Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 328
Filtrar
1.
Cells ; 10(12)2021 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-34944083

RESUMO

Considering the fact that many retinal diseases are yet to be cured, the pathomechanisms of these multifactorial diseases need to be investigated in more detail. Among others, oxidative stress and hypoxia are pathomechanisms that take place in retinal diseases, such as glaucoma, age-related macular degeneration, or diabetic retinopathy. In consideration of these diseases, it is also evidenced that the immune system, including the complement system and its activation, plays an important role. Suitable models to investigate neuroretinal diseases are organ cultures of porcine retina. Based on an established model, the role of the complement system was studied after the induction of oxidative stress or hypoxia. Both stressors led to a loss of retinal ganglion cells (RGCs) accompanied by apoptosis. Hypoxia activated the complement system as noted by higher C3+ and MAC+ cell numbers. In this model, activation of the complement cascade occurred via the classical pathway and the number of C1q+ microglia was increased. In oxidative stressed retinas, the complement system had no consideration, but strong inflammation took place, with elevated TNF, IL6, and IL8 mRNA expression levels. Together, this study shows that hypoxia and oxidative stress induce different mechanisms in the porcine retina inducing either the immune response or an inflammation. Our findings support the thesis that the immune system is involved in the development of retinal diseases. Furthermore, this study is evidence that both approaches seem suitable models to investigate undergoing pathomechanisms of several neuroretinal diseases.


Assuntos
Ativação do Complemento/imunologia , Via Clássica do Complemento/imunologia , Hipóxia/imunologia , Retina/imunologia , Retina/patologia , Células Ganglionares da Retina/patologia , Animais , Apoptose/efeitos dos fármacos , Cobalto/toxicidade , Ativação do Complemento/efeitos dos fármacos , Via Alternativa do Complemento/efeitos dos fármacos , Via Alternativa do Complemento/imunologia , Via Clássica do Complemento/efeitos dos fármacos , Proteínas do Sistema Complemento/metabolismo , Peróxido de Hidrogênio/toxicidade , Lectinas/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Estresse Oxidativo/efeitos dos fármacos , Células Ganglionares da Retina/efeitos dos fármacos , Neurônios Retinianos/efeitos dos fármacos , Neurônios Retinianos/patologia , Estresse Fisiológico/efeitos dos fármacos , Suínos
2.
PLoS One ; 16(9): e0257960, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34591905

RESUMO

Perinatal hypoxic ischemic encephalopathy (HIE) remains a major contributor of infant death and long-term disability worldwide. The role played by the complement system in this ischemia-reperfusion injury remains poorly understood. In order to better understand the role of complement activation and other modifiable mechanisms of injury in HIE, we tested the dual-targeting anti-inflammatory peptide, RLS-0071 in an animal model of HIE. Using the well-established HIE rat pup model we measured the effects of RLS-0071 during the acute stages of the brain injury and on long-term neurocognitive outcomes. Rat pups subject to hypoxia-ischemia insult received one of 4 interventions including normothermia, hypothermia and RLS-0071 with and without hypothermia. We measured histopathological effects, brain C1q levels and neuroimaging at day 1 and 21 after the injury. A subset of animals was followed into adolescence and evaluated for neurocognitive function. On histological evaluation, RLS-0071 showed neuronal protection in combination with hypothermia (P = 0.048) in addition to reducing C1q levels in the brain at 1hr (P = 0.01) and at 8 hr in combination with hypothermia (P = 0.005). MRI neuroimaging demonstrated that RLS-0071 in combination with hypothermia reduced lesion volume at 24 hours (P<0.05) as well as decreased T2 signal at day 21 in combination with hypothermia (P<0.01). RLS-0071 alone or in combination with hypothermia improved both short-term and long-term memory. These findings suggest that modulation by RLS-0071 can potentially decrease brain damage resulting from HIE.


