefritis Lúpica , Proteínas Inactivadoras de Complemento/uso terapéutico , Proteínas del Sistema Complemento , Humanos , Riñón/patología , Lupus Eritematoso Sistémico/complicaciones
2.
Am J Kidney Dis ; 79(4): 570-581, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34571062

RESUMEN

Blocking the complement system as a therapeutic strategy has been proposed for numerous glomerular diseases but presents myriad questions and challenges, not the least of which is demonstrating efficacy and safety. In light of these potential issues and because there are an increasing number of anticomplement therapy trials either planned or under way, the National Kidney Foundation facilitated an all-virtual scientific workshop entitled "Improving Clinical Trials for Anti-Complement Therapies in Complement-Mediated Glomerulopathies." Attended by patient representatives and experts in glomerular diseases, complement physiology, and clinical trial design, the aim of this workshop was to develop standards applicable for designing and conducting clinical trials for anticomplement therapies across a wide spectrum of complement-mediated glomerulopathies. Discussions focused on study design, participant risk assessment and mitigation, laboratory measurements and biomarkers to support these studies, and identification of optimal outcome measures to detect benefit, specifically for trials in complement-mediated diseases. This report summarizes the discussions from this workshop and outlines consensus recommendations.


Asunto(s)
Proteínas Inactivadoras de Complemento , Enfermedades Renales , Proteínas Inactivadoras de Complemento/uso terapéutico , Proteínas del Sistema Complemento , Humanos , Riñón
3.
Front Immunol ; 12: 775168, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34868042

RESUMEN

COVID-19 is characterized by virus-induced injury leading to multi-organ failure, together with inflammatory reaction, endothelial cell (EC) injury, and prothrombotic coagulopathy with thrombotic events. Complement system (C) via its cross-talk with the contact and coagulation systems contributes significantly to the severity and pathological consequences due to SARS-CoV-2 infection. These immunopathological mechanisms overlap in COVID-19 and pre-eclampsia (PE). Thus, mothers contracting SARS-CoV-2 infection during pregnancy are more vulnerable to developing PE. SARS-CoV-2 infection of ECs, via its receptor ACE2 and co-receptor TMPRSS2, can provoke endothelial dysfunction and disruption of vascular integrity, causing hyperinflammation and hypercoagulability. This is aggravated by bradykinin increase due to inhibition of ACE2 activity by the virus. C is important for the progression of normal pregnancy, and its dysregulation can impact in the form of PE-like syndrome as a consequence of SARS-CoV-2 infection. Thus, there is also an overlap between treatment regimens of COVID-19 and PE. C inhibitors, especially those targeting C3 or MASP-2, are exciting options for treating COVID-19 and consequent PE. In this review, we examine the role of C, contact and coagulation systems as well as endothelial hyperactivation with respect to SARS-CoV-2 infection during pregnancy and likely development of PE.


Asunto(s)
COVID-19/inmunología , Proteínas del Sistema Complemento/inmunología , Preeclampsia/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , COVID-19/fisiopatología , Proteínas Inactivadoras de Complemento/uso terapéutico , Endotelio/inmunología , Femenino , Humanos , Preeclampsia/fisiopatología , Preeclampsia/prevención & control , Embarazo , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Complicaciones Infecciosas del Embarazo/fisiopatología , SARS-CoV-2 , Trombosis/inmunología , Tratamiento Farmacológico de COVID-19
4.
J Clin Immunol ; 40(5): 699-707, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32447592

RESUMEN

Hemophagocytic lymphohistiocytosis (HLH) is a syndrome of excessive immune system activation driven mainly by high levels of interferon gamma. The clinical presentation of HLH can have considerable overlap with other inflammatory conditions. We present a cohort of patients with therapy refractory HLH referred to our center who were found to have a simultaneous presentation of complement-mediated thrombotic microangiopathy (TMA). Twenty-three patients had therapy refractory HLH (13 primary, 4 EVB-HLH, 6 HLH without known trigger). Sixteen (69.6%) met high-risk TMA criteria. Renal failure requiring renal replacement therapy, severe hypertension, serositis, and gastrointestinal bleeding were documented only in patients with HLH who had concomitant complement-mediated TMA. Patients with HLH and without TMA required ventilator support mainly due to CNS symptoms, while those with HLH and TMA had respiratory failure predominantly associated with pulmonary hypertension, a known presentation of pulmonary TMA. Ten patients received eculizumab for complement-mediated TMA management while being treated for HLH. All patients who received the complement blocker eculizumab in addition to the interferon gamma blocker emapalumab had complete resolution of their TMA and survived. Our observations suggest co-activation of both interferon and complement pathways as a potential culprit in the evolution of thrombotic microangiopathy in patients with inflammatory disorders like refractory HLH and may offer novel therapeutic approaches for these critically ill patients. TMA should be considered in children with HLH and multi-organ failure, as an early institution of a brief course of complement blocking therapy in addition to HLH-targeted therapy may improve clinical outcomes in these patients.


