Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Clin Immunol ; 230: 108819, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34358691

RESUMO

Acquired angioedema due to C1 inhibitor deficiency (C1-INH-AAE) is a rare disease characterized by adult-onset recurrent non-urticarial angioedema with low levels of C1-INH. It is associated with lymphoproliferative disorders, and treatments are off-label with variable success. We conducted a systematic literature review to include patients with C1-INH-AAE identified in PubMed and Embase databases between January 2006 and February 2021. Clinical features of these patients were summarized, and factors associated with disease remission were explored. A total of 121 patients were included in the current study with a median age at diagnosis of 64 years and 45.5% being male. An associated disease was recorded in 94 patients (77.7%), and lymphoproliferative disorder was the most reported (59/94, 62.8%). Anti-C1-INH autoantibodies were present in 45 of 71 patients (63.4%). Factors impacting disease remissions included age (odds ratio [OR] 0.951, 95% confidence interval [CI] 0.909-0.994, p = 0.027), male (OR 0.327, 95% CI 0.124-0.866, p = 0.025), presence of monoclonal gammopathy (OR 0.133, 95% CI 0.041-0.429, p = 0.001), requirement of specific on-demand treatment (OR 0.216, 95% CI 0.066-0.709, p = 0.012) and rituximab use (OR 2.865, 95% CI 1.038-7.911, p = 0.042). A total of nine patients (7.4%) died at last follow up and most were unrelated to C1-INH-AAE. Our results imply that C1-INH-AAE is primarily associated with underlying B or plasma cell abnormalities, and clone-directed therapies could be promising options for its long-term management.


Assuntos
Angioedema/etiologia , Proteína Inibidora do Complemento C1/metabolismo , Complemento C1/antagonistas & inibidores , Idoso , Angioedema/imunologia , Angioedema/terapia , Autoanticorpos/sangue , Proteína Inibidora do Complemento C1/imunologia , Proteína Inibidora do Complemento C1/uso terapêutico , Feminino , Humanos , Transtornos Linfoproliferativos/complicações , Masculino , Pessoa de Meia-Idade , Paraproteinemias/complicações , Resultado do Tratamento
2.
Front Immunol ; 11: 2072, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32922409

RESUMO

A dysregulated immune response with hyperinflammation is observed in patients with severe coronavirus disease 2019 (COVID-19). The aim of the present study was to assess the safety and potential benefits of human recombinant C1 esterase inhibitor (conestat alfa), a complement, contact activation and kallikrein-kinin system regulator, in severe COVID-19. Patients with evidence of progressive disease after 24 h including an oxygen saturation <93% at rest in ambient air were included at the University Hospital Basel, Switzerland in April 2020. Conestat alfa was administered by intravenous injections of 8400 IU followed by 3 additional doses of 4200 IU in 12-h intervals. Five patients (age range, 53-85 years; one woman) with severe COVID-19 pneumonia (11-39% lung involvement on computed tomography scan of the chest) were treated a median of 1 day (range 1-7 days) after admission. Treatment was well-tolerated. Immediate defervescence occurred, and inflammatory markers and oxygen supplementation decreased or stabilized in 4 patients (e.g., median C-reactive protein 203 (range 31-235) mg/L before vs. 32 (12-72) mg/L on day 5). Only one patient required mechanical ventilation. All patients recovered. C1INH concentrations were elevated before conestat alfa treatment. Levels of complement activation products declined after treatment. Viral loads in nasopharyngeal swabs declined in 4 patients. In this uncontrolled case series, targeting multiple inflammatory cascades by conestat alfa was safe and associated with clinical improvements in the majority of severe COVID-19 patients. Controlled clinical trials are needed to assess its safety and efficacy in preventing disease progression.


Assuntos
Betacoronavirus/efeitos dos fármacos , Proteína Inibidora do Complemento C1/uso terapêutico , Complemento C1/antagonistas & inibidores , Infecções por Coronavirus/tratamento farmacológico , Síndrome da Liberação de Citocina/tratamento farmacológico , Sistema Calicreína-Cinina/efeitos dos fármacos , Pneumonia Viral/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , COVID-19 , Proteína Inibidora do Complemento C1/análise , Fator XIa/antagonistas & inibidores , Feminino , Humanos , Calicreínas/antagonistas & inibidores , Masculino , Pessoa de Meia-Idade , Pandemias , Proteínas Recombinantes/uso terapêutico , SARS-CoV-2 , Carga Viral/efeitos dos fármacos
3.
Immunohorizons ; 3(11): 519-530, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31690560