Assuntos
Encéfalo/efeitos dos fármacos , Complemento C1q/antagonistas & inibidores , Inativadores do Complemento/farmacologia , Via Clássica do Complemento/efeitos dos fármacos , Hipóxia-Isquemia Encefálica/terapia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Terapia Combinada , Complemento C1q/metabolismo , Inativadores do Complemento/uso terapêutico , Modelos Animais de Doenças , Hipotermia Induzida , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Destreza Motora/efeitos dos fármacos , Destreza Motora/fisiologia , Ratos , Reconhecimento Psicológico/efeitos dos fármacos , Reconhecimento Psicológico/fisiologia , Resultado do Tratamento
3.
J Autoimmun ; 124: 102728, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34592707

RESUMO

Extremely rare reactions characterized by thrombosis and thrombocytopenia have been described in subjects that received ChAdOx1 nCoV-19 vaccination 5-16 days earlier. Although patients with vaccine-induced thrombotic thrombocytopenia (VITT) have high levels of antibodies to platelet factor 4 (PF4)-polyanion complexes, the exact mechanism of the development of thrombosis is still unknown. Here we reported serum studies as well as proteomics and genomics analyses demonstrating a massive complement activation potentially linked to the presence of anti-PF4 antibodies in a patient with severe VITT. At admission, complement activity of the classical and lectin pathways were absent (0% for both) with normal levels of the alternative pathway (73%) in association with elevated levels of the complement activation marker sC5b-9 (630 ng/mL [n.v. 139-462 ng/mL]) and anti-PF4 IgG (1.918 OD [n.v. 0.136-0.300 OD]). The immunoblotting analysis of C2 showed the complete disappearance of its normal band at 110 kDa. Intravenous immunoglobulin treatment allowed to recover complement activity of the classical pathway (91%) and lectin pathway (115%), to reduce levels of sC5b-9 (135 ng/mL) and anti-PF4 IgG (0.681 OD) and to normalize the C2 pattern at immunoblotting. Proteomics and genomics analyses in addition to serum studies showed that the absence of complement activity during VITT was not linked to alterations of the C2 gene but rather to a strong complement activation leading to C2 consumption. Our data in a single patient suggest monitoring complement parameters in other VITT patients considering also the possibility to target complement activation with specific drugs.


Assuntos
Vacinas contra COVID-19/efeitos adversos , COVID-19/prevenção & controle , Complemento C2 , Complexo de Ataque à Membrana do Sistema Complemento , Via Clássica do Complemento , Lectina de Ligação a Manose da Via do Complemento , Púrpura Trombocitopênica Trombótica , SARS-CoV-2 , Adulto , Autoanticorpos/sangue , Vacinas contra COVID-19/administração & dosagem , ChAdOx1 nCoV-19 , Complemento C2/genética , Complemento C2/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/genética , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Via Clássica do Complemento/efeitos dos fármacos , Via Clássica do Complemento/genética , Lectina de Ligação a Manose da Via do Complemento/efeitos dos fármacos , Lectina de Ligação a Manose da Via do Complemento/genética , Feminino , Humanos , Fator Plaquetário 4/sangue , Púrpura Trombocitopênica Trombótica/sangue , Púrpura Trombocitopênica Trombótica/induzido quimicamente , Púrpura Trombocitopênica Trombótica/genética
4.
Am J Physiol Renal Physiol ; 321(4): F505-F516, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34459222

RESUMO

Focal segmental glomerulosclerosis (FSGS) and minimal change disease (MCD) are common forms of idiopathic nephrotic syndrome. The causes of these diseases are incompletely understood, but the response of patients to immunosuppressive therapies suggests that their pathogenesis is at least in part immune mediated. Preclinical and clinical research indicates that activation of the classical pathway of complement contributes to glomerular injury in FSGS. Glomerular IgM deposits are also prominent in some patients, raising the possibility that IgM is a trigger of classical pathway activation. In the present study, we examined the pattern of complement activation in the glomeruli and plasma of patients with nephrotic syndrome. We also tested whether patients with FSGS and MCD have elevated levels of natural IgM reactive with epitopes on glomerular endothelial cells and cardiolipin. We found evidence of classical pathway activation in patients with idiopathic nephrotic syndrome compared with healthy control subjects. We also detected higher levels of self-reactive IgM to both targets. Based on these results, IgM and classical pathway activation may contribute to disease pathogenesis in some patients with FSGS and MCD.NEW & NOTEWORTHY IgM is detected in biopsies from some patients with nephrotic syndrome, although this has been attributed to passive trapping of the protein. We found, however, that IgM colocalizes with complement activation fragments in some glomeruli. We also found that affected patients had higher levels of IgM reactive to glomerular endothelial cell epitopes. Thus, IgM activates the complement system in the glomeruli of some patients with nephrotic syndrome and may contribute to injury.