Asunto(s)
Interferón gamma/metabolismo , Linfohistiocitosis Hemofagocítica/inmunología , Microangiopatías Trombóticas/inmunología , Adolescente , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Niño , Preescolar , Estudios de Cohortes , Proteínas Inactivadoras de Complemento/uso terapéutico , Proteínas del Sistema Complemento/metabolismo , Femenino , Humanos , Hipertensión Pulmonar , Lactante , Interferón gamma/antagonistas & inhibidores , Linfohistiocitosis Hemofagocítica/mortalidad , Masculino , Insuficiencia Respiratoria , Microangiopatías Trombóticas/mortalidad , Adulto Joven
5.
J Innate Immun ; 10(5-6): 455-464, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30149378

RESUMEN

Complement is a complex protein network of plasma, and an integral part of the innate immune system. Complement activation results in the rapid clearance of bacteria by immune cells, and direct bacterial killing via large pore-forming complexes. Here we review important recent discoveries in the complement field, focusing on interactions relevant for the defense against bacteria. Understanding the molecular interplay between complement and bacteria is of great importance for future therapies for infectious and inflammatory diseases. Antibodies that support complement-dependent bacterial killing are of interest for the development of alternative therapies to treat infections with antibiotic-resistant bacteria. Furthermore, a variety of novel therapeutic complement inhibitors have been developed to prevent unwanted complement activation in autoimmune inflammatory diseases. A better understanding of how such inhibitors may increase the risk of bacterial infections is essential if such therapies are to be successful.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Infecciones Bacterianas/inmunología , Proteínas del Sistema Complemento/metabolismo , Animales , Antibacterianos/uso terapéutico , Citotoxicidad Celular Dependiente de Anticuerpos , Infecciones Bacterianas/tratamiento farmacológico , Activación de Complemento , Proteínas Inactivadoras de Complemento/uso terapéutico , Resistencia a Medicamentos , Interacciones Huésped-Patógeno , Humanos , Fagocitosis
6.
Semin Hematol ; 55(3): 150-158, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30032752

RESUMEN

The dissection of the pathogenic mechanisms of the various forms of the hemolytic uremic syndrome (HUS) has paved the way for the design of specific efficacious treatments. Such mechanistic approach led to a revolution in the management of atypical HUS with the use of the first-in class C5 blocker, eculizumab. The availability of this anticomplement drug has also raised unsettled questions regarding the cost or burden and optimal duration of therapy and its use in secondary HUS. The efficacy of eculizumab in Shiga toxin producing Escherichia coli-associated HUS is not to date established and the results of ongoing prospective studies are eagerly awaited. Nevertheless, the emergence of anticomplement therapies (eculizumab and other drugs in development) has transformed our approach of HUS.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Proteínas Inactivadoras de Complemento/uso terapéutico , Síndrome Hemolítico Urémico Atípico/patología , Proteínas Inactivadoras de Complemento/farmacología , Humanos , Resultado del Tratamiento
7.
Inflammation ; 41(2): 449-463, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29198014