RESUMO

Terminal complement pathway inhibition at the level of C5 alleviates symptoms of several diseases associated with complement overactivation. However, C5 blockade is associated with an increased risk of invasive meningococcal disease despite immunization. Targeting specific complement pathways proximal to C5 provides the theoretical advantage of leaving the other pathways (including the terminal pathway) intact for immune surveillance. We aimed to address the risk of Neisseria meningitidis and Streptococcus pneumoniae infections when inhibiting the classical pathway (CP) using a specific C1s inhibitor (TNT005). Addition of TNT005 to 20% normal human serum that contained anti-meningococcal capsular Ab decreased C4 deposition 8-fold and abrogated killing of N. meningitidis, despite leaving C3 deposition intact. TNT005 impaired killing of N. meningitidis in 78% nonimmune human plasma and 78% whole blood but permitted killing in both when specific anti-capsular Ab was added. Simultaneously inhibiting both the CP and alternative pathway (AP) blocked killing of Ab-coated N. meningitidis in whole blood. Blocking the AP alone abrogated C3 deposition, whereas TNT005 only partially inhibited (∼40% decrease) C3 deposition on S. pneumoniae coated with anti-capsular Ab. Blocking either the CP or AP alone did not impair killing of pneumococci in whole blood containing specific Ab (<10% survival at 3 h); however, blocking both pathways resulted in ∼35% bacterial survival. These data suggest that killing of N. meningitidis or S. pneumoniae in whole blood containing specific anti-capsular Abs is unimpeded by TNT005. Meningococcal and pneumococcal capsular conjugate vaccines may mitigate risk of these infections in patients receiving C1s inhibitors.


Assuntos
Anticorpos Antibacterianos/imunologia , Cápsulas Bacterianas/imunologia , Complemento C1/antagonistas & inibidores , Neisseria meningitidis/imunologia , Streptococcus pneumoniae/imunologia , Anticorpos Monoclonais/imunologia , Proteínas de Bactérias/imunologia , Complemento C4/imunologia , Via Clássica do Complemento/efeitos dos fármacos , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia
4.
Brain Nerve ; 71(6): 573-580, 2019 Jun.
Artigo em Japonês | MEDLINE | ID: mdl-31171754

RESUMO

Neuromyelitis optica (NMO) is clinically characterized by severe optic neuritis and long myelitis; and is associated with aquaporin-4 immunoglobulin-G (AQP4-IgG), an autoantibody directed against aquaporin-4 (AQP4), which is an astrocytic water channel protein. Until recent years, the treatment of NMO has mainly focused on the suppression of lymphocytes and depletion of autoantibodies. However, several studies on the pathogehesis of the disease in human pathology, cultured cells, and animal models have revealed that astrocyte injury in NMO is mainly caused by the complement-dependent cytotoxicity following AQP4-IgG binding to AQP4. Moreover, complement-targeting therapy has recently been translated into practical applications in several hematological disorders. and similary, also in the cases of NMO. In this article, we review the relevance of the complement system in the pathogenesis of NMO. Additionally, we review the current status and prospects of the complement-targeting therapy for NMO, including the clinical trials of eculizumab and C1 inhibitor for NMO, and the experimental studies on C1 inhibition by monoclonal antibodies.


Assuntos
Aquaporina 4/imunologia , Inativadores do Complemento/uso terapêutico , Proteínas do Sistema Complemento , Neuromielite Óptica/tratamento farmacológico , Neuromielite Óptica/patologia , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Autoanticorpos , Ensaios Clínicos como Assunto , Complemento C1/antagonistas & inibidores , Humanos , Imunoglobulina G
5.
Biomolecules ; 8(2)2018 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-29735903

RESUMO

When excessively activated, C1 is insufficiently regulated, which results in tissue damage. Such tissue damage causes the complement system to become further activated to remove the resulting tissue damage, and a vicious cycle of activation/tissue damage occurs. Current Food and Drug Administration approved treatments include supplemental recombinant C1 inhibitor, but these are extremely costly and a more economical solution is desired. In our work, we have utilized an existing data set of 136 compounds that have been previously tested for activity against C1. Using these compounds and the activity data, we have created models using principal component analysis, genetic algorithm, and support vector machine approaches to characterize activity. The models were then utilized to virtually screen the 72 million compound PubChem repository. This first round of virtual high-throughput screening identified many economical and promising inhibitor candidates, a subset of which was tested to validate their biological activity. These results were used to retrain the models and rescreen PubChem in a second round vHTS. Hit rates for the first round vHTS were 57%, while hit rates for the second round vHTS were 50%. Additional structure⁻property analysis was performed on the active and inactive compounds to identify interesting scaffolds for further investigation.