Assuntos
Cardiolipinas/imunologia , Via Clássica do Complemento , Proteínas do Sistema Complemento/análise , Células Endoteliais/imunologia , Epitopos , Glomerulosclerose Segmentar e Focal/imunologia , Imunoglobulina M/análise , Glomérulos Renais/imunologia , Nefrose Lipoide/imunologia , Síndrome Nefrótica/imunologia , Adulto , Idoso , Especificidade de Anticorpos , Estudos de Casos e Controles , Via Clássica do Complemento/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Feminino , Glomerulosclerose Segmentar e Focal/sangue , Glomerulosclerose Segmentar e Focal/tratamento farmacológico , Glomerulosclerose Segmentar e Focal/patologia , Humanos , Imunoglobulina M/sangue , Imunossupressores/uso terapêutico , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/patologia , Masculino , Pessoa de Meia-Idade , Nefrose Lipoide/tratamento farmacológico , Nefrose Lipoide/patologia , Síndrome Nefrótica/tratamento farmacológico , Síndrome Nefrótica/patologia , Resultado do Tratamento , Adulto Jovem
5.
Front Immunol ; 12: 607641, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33936031

RESUMO

The Sez6 family consists of Sez6, Sez6L, and Sez6L2. Its members are expressed throughout the brain and have been shown to influence synapse numbers and dendritic morphology. They are also linked to various neurological and psychiatric disorders. All Sez6 family members contain 2-3 CUB domains and 5 complement control protein (CCP) domains, suggesting that they may be involved in complement regulation. We show that Sez6 family members inhibit C3b/iC3b opsonization by the classical and alternative pathways with varying degrees of efficacy. For the classical pathway, Sez6 is a strong inhibitor, Sez6L2 is a moderate inhibitor, and Sez6L is a weak inhibitor. For the alternative pathway, the complement inhibitory activity of Sez6, Sez6L, and Sez6L2 all equaled or exceeded the activity of the known complement regulator MCP. Using Sez6L2 as the representative family member, we show that it specifically accelerates the dissociation of C3 convertases. Sez6L2 also functions as a cofactor for Factor I to facilitate the cleavage of C3b; however, Sez6L2 has no cofactor activity toward C4b. In summary, the Sez6 family are novel complement regulators that inhibit C3 convertases and promote C3b degradation.


Assuntos
Convertases de Complemento C3-C5/metabolismo , Complemento C3b/imunologia , Fibrinogênio/metabolismo , Proteínas de Membrana/metabolismo , Complemento C3b/metabolismo , Proteínas Inativadoras do Complemento/genética , Proteínas Inativadoras do Complemento/metabolismo , Via Alternativa do Complemento/efeitos dos fármacos , Via Clássica do Complemento/efeitos dos fármacos , Expressão Gênica , Humanos , Imuno-Histoquímica , Proteínas de Membrana/genética , Proteínas de Membrana/farmacologia , Proteólise , Proteínas Recombinantes de Fusão
6.
Blood ; 137(4): 443-455, 2021 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-33507296