RESUMEN

Salidroside is neuroprotective across a wide therapeutic time-window after cerebral ischemia-reperfusion injury (IRI). Here, we investigated the role of complement in mediating effects of salidroside after cerebral IRI in rats. Rats were administrated with vehicle or salidroside 50 mg/kg, given daily for either 24 or 48 h, after middle cerebral artery occlusion (MCAO) for 2 h and reperfusion for 1 h. Levels of proteins in ischemic brain were measured by immunofluorescence and western blotting. We observed early increases in the deposition of immunoglobulin M, mannose-binding lectin 2, and annexin IV on cerebral endothelial cells, induction of the complement components C3 and C3a, by 24 h after IRI, and a later significant increase in the complement component C1q by 48 h. Salidroside prevented these changes. The neuroplasticity-related early growth response proteins Egr1, Egr2, and Egr4 and activity-regulated cytoskeleton-associated protein increased transiently in the first 6 h after IRI but then decreased below baseline by 48 h after IRI. Neither salidroside nor a C3a receptor antagonist (C3aRA) affected these proteins 24 h after IRI, but both reversed their later decreases to similar and non-additive extents. Salidroside and C3aRA increased NeuN in a non-additive manner after IRI. Our results suggest that salidroside exerts neuroprotection by reducing early activation of the lectin pathway on the cerebral endothelium and inhibiting the gradual activation of the classical pathway after cerebral IRI. This prolonged neuroprotection may depend, at least in part, on increased expression of neuroplasticity-related genes driven by reduced complement activation.


Asunto(s)
Proteínas Inactivadoras de Complemento/farmacología , Factores de Transcripción de la Respuesta de Crecimiento Precoz/metabolismo , Glucósidos/farmacología , Neuroprotección/efectos de los fármacos , Fenoles/farmacología , Daño por Reperfusión/tratamiento farmacológico , Animales , Isquemia Encefálica/tratamiento farmacológico , Complemento C3/antagonistas & inhibidores , Proteínas Inactivadoras de Complemento/uso terapéutico , Proteínas del Sistema Complemento/efectos de los fármacos , Glucósidos/uso terapéutico , Infarto de la Arteria Cerebral Media , Fenoles/uso terapéutico , Ratas , Factores de Tiempo
8.
J Am Soc Nephrol ; 29(2): 620-635, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29042454

RESUMEN

Complement-activating anti-HLA donor-specific antibodies (DSAs) are associated with impaired kidney transplant outcome; however, whether these antibodies induce a specific rejection phenotype and influence response to therapy remains undetermined. We prospectively screened 931 kidney recipients for complement-activating DSAs and used histopathology, immunostaining, and allograft gene expression to assess rejection phenotypes. Effector cells were evaluated using in vitro human cell cultures. Additionally, we assessed the effect of complement inhibition on kidney allograft rejection phenotype and the clinical response to complement inhibition in 116 independent kidney recipients with DSAs at transplant receiving rejection prophylaxis with eculizumab or standard of care (plasma exchange and intravenous Ig) at ten international centers. The histomolecular rejection phenotype associated with complement-activating DSA was characterized by complement deposition and accumulation of natural killer cells and monocytes/macrophages in capillaries and increased expression of five biologically relevant genes (CXCL11, CCL4, MS4A7, MS4A6A, and FCGR3A) indicative of endothelial activation, IFNγ response, CD16-mediated natural killer cell activation, and monocyte/macrophage activation. Compared with standard of care, eculizumab specifically abrogated this histomolecular rejection phenotype and associated with a decreased 3-month rejection incidence rate in patients with complement-activating DSAs (56%; 95% confidence interval [95% CI], 38% to 74% versus 19%; 95% CI, 8% to 35%; P=0.001) but not in those with noncomplement-activating DSAs (9%; 95% CI, 2% to 25% versus 13%; 95% CI, 2% to 40%; P=0.65). In conclusion, circulating complement-activating anti-HLA DSAs are associated with a specific histomolecular kidney allograft rejection phenotype that can be abrogated by complement inhibition.


Asunto(s)
Aloinjertos/inmunología , Anticuerpos/sangre , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Antígenos HLA/inmunología , Transcriptoma , Adulto , Anciano , Aloinjertos/metabolismo , Aloinjertos/patología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Células Cultivadas , Quimiocina CCL4/genética , Quimiocina CXCL11/genética , Proteínas Inactivadoras de Complemento/uso terapéutico , Proteínas del Sistema Complemento/metabolismo , Femenino , Rechazo de Injerto/terapia , Humanos , Inmunoglobulinas Intravenosas/uso terapéutico , Factores Inmunológicos/uso terapéutico , Riñón/patología , Trasplante de Riñón , Células Asesinas Naturales , Macrófagos , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Monocitos , Fenotipo , Intercambio Plasmático , Receptores de IgG/genética
9.
Br J Haematol ; 178(2): 181-195, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28339096