Assuntos
Complemento C1/metabolismo , Descoberta de Drogas/métodos , Ensaios de Triagem em Larga Escala/métodos , Aprendizado de Máquina , Bibliotecas de Moléculas Pequenas/química , Complemento C1/antagonistas & inibidores , Bases de Dados de Compostos Químicos , Descoberta de Drogas/economia , Ensaios de Triagem em Larga Escala/economia , Humanos , Relação Quantitativa Estrutura-Atividade , Bibliotecas de Moléculas Pequenas/farmacologia
6.
J Immunol ; 199(11): 3883-3891, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29061764

RESUMO

Complement is crucial to the immune response, but dysregulation of the system causes inflammatory disease. Complement is activated by three pathways: classical, lectin, and alternative. The classical and lectin pathways are initiated by the C1r/C1s (classical) and MASP-1/MASP-2 (lectin) proteases. Given the role of complement in disease, there is a requirement for inhibitors to control the initiating proteases. In this article, we show that a novel inhibitor, gigastasin, from the giant Amazon leech, potently inhibits C1s and MASP-2, whereas it is also a good inhibitor of MASP-1. Gigastasin is a poor inhibitor of C1r. The inhibitor blocks the active sites of C1s and MASP-2, as well as the anion-binding exosites of the enzymes via sulfotyrosine residues. Complement deposition assays revealed that gigastasin is an effective inhibitor of complement activation in vivo, especially for activation via the lectin pathway. These data suggest that the cumulative effects of inhibiting both MASP-2 and MASP-1 have a greater effect on the lectin pathway than the more potent inhibition of only C1s of the classical pathway.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Complemento C1/antagonistas & inibidores , Inativadores do Complemento/química , Via Clássica do Complemento/efeitos dos fármacos , Lectina de Ligação a Manose da Via do Complemento/efeitos dos fármacos , Sanguessugas/química , Serina Proteases Associadas a Proteína de Ligação a Manose/antagonistas & inibidores , Peptídeos/química , Inibidores de Serina Proteinase/química , Animais , Domínio Catalítico/efeitos dos fármacos , Células Cultivadas , Inativadores do Complemento/farmacologia , Endotélio Vascular/efeitos dos fármacos , Humanos , Peptídeos/farmacologia , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Inibidores de Serina Proteinase/farmacologia
7.
PLoS One ; 12(8): e0183701, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28832655

RESUMO

The complement system is an essential component of innate immunity and plays a major role in the pathogenesis of ischemia-reperfusion injury (IRI). In this study, we investigated the impact of human C1-inhibitor (C1INH) on the early inflammatory response to IRI and the subsequent progression to fibrosis in mice. We evaluated structural damage, renal function, acute inflammatory response, progression to fibrosis and overall survival at 90-days post-injury. Animals receiving C1INH prior to reperfusion had a significant improvement in survival rate along with superior renal function when compared to vehicle (PBS) treated counterparts. Pre-treatment with C1INH also prevented acute IL-6, CXCL1 and MCP-1 up-regulation, C5a release, C3b deposition and infiltration by neutrophils and macrophages into renal tissue. This anti-inflammatory effect correlated with a significant reduction in the expression of markers of fibrosis alpha smooth muscle actin, desmin and picrosirius red at 30 and 90 days post-IRI and reduced renal levels of TGF-ß1 when compared to untreated controls. Our findings indicate that intravenous delivery of C1INH prior to ischemic injury protects kidneys from inflammatory injury and subsequent progression to fibrosis. We conclude that early complement blockade in the context of IRI constitutes an effective strategy in the prevention of fibrosis after ischemic acute kidney injury.


Assuntos
Injúria Renal Aguda/prevenção & controle , Complemento C1/antagonistas & inibidores , Inativadores do Complemento/farmacologia , Nefropatias/patologia , Traumatismo por Reperfusão/complicações , Injúria Renal Aguda/etiologia , Animais , Complemento C1/fisiologia , Modelos Animais de Doenças , Progressão da Doença , Fibrose , Masculino , Camundongos , Camundongos Endogâmicos C57BL
8.
Nihon Jibiinkoka Gakkai Kaiho ; 120(3): 217-23, 2017 03.
Artigo em Japonês | MEDLINE | ID: mdl-30010306