RESUMO

Blocking the terminal complement pathway with the C5 inhibitor eculizumab has revolutionized the clinical management of several complement-mediated diseases and has boosted the clinical development of new inhibitors. Data on the C3 inhibitor Compstatin and the C5 inhibitors eculizumab and Coversin reported here demonstrate that C3/C5 convertases function differently from prevailing concepts. Stoichiometric C3 inhibition failed to inhibit C5 activation and lytic activity during strong classical pathway activation, demonstrating a "C3 bypass" activation of C5. We show that, instead of C3b, surface-deposited C4b alone can also recruit and prime C5 for consecutive proteolytic activation. Surface-bound C3b and C4b possess similar affinities for C5. By demonstrating that the fluid phase convertase C3bBb is sufficient to cleave C5 as long as C5 is bound on C3b/C4b-decorated surfaces, we show that surface fixation is necessary only for the C3b/C4b opsonins that prime C5 but not for the catalytic convertase unit C3bBb. Of note, at very high C3b densities, we observed membrane attack complex formation in absence of C5-activating enzymes. This is explained by a conformational activation in which C5 adopts a C5b-like conformation when bound to densely C3b-opsonized surfaces. Stoichiometric C5 inhibitors failed to prevent conformational C5 activation, which explains the clinical phenomenon of residual C5 activity documented for different inhibitors of C5. The new insights into the mechanism of C3/C5 convertases provided here have important implications for the development and therapeutic use of complement inhibitors as well as the interpretation of former clinical and preclinical data.


Assuntos
C3 Convertase da Via Alternativa do Complemento/fisiologia , Complemento C3/antagonistas & inibidores , Complemento C4b/fisiologia , Complemento C5/antagonistas & inibidores , Inativadores do Complemento/farmacologia , Via Clássica do Complemento/efeitos dos fármacos , Modelos Imunológicos , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Membrana Celular/imunologia , Complemento C5/química , Inativadores do Complemento/uso terapêutico , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Resistência a Medicamentos , Células Endoteliais da Veia Umbilical Humana , Humanos , Modelos Moleculares , Peptídeos Cíclicos/farmacologia , Peptídeos Cíclicos/uso terapêutico , Conformação Proteica
7.
Carbohydr Polym ; 254: 117251, 2021 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-33357847

RESUMO

The research described here presents data on the effect of galactans of red algae, carrageenans (λ/µ/ν-, κ-, κ/ß-, and ι/κ-types), and agar on complement system activation in normal human serum. The experiments were based on well surfaces coated with triggering agents for binding initiating complement components -C3 and C4. The sulfated galactans inhibited C3 binding to lipopolysaccharide with direct dependence on the sulfation degree of polysaccharides. Sulfation degree was also important in carrageenans' capacity to reduce C4 binding to mannan. However, C4 binding to antibodies was considerably activated by carrageenans, especially with 3,6-anhydrogalactose. The gelling carrageenans were able to block antigen binding centers of total serum IgM and with more intensity than non-gelling. No structural characteristics mattered in ameliorating C5 cleavage by plasmin in extrinsic protease complement activation, but λ/µ/ν- and κ/ß-carrageenans almost completely inhibited C5 cleavage. Thus, galactans participated in cell surface biology by imitating surface glycans in inhibition of C3 binding and mannose binding lectin, but as to the tthe heclassical pathway these substances stimulated complement, probably due to their structure based on carrabiose.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Galactanos/química , Galactanos/farmacologia , Alga Marinha/química , Anticorpos/sangue , Configuração de Carboidratos , Sequência de Carboidratos , Carragenina/química , Carragenina/farmacologia , Via Alternativa do Complemento/efeitos dos fármacos , Via Clássica do Complemento/efeitos dos fármacos , Galactanos/sangue , Humanos , Técnicas In Vitro , Estrutura Molecular , Ressonância Magnética Nuclear Biomolecular , Polissacarídeos/química , Polissacarídeos/farmacologia , Rodófitas/química , Espectroscopia de Infravermelho com Transformada de Fourier , Sulfatos/química
8.
Adv Chronic Kidney Dis ; 27(2): 138-148, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32553246