RESUMEN

Antiphospholipid antibodies are a heterogeneous group of autoantibodies that have clear associations with thrombosis and pregnancy morbidity, and which together constitute the 'antiphospholipid syndrome' (APS). However, the pathophysiology of these complications is not well understood and their heterogeneity suggests that more than one pathogenic process may be involved. Diagnosis remains a combination of laboratory analysis and clinical observation but there have been significant advances in identifying specific pathogenic features, such as domain I-specific anti-ß2-glycoprotein-I antibodies. This in turn has pointed to endothelial and complement activation as important factors in the pathogenesis of APS. Consequently, although anticoagulation remains the standard treatment for thrombotic APS and during pregnancy, the realisation that these additional pathways are involved in the pathogenesis of APS has significant implications for treatment: agents acting outside the coagulation system, such as hydroxychloroquine for pregnancy complications and sirolimus as an inhibitor of the mammalian target of rapamycin (mTOR) pathway, are now under evaluation and represent a radical change in thinking for haematologists. Conventional anticoagulation is also under challenge from new, direct acting anticoagulants. This review will provide a comprehensive overview of the evolving understanding of APS pathogenesis and how this and novel therapeutics will alter diagnosis and management.


Asunto(s)
Síndrome Antifosfolípido/etiología , Anticoagulantes/uso terapéutico , Síndrome Antifosfolípido/tratamiento farmacológico , Autoanticuerpos/inmunología , Proteínas Inactivadoras de Complemento/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Femenino , Predicción , Humanos , Hidroxicloroquina/uso terapéutico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Inmunosupresores/uso terapéutico , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Complicaciones del Embarazo/etiología , Rituximab/uso terapéutico , Sirolimus/uso terapéutico , Trombosis/tratamiento farmacológico , Trombosis/etiología , Trombosis/inmunología , beta 2 Glicoproteína I/inmunología
10.
Semin Immunol ; 28(3): 223-40, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27346521

RESUMEN

The introduction in the clinic of anti-complement agents represented a major achievement which gave to physicians a novel etiologic treatment for different human diseases. Indeed, the first anti-complement agent eculizumab has changed the treatment paradigm of paroxysmal nocturnal hemoglobinuria (PNH), dramatically impacting its severe clinical course. In addition, eculizumab is the first agent approved for atypical Hemolytic Uremic Syndrome (aHUS), a life-threatening inherited thrombotic microangiopathy. Nevertheless, such remarkable milestone in medicine has brought to the fore additional challenges for the scientific community. Indeed, the list of complement-mediated anemias is not limited to PNH and aHUS, and other human diseases can be considered for anti-complement treatment. They include other thrombotic microangiopathies, as well as some antibody-mediated hemolytic anemias. Furthermore, more than ten years of experience with eculizumab led to a better understanding of the individual steps of the complement cascade involved in the pathophysiology of different human diseases. Based on this, new unmet clinical needs are emerging; a number of different strategies are currently under development to improve current anti-complement treatment, trying to address these specific clinical needs. They include: (i) alternative anti-C5 agents, which may improve the heaviness of eculizumab treatment; (ii) broad-spectrum anti-C3 agents, which may improve the efficacy of anti-C5 treatment by intercepting the complement cascade upstream (i.e., preventing C3-mediated extravascular hemolysis in PNH); (iii) targeted inhibitors of selective complement activating pathways, which may prevent early pathogenic events of specific human diseases (e.g., anti-classical pathway for antibody-mediated anemias, or anti-alternative pathway for PNH and aHUS). Here we briefly summarize the status of art of current and future complement inhibition for different complement-mediated anemias, trying to identify the most promising approaches for each individual disease.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Proteínas Inactivadoras de Complemento/uso terapéutico , Proteínas del Sistema Complemento/metabolismo , Hemoglobinuria Paroxística/terapia , Síndrome Hemolítico-Urémico/terapia , Inmunoterapia/métodos , Microangiopatías Trombóticas/terapia , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Activación de Complemento , Hemoglobinuria Paroxística/inmunología , Síndrome Hemolítico-Urémico/inmunología , Humanos , Medicina de Precisión , Microangiopatías Trombóticas/tratamiento farmacológico , Microangiopatías Trombóticas/inmunología
12.
Biomed Res Int ; 2015: 363278, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25705656