RESUMO

Angioedema is characterized by rapid and severe swelling of the subcutaneous and submucosal tissues. Angioedema involving the upper airway can lead to life-threatening airway obstruction, and needs prompt diagnosis and treatment. Herein, we report a case of acute angioedema which was suspected as having been caused by estrogen imbalance. A 32-year-old woman who was taking a fertility drug for infertility treatment, presented with sudden swelling of the face and neck region and breathing difficulty. Her symptoms continued to progress despite antibiotic and corticosteroid administration. We suspected hereditary angioedema (HAE), and administered a C1-inactivator, which led to immediate and dramatic resolution of the symptoms. Since the C4 and C1-inhibitor levels were normal, the possibility of HAE type III was considered. However, another possibility was that her complicated hormonal condition, including oral intake of a fertility drug, menstruation, and mental stress may have led to estrogen imbalance causing angioedema. Currently, a variety of hormone therapies is widely used ; therefore, caution is needed against the development of estrogen-dependent angioedema.


Assuntos
Angioedema/tratamento farmacológico , Complemento C1/antagonistas & inibidores , Doença Aguda , Adulto , Angioedema/diagnóstico por imagem , Feminino , Humanos , Tomografia Computadorizada por Raios X , Resultado do Tratamento
9.
Transfusion ; 56(8): 2133-45, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27282513

RESUMO

BACKGROUND: Acute hemolytic transfusion reactions have a broad clinical presentation from mild and transitory signs and symptoms to shock, disseminated intravascular coagulation, renal failure, and death. We have recently developed a rat model of acute intravascular hemolysis showing that the classical complement pathway mediates antibody-dependent hemolysis. The objective of this study was to evaluate the role of the classical pathway inhibitor peptide inhibitor of complement C1 (PIC1) in this animal model. STUDY DESIGN AND METHODS: Male Wistar rats received a 15% transfusion of human red blood cells (RBCs) and blood was isolated from the animals up to 120 minutes. Animals received PIC1 either 2 minutes before or 0.5 minutes after transfusion. Sham-, vehicle-, and cobra venom factor (CVF)-treated animals were used as control groups with a subset of rats also receiving an equivalent dose of intravenous immunoglobulin (IVIG) before transfusion. Blood was analyzed for transfused RBC survival by flow cytometry and free hemoglobin (Hb) in isolated plasma by spectrophotometry. RESULTS: Vehicle-treated rats showed decreased human RBC survival and increased free Hb as expected. Rats receiving PIC1 before transfusion showed increased human RBC survival and decreased Hb similar to CVF-treated rats. Notably, rats receiving PIC1 after initiation of transfusion showed similar decreases in hemolysis as animals receiving PIC1 before transfusion. Compared to IVIG and saline controls, PIC1-treated animals demonstrated decreased hemolysis and protection from acute kidney injury. CONCLUSIONS: These results demonstrate that PIC1 has efficacy in an animal model of acute intravascular hemolysis in both prevention and rescue scenarios.


Assuntos
Complemento C1/antagonistas & inibidores , Hemólise/efeitos dos fármacos , Peptídeos/farmacologia , Animais , Incompatibilidade de Grupos Sanguíneos/tratamento farmacológico , Contagem de Eritrócitos , Eritrócitos/citologia , Eritrócitos/efeitos dos fármacos , Hemoglobinas/metabolismo , Humanos , Masculino , Peptídeos/uso terapêutico , Ratos , Ratos Wistar , Reação Transfusional/tratamento farmacológico
10.
Sci Rep ; 6: 19300, 2016 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-26758086

RESUMO

Blood-feeding insects inject potent salivary components including complement inhibitors into their host's skin to acquire a blood meal. Sand fly saliva was shown to inhibit the classical pathway of complement; however, the molecular identity of the inhibitor remains unknown. Here, we identified SALO as the classical pathway complement inhibitor. SALO, an 11 kDa protein, has no homology to proteins of any other organism apart from New World sand flies. rSALO anti-complement activity has the same chromatographic properties as the Lu. longipalpis salivary gland homogenate (SGH)counterparts and anti-rSALO antibodies blocked the classical pathway complement activity of rSALO and SGH. Both rSALO and SGH inhibited C4b deposition and cleavage of C4. rSALO, however, did not inhibit the protease activity of C1s nor the enzymatic activity of factor Xa, uPA, thrombin, kallikrein, trypsin and plasmin. Importantly, rSALO did not inhibit the alternative or the lectin pathway of complement. In conclusion our data shows that SALO is a specific classical pathway complement inhibitor present in the saliva of Lu. longipalpis. Importantly, due to its small size and specificity, SALO may offer a therapeutic alternative for complement classical pathway-mediated pathogenic effects in human diseases.