RESUMO

Antibody-mediated rejection (AMR) is one of the leading causes of kidney allograft failure and is usually mediated by anti-human leukocyte antigen donor-specific antibodies (DSAs). Activation of classical pathway of the complement system is responsible for downstream effects of DSA and account for significant manifestations of AMR. Currently, the treatment of AMR is based on strategies to remove preformed antibodies or to prevent their production; however, these strategies are often unsuccessful. It is theoretically possible to inhibit complement activity to prevent the effect of DSA on kidney allograft function. Complement inhibitors such as eculizumab, a complement 5 monoclonal antibody, and complement 1 esterase inhibitors (C1 INHs) have been used in prevention and treatment of AMR with variable success. Eculizumab and C1 INH seem to reduce the incidence of early AMR and allow transplantation in highly sensitized kidney transplant recipients, but data on their long-term effect on kidney allograft function are limited. Several case reports described the successful use of eculizumab in the treatment of AMR, but there are no randomized controlled studies that showed efficacy. Treatment of AMR with C1 INH, in addition to standard of care, did not change short-term outcome but long-term studies are underway.


Assuntos
Anticorpos Monoclonais Humanizados , Proteína Inibidora do Complemento C1 , Via Clássica do Complemento , Rejeição de Enxerto , Antígenos HLA/imunologia , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/farmacologia , Proteína Inibidora do Complemento C1/imunologia , Proteína Inibidora do Complemento C1/farmacologia , Inativadores do Complemento/imunologia , Inativadores do Complemento/farmacologia , Via Clássica do Complemento/efeitos dos fármacos , Via Clássica do Complemento/imunologia , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Humanos , Falência Renal Crônica/cirurgia , Transplante de Rim/efeitos adversos
9.
Clin Cancer Res ; 26(13): 3481-3490, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31919135

RESUMO

PURPOSE: Application of allogeneic hematopoietic cell transplantation (allo-HCT) for patients with hematologic disorders is limited by the development of GVHD. Separation of GVHD and graft-versus-leukemia (GVL) remains a great challenge in the field. We investigated the contribution of individual pathways involved in the complement cascade in GVH and GVL responses to identify specific targets by which to separate these two processes. EXPERIMENTAL DESIGN: We used multiple preclinical murine and human-to-mouse xenograft models involving allo-HCT recipients lacking components of the alternative pathway (AP) or classical pathway (CP)/lectin pathway (LP) to dissect the role of each individual pathway in GVHD pathogenesis and the GVL effect. For translational purposes, we used the AP-specific complement inhibitor, CR2-fH, which localizes in injured target organs to allow specific blockade of complement activation at sites of inflammation. RESULTS: Complement deposition was evident in intestines of mice and patients with GVHD. In a preclinical setting, ablation of the AP, but not the CP/LP, significantly improved GVHD outcomes. Complement activation through the AP in host hematopoietic cells, and specifically dendritic cells (DC), was required for GVHD progression. AP deficiency in recipients decreased donor T-cell migration and Th1/Th2 differentiation, while increasing the generation of regulatory T cells. This was because of decreased activation and stimulatory activity of recipient DCs in GVHD target organs. Treatment with CR2-fH effectively prevented GVHD while preserving GVL activity. CONCLUSIONS: This study highlights the AP as a new therapeutic target to prevent GVHD and tumor relapse after allo-HCT. Targeting the AP by CR2-fH represents a promising therapeutic approach for GVHD treatment.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Proteínas do Sistema Complemento/imunologia , Doença Enxerto-Hospedeiro/etiologia , Doença Enxerto-Hospedeiro/prevenção & controle , Animais , Via Clássica do Complemento/efeitos dos fármacos , Via Clássica do Complemento/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Imunofenotipagem , Leucemia/complicações , Leucemia/terapia , Camundongos , Camundongos Knockout , Prognóstico , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Transplante Homólogo
10.
Clin Rev Allergy Immunol ; 58(2): 155-171, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31144209

RESUMO

Approved for the treatment of autoimmune diseases, hematological malignancies, and solid cancers, several monoclonal antibodies (mAb) make use of complement in their mechanism of action. Such an assessment is based on comprehensive investigations that used mouse models, in vitro studies, and analyses from patients at initiation (basal level to highlight deficiencies) and after treatment initiation (mAb impact on complement), which have further provided key insights into the importance of the complement activation and/or complement deficiencies in mAb activity. Accordingly, new approaches can now be developed with the final objective of increasing the clinical efficacy of mAb. These improvements include (i) the concurrent administration of fresh frozen plasma during mAb therapy; (ii) mAb modifications such as immunoglobulin G subclass switching, Fc mutation, or IgG hexamerization to improve the fixation and activation of C1q; (iii) optimization of the target recognition to induce a higher complement-dependent cytotoxicity (CDC) and/or complement-dependant cellular cytotoxicity (CDCC); and (iv) the control of soluble and cellular complement inhibitors.