RESUMEN

Autoimmune hemolytic anemia (AIHA) is a collective term for several diseases characterized by autoantibody-initiated destruction of red blood cells (RBCs). Exact subclassification is essential. We provide a review of the respective types of AIHA with emphasis on mechanisms of RBC destruction, focusing in particular on complement involvement. Complement activation plays a definitive but limited role in warm-antibody AIHA (w-AIHA), whereas primary cold agglutinin disease (CAD), secondary cold agglutinin syndrome (CAS), and paroxysmal cold hemoglobinuria (PCH) are entirely complement-dependent disorders. The details of complement involvement differ among these subtypes. The theoretical background for therapeutic complement inhibition in selected patients is very strong in CAD, CAS, and PCH but more limited in w-AIHA. The optimal target complement component for inhibition is assumed to be important and highly dependent on the type of AIHA. Complement modulation is currently not an evidence-based therapy modality in any AIHA, but a number of experimental and preclinical studies are in progress and a few clinical observations have been reported. Clinical studies of new complement inhibitors are probably not far ahead.


Asunto(s)
Anemia Hemolítica Autoinmune/patología , Autoanticuerpos/inmunología , Proteínas Inactivadoras de Complemento/uso terapéutico , Eritrocitos/inmunología , Anemia Hemolítica Autoinmune/sangre , Anemia Hemolítica Autoinmune/inmunología , Proteínas del Sistema Complemento/inmunología , Eritrocitos/patología , Hemoglobinuria Paroxística/sangre , Hemoglobinuria Paroxística/inmunología , Humanos
13.
Pediatr Blood Cancer ; 62(2): 224-228, 2015 02.
Artículo en Inglés | MEDLINE | ID: mdl-25382742

RESUMEN

BACKGROUND: Addition of anti-GD2 antibody ch14.18 to the treatment of neuroblastoma has improved outcomes. The most common side effect of ch14.18 is neuropathic pain, which may in part be complement-mediated. Hu14.18K322A is a humanized anti-GD2 antibody designed to diminish complement activation and induce less pain. We compare the pain outcomes in patients treated with ch14.18 and those treated with hu14.18K322A, and explore dose-dependent relationships between pain scores, opioid requirements, and complement levels in patients treated with hu14.18K322A. PROCEDURE: Opioid (morphine equivalent mg/kg) and anxiolytic requirements during course 1 (4 days) in patients treated with hu14.18K322A and ch14.18 were reviewed. Correlations between antibody dose and pain scores, opioid requirements, and complement levels were examined for patients receiving hu14.18K322A. RESULTS: Patients treated with hu14.18K322A (n = 19) had lower opioid requirements than those who received ch14.18 (n = 9). The differences in median opioid requirements (mg/kg) were statistically significant for the overall course (1.57 vs. 2.41, P = 0.019) as well as for Days 3 (0.34 vs. 0.65, P = 0.005), and 4 (0.32 vs. 0.64, P = 0.010). No difference in anxiolytic use was observed between the two groups. In the group treated with hu14.18K322A, we found a positive correlation between antibody dose administered and pain scores, but no correlation between antibody dose and opioid requirements or changes in complement levels. CONCLUSIONS: In this retrospective analysis, hu14.18K322A induced less pain than ch14.18 based on opioid requirements. Pediatr Blood Cancer 2015;62:224-228. © 2014 Wiley Periodicals, Inc.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Proteínas Inactivadoras de Complemento/uso terapéutico , Gangliósidos/antagonistas & inhibidores , Inmunoterapia/métodos , Neuralgia/tratamiento farmacológico , Neuroblastoma/terapia , Adolescente , Niño , Preescolar , Activación de Complemento/inmunología , Proteínas del Sistema Complemento/efectos de los fármacos , Femenino , Gangliósidos/inmunología , Humanos , Lactante , Masculino , Estudios Retrospectivos , Resultado del Tratamiento
15.
Biochem Pharmacol ; 88(1): 12-22, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24384116

RESUMEN

Ischemia/reperfusion injury (IRI) may occur from ischemia due to thrombotic occlusion, trauma or surgical interventions, including transplantation, with subsequent reestablishment of circulation. Time-dependent molecular and structural changes result from the deprivation of blood and oxygen in the affected tissue during ischemia. Upon restoration of blood flow a multifaceted network of plasma cascades is activated, including the complement-, coagulation-, kinin-, and fibrinolytic system, which plays a major role in the reperfusion-triggered inflammatory process. The plasma cascade systems are therefore promising therapeutic targets for attenuation of IRI. Earlier studies showed beneficial effects through inhibition of the complement system using specific complement inhibitors. However, pivotal roles in IRI are also attributed to other cascades. This raises the question, whether drugs, such as C1 esterase inhibitor, which regulate more than one cascade at a time, have a higher therapeutic potential. The present review discusses different therapeutic approaches ranging from specific complement inhibition to simultaneous inhibition of plasma cascade systems for reduction of IRI, gives an overview of the plasma cascade systems in IRI as well as highlights recent findings in this field.