Assuntos
Inativadores do Complemento/farmacologia , Via Clássica do Complemento/efeitos dos fármacos , Proteínas de Insetos/farmacologia , Psychodidae/imunologia , Psychodidae/metabolismo , Saliva/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Ativação do Complemento/efeitos dos fármacos , Complemento C1/antagonistas & inibidores , Complemento C1/imunologia , Complemento C1/metabolismo , Complemento C4/antagonistas & inibidores , Complemento C4/imunologia , Complemento C4/metabolismo , Humanos , Proteínas Recombinantes/farmacologia
11.
PLoS One ; 9(9): e106824, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25191939

RESUMO

Neuromyelitis optica (NMO) is an autoimmune demyelinating disease of the central nervous system in which binding of anti-aquaporin-4 (AQP4) autoantibodies (NMO-IgG) to astrocytes causes complement-dependent cytotoxicity (CDC) and inflammation resulting in oligodendrocyte and neuronal injury. There is compelling evidence for a central role of complement in NMO pathogenesis. Here, we evaluated the potential of C1-esterase inhibitor (C1-inh) for complement-targeted therapy of NMO. C1-inh is an anti-inflammatory plasma protein with serine protease inhibition activity that has a broad range of biological activities on the contact (kallikrein), coagulation, fibrinolytic and complement systems. C1-inh is approved for therapy of hereditary angioedema (HAE) and has been studied in a small safety trial in acute NMO relapses (NCT 01759602). In vitro assays of NMO-IgG-dependent CDC showed C1-inh inhibition of human and rat complement, but with predicted minimal complement inhibition activity at a dose of 2000 units in humans. Inhibition of complement by C1-inh was potentiated by ∼10-fold by polysulfated macromolecules including heparin and dextran sulfate. In rats, intravenous C1-inh at a dose 30-fold greater than that approved to treat HAE inhibited serum complement activity by <5%, even when supplemented with heparin. Also, high-dose C1-inh did not reduce pathology in a rat model of NMO produced by intracerebral injection of NMO-IgG. Therefore, although C1r and C1s are targets of C1-inh, our in vitro data with human serum and in vivo data in rats suggest that the complement inhibition activity of C1-inh in serum is too low to confer clinical benefit in NMO.


Assuntos
Aquaporina 4/imunologia , Astrócitos/imunologia , Proteína Inibidora do Complemento C1/administração & dosagem , Complemento C1/antagonistas & inibidores , Neuromielite Óptica/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Autoanticorpos/sangue , Células CHO , Proteína Inibidora do Complemento C1/farmacologia , Cricetulus , Sulfato de Dextrana/farmacologia , Modelos Animais de Doenças , Sinergismo Farmacológico , Heparina/farmacologia , Humanos , Imunoglobulina G/efeitos adversos , Imunoglobulina G/imunologia , Técnicas In Vitro , Terapia de Alvo Molecular , Neuromielite Óptica/sangue , Neuromielite Óptica/patologia , Ratos , Ratos Endogâmicos Lew
12.
Chem Immunol Allergy ; 100: 140-7, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24925394

RESUMO

Diseases which have been demonstrated to be caused by increased plasma levels of bradykinin all have angioedema as the common major clinical manifestation. Angioedema due to therapy with angiotensin-converting enzyme (ACE) inhibitors is caused by suppressed bradykinin degradation so that it accumulates. This occurs because ACE metabolizes bradykinin by removal of Phe-Arg from the C-terminus, which inactivates it. By contrast, angioedema due to C1 inhibitor deficiency (either hereditary types I and II, or acquired) is caused by bradykinin overproduction. C1 inhibitor inhibits factor XIIa, kallikrein and activity associated with the prekallikrein-HK (high-molecular-weight kininogen) complex. In its absence, uncontrolled activation of the plasma bradykinin cascade is seen once there has been an initiating stimulus. C4 levels are low in all types of C1 inhibitor deficiency due to the instability of C1 (C1r, in particular) such that some activated C1 always circulates and depletes C4. In the hereditary disorder, formation of factor XIIf (factor XII fragment) during attacks of swelling causes C4 levels to drop toward zero, and C2 levels decline. A kinin-like molecule, once thought to be a cleavage product derived from C2 that contributes to the increased vascular permeability seen in hereditary angioedema (HAE), is now thought to be an artifact, i.e. no such molecule is demonstrable. The acquired C1 inhibitor deficiency is associated with clonal disorders of B cell hyperreactivity, including lymphoma and monoclonal gammopathy. Most cases have an IgG autoantibody to C1 inhibitor which inactivates it so that the presentation is strikingly similar to type I HAE. New therapies for types I and II HAE include C1 inhibitor replacement therapy, ecallantide, a kallikrein antagonist, and icatibant, a B2 receptor antagonist. A newly described type III HAE has normal C1 inhibitor, although it is thought to be mediated by bradykinin, as is an antihistamine-resistant subpopulation of patients with 'idiopathic' angioedema. The mechanism(s) for the formation of bradykinin in these disorders is unknown.