Assuntos
Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Biomarcadores , Ativação do Complemento/efeitos dos fármacos , Via Alternativa do Complemento/efeitos dos fármacos , Via Alternativa do Complemento/imunologia , Via Clássica do Complemento/efeitos dos fármacos , Via Clássica do Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Humanos , Fatores Imunológicos/farmacologia , Fatores Imunológicos/uso terapêutico , Imunoterapia , Resultado do Tratamento
11.
J Immunotoxicol ; 16(1): 191-200, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31684787

RESUMO

In preclinical toxicity studies, species-foreign proteins administered to animals frequently leads to formation of anti-drug antibodies (ADA). Such antibodies may form circulating immune complexes (CIC) with the administered protein. These CIC can activate the classical complement pathway, thereby forming complement-bound CIC (cCIC); if large of amounts of CIC or cCIC is formed, the clearance mechanism may become saturated which potentially leads to vascular immune complex (IC) deposition and inflammation. Limited information is available on the effect of different treatment related procedures as well as biomarkers of IC-related vascular disease. In order to explore the effect of different dose regimens on IC formation and deposition, and identification of possible biomarkers of IC deposition and IC-related pathological changes, C57BL/6J and BALB/c mice were dosed subcutaneously twice weekly with bovine serum albumin (BSA) for 13 weeks without adjuvant. After 6 and 13 weeks, CIC and cCIC were detected in plasma; after 13 weeks, IC deposition was detected in kidney glomeruli. In particular immunohistochemistry double-staining was shown to be useful for detection of IC deposition. Increasing dosing frequency or changing BSA dose level on top of an already established CIC and cCIC response did not cause changes in IC deposition, but CIC and cCIC concentrations tended to decrease with increased dose level, and increased cCIC formation was observed after more frequent dosing. The presence of CIC in plasma was associated with glomerular IC deposits in the dose regimen study; however, the use of CIC or cCIC as potential biomarkers for IC deposition and IC-related pathological changes, needs to be explored further.


Assuntos
Complexo Antígeno-Anticorpo/análise , Glomerulonefrite/imunologia , Soroalbumina Bovina/toxicidade , Vasculite Sistêmica/imunologia , Animais , Complexo Antígeno-Anticorpo/imunologia , Biomarcadores/análise , Via Clássica do Complemento/efeitos dos fármacos , Via Clássica do Complemento/imunologia , Modelos Animais de Doenças , Estudos de Viabilidade , Feminino , Glomerulonefrite/sangue , Glomerulonefrite/induzido quimicamente , Glomerulonefrite/diagnóstico , Humanos , Imuno-Histoquímica , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/imunologia , Glomérulos Renais/patologia , Masculino , Camundongos , Soroalbumina Bovina/administração & dosagem , Soroalbumina Bovina/imunologia , Vasculite Sistêmica/sangue , Vasculite Sistêmica/induzido quimicamente , Vasculite Sistêmica/diagnóstico , Testes de Toxicidade/métodos
12.
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
13.
Biomacromolecules ; 20(10): 3809-3818, 2019 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-31461260

RESUMO

The complement system is a powerful mechanism of the innate immune defense system. Dysregulation may contribute to several diseases. Heparin is a known regulator of the complement system, but its application is limited due to its anticoagulative activity. A promising alternative is the synthetic analogue dendritic polyglycerol sulfate (dPGS). Although dPGS-mediated inhibition of the classical and alternative pathway has been roughly described previously, here we analyzed the effects of dPGS regarding the three pathways at different levels of the proteolytic cascades for the first time. Regarding the final outcome (membrane attack complex formation), IC50 values for dPGS varied between the alternative (900 nM), the classical (300 nM), and the lectin pathway (60 nM). In a backward approach, processing of proteins C5 and C3 via the respective convertase was analyzed by ELISA to narrow down dPGS targets. A dose-dependent reduction of C5a and C3a levels was detected. Further, the analysis via surface plasmon resonance revealed novel dPGS binding proteins; the pro-inflammatory anaphylatoxins C3a and C5a and the classical pathway activator C1q showed down to nanomolar binding affinities. The fully synthetic multivalent polymer dPGS seems to be a promising candidate for the further development to counteract excessive complement activation in disease.