Asunto(s)
Coagulación Sanguínea , Proteínas Inactivadoras de Complemento/uso terapéutico , Proteínas del Sistema Complemento/metabolismo , Daño por Reperfusión/sangre , Animales , Anticuerpos Monoclonales/uso terapéutico , Coagulación Sanguínea/efectos de los fármacos , Coagulación Sanguínea/fisiología , Ensayos Clínicos como Asunto , Proteínas Inactivadoras de Complemento/farmacología , Proteínas del Sistema Complemento/inmunología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Humanos , Cininas/antagonistas & inhibidores , Cininas/inmunología , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología , Tromboplastina/antagonistas & inhibidores , Tromboplastina/inmunología , Resultado del Tratamiento
16.
Blood Rev ; 27 Suppl 1: S1-6, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24331206

RESUMEN

Paroxysmal nocturnal haemoglobinuria (PNH) is a progressive and life-threatening disease that causes thrombosis, end organ damage and impaired quality of life. Chronic uncontrolled complement activation leads to chronic haemolysis, causing progressive morbidities and early mortality. Hence, early diagnosis is essential for improved patient management and prognosis. Eculizumab (SOLIRIS®) specifically inhibits chronic, uncontrolled complement activation and is the first-in-class, humanised, monoclonal antibody targeting C5 within the terminal complement pathway. Eculizumab is the first and only approved treatment for PNH in adults and children.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Activación de Complemento/efectos de los fármacos , Proteínas Inactivadoras de Complemento/uso terapéutico , Hemoglobinuria Paroxística/complicaciones , Hemoglobinuria Paroxística/tratamiento farmacológico , Trombosis/etiología , Animales , Proteínas del Sistema Complemento/inmunología , Hemoglobinuria Paroxística/inmunología , Hemólisis , Humanos , Terapia Molecular Dirigida
17.
Immunobiology ; 218(5): 817-27, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23182708

RESUMEN

The complement cascade is a major contributor to the innate immune response. It has now been well accepted that complement plays a critical role in hyperacute rejection and acute antibody-mediated rejection of transplanted organ. There is also increasing evidence that complement proteins contribute to the pathogenesis of organ ischemia-reperfusion injury, and even to cell-mediated rejection. Furthermore, the chemoattractants C3a and C5a and the terminal membrane attack complex that are generated by complement activation can directly or indirectly mediate tissue injury and trigger adaptive immune responses. Here, we review recent findings concerning the role of complement in graft ischemia-reperfusion injury, antibody-mediated rejection and accommodation, and cell-mediated rejection. We also discuss the current status of complement intervention therapies in clinical transplantation and describe potential new therapeutic strategies for clinical application.


Asunto(s)
Proteínas Inactivadoras de Complemento/uso terapéutico , Proteínas del Sistema Complemento/inmunología , Rechazo de Injerto/prevención & control , Trasplante de Órganos/tendencias , Daño por Reperfusión/prevención & control , Anticuerpos Monoclonales Humanizados/uso terapéutico , Activación de Complemento/efectos de los fármacos , Proteína Inhibidora del Complemento C1/uso terapéutico , Proteínas del Sistema Complemento/genética , Rechazo de Injerto/inmunología , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Inmunoglobulinas Intravenosas/uso terapéutico , Péptidos Cíclicos/uso terapéutico , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/inmunología , Daño por Reperfusión/inmunología
18.
Immunobiology ; 217(2): 127-46, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21868123

RESUMEN

Age-related macular degeneration (AMD) is the leading cause of blindness in the developed world. It is a complex multifactorial disease, and despite new advances in treatment, many patients still succumb to visual impairment. The complement pathway has been implicated in the pathogenesis of many diseases, and recently variants in several genes encoding complement pathway proteins have been associated with AMD. Complement proteins have been found in histological specimens of eyes with AMD. Altered levels of both intrinsic complement proteins and activated products have been found in the circulation of patients with AMD. Complement activation may be triggered by oxidative stress, resulting from retinal exposure to incoming light; indeed an inter-play between these two pathological processes seems to exist. Finally, complement inhibitors are currently being evaluated in clinical trials. This article reviews the role of the complement system in AMD, and the potential of complement inhibition in preventing the devastating blindness resulting from this disease.