Assuntos
Angioedema/etiologia , Bradicinina/metabolismo , Angioedema/tratamento farmacológico , Angioedema/imunologia , Angioedemas Hereditários/tratamento farmacológico , Angioedemas Hereditários/etiologia , Angioedemas Hereditários/imunologia , Inibidores da Enzima Conversora de Angiotensina/efeitos adversos , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Anti-Inflamatórios não Esteroides/uso terapêutico , Antifibrinolíticos/uso terapêutico , Bradicinina/análogos & derivados , Bradicinina/sangue , Complemento C1/antagonistas & inibidores , Complemento C1/genética , Complemento C1/metabolismo , Fator XII/metabolismo , Humanos , Calicreínas/antagonistas & inibidores , Calicreínas/metabolismo
13.
BMJ Case Rep ; 20142014 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-24850548

RESUMO

Hereditary angioedema (HAE) is a rare inherited disorder, characterised by recurrent oedema attacks in various regions of the body. In HAE, mutations in the C1 esterase inhibitor (C1-INH) gene result in decreased C1-INH concentrations (type I HAE) or functionally deficient C1-INH (type II HAE), leading to inappropriate activation of the kallikrein-kinin system and release of vasoactive mediators. Treatment of HAE aims to manage acute attacks (using replacement C1-INH or bradykinin B2 receptor antagonist) or prevent attacks through prophylaxis (using C1-INH or attenuated androgens). We present a case of a 67-year-old man with HAE who suffered a high number of breakthrough HAE attacks while undergoing long-term prophylaxis with attenuated androgens. Androgen therapy was safely discontinued and routine prevention therapy with C1-INH (1000 U) introduced as part of an individualised management approach, in line with published clinical trial data, which improved patient outcomes in terms of HAE attack frequency and severity.


Assuntos
Angioedemas Hereditários/prevenção & controle , Proteína Inibidora do Complemento C1/uso terapêutico , Complemento C1/antagonistas & inibidores , Inativadores do Complemento/uso terapêutico , Idoso , Humanos , Masculino
14.
BMJ Case Rep ; 20132013 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-24096073

RESUMO

ACE-inhibitor is an antihypertensive drug which is increasingly used to treat a wide range of medical conditions. A known adverse reaction is angio-oedema of the head and neck, which can become fatal when the upper airway is involved, causing asphyxia. We present a Caucasian man, who developed severe angio-oedema of the tongue and floor of the mouth. He was successfully treated with complement C1-concentrate causing the swelling to regress within 20 min. This treatment option can be an effective alternative to bradykinin antagonists, which might not be available in the emergency room, or more invasive measures like intubation or emergency airway puncture.


Assuntos
Angioedema/induzido quimicamente , Angioedema/tratamento farmacológico , Inibidores da Enzima Conversora de Angiotensina/efeitos adversos , Anti-Inflamatórios não Esteroides/uso terapêutico , Bradicinina/análogos & derivados , Complemento C1/antagonistas & inibidores , Angioedema/diagnóstico , Bradicinina/uso terapêutico , Diagnóstico Diferencial , Humanos , Masculino , Pessoa de Meia-Idade , Palato Mole , Língua
15.
Biochemistry (Mosc) ; 78(6): 667-73, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23980893

RESUMO

Fc fragments (hFc) of human myeloma IgG2 proteins LOM and SIN having core hinge (Cys-Cys-Val-Glu-Cys-Pro-Pro-Cys) were first obtained by a modified proteolytic procedure. The thermostability of CH2 domains inside of standard Fc, hFc fragments, and intact IgG2 LOM and SIN was studied by fluorescence spectroscopy. It was found that CH2 domains of intact IgG2 are destabilized. The destabilization is accompanied by reduced ability of IgG2 to inhibit the activation of complement system by classical pathway. This could be due to the decrease in the affinity of CH2 domains to factor C1q.


Assuntos
Complemento C1/metabolismo , Imunoglobulina G/metabolismo , Sequência de Aminoácidos , Animais , Complemento C1/antagonistas & inibidores , Humanos , Concentração de Íons de Hidrogênio , Regiões Constantes de Imunoglobulina/química , Regiões Constantes de Imunoglobulina/imunologia , Regiões Constantes de Imunoglobulina/metabolismo , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/imunologia , Fragmentos Fc das Imunoglobulinas/metabolismo , Imunoglobulina G/química , Imunoglobulina G/imunologia , Ligação Proteica , Estabilidade Proteica , Coelhos , Espectrometria de Fluorescência , Temperatura , Termodinâmica
16.
J Eur Acad Dermatol Venereol ; 27(2): 133-41, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22568407

RESUMO

Hereditary angio-oedema (HAE) is characterized by recurrent, localized, non-pitting, non-pruritic, non-urticarial oedema. Nearly all patients experience skin swelling as a feature of HAE. There may be painful abdominal attacks, accompanied by nausea and vomiting. The disease is life-threatening should laryngeal oedema occur. HAE results from a deficiency or dysfunction of C1 inhibitor, a plasma protein with an important role in regulating the contact, complement and fibrinolytic systems. Effective management of HAE should include a plan for treatment of attacks, as well as routine and preprocedure prevention. Acute and prophylactic therapy with C1 inhibitor therapy for correcting the underlying deficiency in HAE is a valuable option.


Assuntos
Angioedemas Hereditários/diagnóstico , Angioedemas Hereditários/tratamento farmacológico , Complemento C1/antagonistas & inibidores , Angioedemas Hereditários/genética , Angioedemas Hereditários/prevenção & controle , Proteínas Inativadoras do Complemento 1/uso terapêutico , Humanos , Mutação
17.
PLoS One ; 7(10): e47296, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23077587

RESUMO

Oversulfated chondroitin sulfate (OSCS) has become the subject of multidisciplinary investigation as a non-traditional contaminant in the heparin therapeutic preparations that were linked to severe adverse events. In this study, it was found that OSCS inhibited complement fixation on bacteria and bacterial lysis mediated by the complement classical pathway. The inhibition of complement by OSCS is not due to interference with antibody/antigen interaction or due to consumption of C3 associated with FXII-dependent contact system activation. However, OSCS complement inhibition is dependent on C1 inhibitor (C1inh) since the depletion of C1inh from either normal or FXII-deficient complement plasma prevents OSCS inhibition of complement activity. Surface plasmon resonance measurements revealed that immobilized C1inhibitor bound greater than 5-fold more C1s in the presence of OSCS than in presence of heparin. Although heparin can also inhibit complement, OSCS and OSCS contaminated heparin are more potent inhibitors of complement. Furthermore, polysulfated glycosaminoglycan (PSGAG), an anti-inflammatory veterinary medicine with a similar structure to OSCS, also inhibited complement in the plasma of dogs and farm animals. This study provides a new insight that in addition to the FXII-dependent activation of contact system, oversulfated and polysulfated chondroitin-sulfate can inhibit complement activity by potentiating the classical complement pathway regulator C1inh. This effect on C1inh may play a role in inhibiting inflammation as well as impacting bacterial clearance.


Assuntos
Sulfatos de Condroitina/farmacologia , Proteína Inibidora do Complemento C1/metabolismo , Heparina/química , Animais , Anticoagulantes/efeitos adversos , Anticoagulantes/química , Coagulação Sanguínea/efeitos dos fármacos , Sulfatos de Condroitina/química , Complemento C1/antagonistas & inibidores , Complemento C1/química , Proteína Inibidora do Complemento C1/química , Cães , Contaminação de Medicamentos , Fator XII/química , Fator XII/efeitos dos fármacos , Heparina/efeitos adversos , Plasma/química , Plasma/metabolismo
18.
Biomed Khim ; 58(2): 176-88, 2012.
Artigo em Russo | MEDLINE | ID: mdl-22724357

RESUMO

Serpins (SERine Protease INhibitors)--is large and diverse group of proteins with similar structures, which can inhibit both serine and cysteine proteases by an irreversible suicide mechanism. A novel serpin from hepatopancreas of Red King Crab (Paralithosed camtschaticus) was obtained and was studied its effect on the process of human blood plasma clotting. The investigated serpin shows a noticeable anticoagulative activity, which increases dramatically in the combined action with heparine. Though the inhibitor has almost no effect on thrombin, it inhibits C1s (C1-esterase). We studied the action of the serpin from P. camtschaticus on C1s via its competitive inhibition by C1 inhibitor and the novel enzyme. The calculated inhibition constant of the serpin from P. camtschaticus towards C1s is 2.02 +/- 0.71 M. Unlike C1 inhibitor, the novel serpin from P. camtschaticus doesn't suppress fibrinolysis and at the same time prevents blood clotting. These features may be of interest for medical purposes.