Asunto(s)
Activación de Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Degeneración Macular/patología , Retina/patología , Factores de Edad , Ceguera/etiología , Proteínas Inactivadoras de Complemento/uso terapéutico , Proteínas del Sistema Complemento/genética , Humanos , Degeneración Macular/complicaciones , Degeneración Macular/genética , Degeneración Macular/inmunología , Estrés Oxidativo , Retina/inmunología
19.
J Immunol ; 187(9): 4913-9, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21964028

RESUMEN

Experimental evidence suggests that C inhibition and more particularly combined inhibition of C and the TLR coreceptor CD14 may be of therapeutic benefit in sepsis and other inflammatory conditions. A barrier to the testing and further development of many inhibitors is that their activity is species specific. Pig is a relevant species for experimental models of human disease, and this study undertakes a comprehensive comparison of the inhibitory efficacy of the C5 inhibitor Ornithodoros moubata C inhibitor (OmCI) in human and porcine whole blood ex vivo models of Escherichia coli-induced sepsis. The effect of OmCI on complement activity in pigs undergoing E. coli sepsis was also examined. Porcine and human serum, and whole blood anticoagulated with lepirudin, was incubated with E. coli and the effect of OmCI investigated. The ex vivo results were virtually identical in pig and human. OmCI completely ablated the activity of all three C pathways at 0.64 µM. E. coli-induced C activation and expression of CD11b (wCD11R3 in the pig), was abolished ex vivo at 0.32 µM OmCI. Combining anti-CD14 and OmCI reduced the formation of IL-8 and TNF-α more potently than the single inhibitors. OmCI also efficiently bound E. coli-induced leukotriene B(4) in pig and human plasma. In support of our ex vivo findings, in vivo the activity of all C pathways was inhibited at 0.6 mg OmCI/kg pig. In conclusion, OmCI efficiently inhibited pig and human C activation, has accompanying anti-inflammatory effects and is a promising candidate inhibitor for further in vivo studies of sepsis.


Asunto(s)
Complemento C5a/antagonistas & inhibidores , Proteínas Inactivadoras de Complemento/fisiología , Ornithodoros/inmunología , Animales , Complemento C5a/metabolismo , Proteínas Inactivadoras de Complemento/uso terapéutico , Vía Alternativa del Complemento/inmunología , Vía Clásica del Complemento/inmunología , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/prevención & control , Femenino , Humanos , Masculino , Proyectos Piloto , Proteínas y Péptidos Salivales/fisiología , Proteínas y Péptidos Salivales/uso terapéutico , Sepsis/inmunología , Sepsis/prevención & control , Porcinos
20.
J Invertebr Pathol ; 107 Suppl: S71-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21784233

RESUMEN

Serum inactivation of baculovirus vectors is a significant barrier to the development of these highly efficient vectors for therapeutic gene delivery. In this review we will describe the efforts taken to avoid complement attack by passive or active measures. Evidently good targets for baculovirus-mediated gene delivery include immunoprivileged tissues, such as eye, brain and testis. Similarly baculovirus vectors have also proven their efficacy in an ex vivo setting for tissue engineering. Active measures to inhibit complement include the use of pharmacological inhibitors of complement as well as surface engineering of the baculoviral vectors through the use of synthetic polymers, pseudotyping or display of complement inhibitors. Lessons learned from these studies will significantly increase the possibility of using baculovirus vectors for therapeutic applications.


Asunto(s)
Baculoviridae/inmunología , Activación de Complemento , Proteínas Inactivadoras de Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Terapia Genética/métodos , Nucleopoliedrovirus/inmunología , Animales , Baculoviridae/genética , Baculoviridae/metabolismo , Proteínas Inactivadoras de Complemento/uso terapéutico , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Técnicas de Transferencia de Gen , Humanos , Factores Inmunológicos , Nucleopoliedrovirus/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...