Assuntos
Anomuros/química , Anticoagulantes/farmacologia , Inibidores de Serina Proteinase/farmacologia , Serpinas/farmacologia , Animais , Ligação Competitiva , Coagulação Sanguínea/efeitos dos fármacos , Complemento C1/antagonistas & inibidores , Complemento C1/metabolismo , Fibrinolisina/antagonistas & inibidores , Heparina/farmacologia , Hepatopâncreas/química , Humanos , Serpinas/metabolismo , Trombina/antagonistas & inibidores
19.
J Burn Care Res ; 33(4): 544-51, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22447201

RESUMO

In a previous study, the authors found persistent presence of acute inflammation markers such as C-reactive protein and complement factors locally in burn wounds. This persistence of acute inflammation may not only delay local burn wound healing but also have a systemic effect, for instance on the heart. Here, the effects of C1 esterase inhibitor (C1inh), an inhibitor of complement activation, on burn wound progression and the heart were analyzed in rats. Dorsal full-thickness burn wounds (2 × 4 cm) were induced on female Wistar rats (n = 14). The rats were divided into two groups (n = 7): a control group (just burns) and a C1inh group. C1inh was administered daily intravenously for 14 days. The burn wound, healthy skin from the hind leg (internal control), and the heart were then fixed in formalin. Tissues were analyzed for granulation tissue formation, reepithelialization, amount and type of infiltrating inflammatory cells (granulocytes and macrophages), and inflammatory markers (complement factors C3 and C4). C1inh treatment significantly reduced the amount of granulation tissue and significantly increased reepithelialization. C1inh also significantly reduced macrophage infiltration. Burns induced infiltration of macrophages into the ventricles of the heart and remarkably also into the atria of the heart. This effect could be counteracted by C1inh. These data show that systemic treatment with C1inh acts at different levels resulting in improved healing locally in burn wounds and systemically reduced inflammation in the heart. Therefore, C1inh might be a possible therapeutic intervention for burn wound patients.


Assuntos
Queimaduras/tratamento farmacológico , Complemento C1/antagonistas & inibidores , Inativadores do Complemento/farmacologia , Miocardite/prevenção & controle , Cicatrização/efeitos dos fármacos , Análise de Variância , Animais , Queimaduras/mortalidade , Queimaduras/patologia , Distribuição de Qui-Quadrado , Complemento C1/farmacologia , Modelos Animais de Doenças , Esquema de Medicação , Feminino , Imuno-Histoquímica , Mediadores da Inflamação/sangue , Infusões Intravenosas , Escala de Gravidade do Ferimento , Miocardite/sangue , Distribuição Aleatória , Ratos , Ratos Wistar , Valores de Referência , Taxa de Sobrevida , Cicatrização/fisiologia
20.
Immunobiology ; 217(2): 251-5, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21852020

RESUMO

This study assessed variation in plasma levels of the complement regulatorC1 inhibitor (C1inh) in patients with age related macular degeneration (AMD) and controls. Plasma from391 AMD cases and 370 controls was assayed by rate nephelometry to determine C1inh protein levels. Protein levels were analysed for relationships with age, gender, smoking, AMD disease status and genetic variation in the SERPING1 gene, which encodes C1inh, using a multivariate analysis. t-Tests show a significant difference in C1inh levels in AMD cases compared with controls (p=2.340E-6), smokers compared to non-smokers (p=1.022E-4) and females compared to males (p=1.661E-7). Multivariate analysis shows that after accounting for gender and smoking AMD status remained significant. Age was included in the model but was not significant. Including genetic variation in the model shows that one significant SNP (rs2649663) 5' of the SERPING1 gene is associated with C1inh levels though this SNP is not associated with AMD. This suggests that genetic variation in the promoter region of the SERPING1 gene may influence expression of the gene.


Assuntos
Proteínas Inativadoras do Complemento 1/genética , Proteína Inibidora do Complemento C1/análise , Proteína Inibidora do Complemento C1/imunologia , Degeneração Macular/genética , Idoso , Idoso de 80 Anos ou mais , Complemento C1/antagonistas & inibidores , Feminino , Predisposição Genética para Doença , Variação Genética , Genótipo , Humanos , Degeneração Macular/sangue , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas/genética , Fatores Sexuais , Fumar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...