Assuntos
Complemento C3/antagonistas & inibidores , Complemento C5/antagonistas & inibidores , Glicerol/farmacologia , Polímeros/farmacologia , Via Alternativa do Complemento/efeitos dos fármacos , Via Clássica do Complemento/efeitos dos fármacos , Lectina de Ligação a Manose da Via do Complemento/efeitos dos fármacos , Glicerol/química , Humanos , Polímeros/química , Proteólise/efeitos dos fármacos
14.
J Invest Dermatol ; 139(12): 2417-2424.e2, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31229501

RESUMO

Deposition of autoantibodies (α-BP180 and BP230) and complement along the dermal-epidermal-junction is a hallmark of bullous pemphigoid and was shown to be important for pathogenesis. Given the adverse effects of standard treatment (glucocorticoids, immunosuppressants), there is an unmet need for safe and effective therapies. In this phase 1 trial, we evaluated the safety and activity of BIVV009 (sutimlimab, previously TNT009), a targeted C1s inhibitor, in 10 subjects with active or past bullous pemphigoid (NCT02502903). Four weekly 60 mg/kg infusions of BIVV009 proved sufficient for inhibition of the classical complement pathway in all patients, as measured by CH50. C3c deposition along the dermal-epidermal junction was partially or completely abrogated in 4 of 5 patients, where it was present at baseline. BIVV009 was found to be safe and tolerable in this elderly population, with only mild to moderate adverse events reported (e.g., headache, fatigue). One serious adverse event (i.e., fatal cardiac decompensation) occurred at the end of the post-treatment observation period in an 84-year-old patient with a history of diabetes and heart failure, but was deemed unlikely to be related to the study drug. This trial provides the first results with a complement-targeting therapy in bullous pemphigoid, to our knowledge, and supports further studies on BIVV009's efficacy and safety in this population.


Assuntos
Autoantígenos/administração & dosagem , Complemento C3/metabolismo , Via Clássica do Complemento/efeitos dos fármacos , Derme/patologia , Distonina/administração & dosagem , Epiderme/patologia , Colágenos não Fibrilares/administração & dosagem , Penfigoide Bolhoso/tratamento farmacológico , Idoso , Idoso de 80 Anos ou mais , Derme/metabolismo , Epiderme/metabolismo , Feminino , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Penfigoide Bolhoso/metabolismo , Penfigoide Bolhoso/patologia , Colágeno Tipo XVII
15.
Haematologica ; 104(2): 403-416, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30262558

RESUMO

High titers of HLA antibodies are associated with platelet refractoriness, causing poor platelet increments after transfusions in a subset of patients with HLA antibodies. Currently, we do not know the biological mechanisms that explain the variability in clinical responses in HLA alloimmunized patients receiving platelet transfusions. Previously we showed that a subset of anti-HLA IgG-antibodies induces FcγRIIa-dependent platelet activation and enhanced phagocytosis. Here, we investigated whether anti-HLA IgG can induce complement activation on platelets. We found that a subset of anti-HLA IgG induced complement activation via the classical pathway, causing C4b and C3b deposition and formation of the membrane-attack complex. This resulted in permeabilization of platelet membranes and increased calcium influx. Complement activation also caused enhanced α-granule release, as measured by CD62P surface exposure. Blocking studies revealed that platelet activation was caused by FcγRIIa-dependent signaling as well as HLA antibody induced complement activation. Synergistic complement activation employing combinations of monoclonal IgGs suggested that assembly of oligomeric IgG complexes strongly promoted complement activation through binding of IgGs to different antigenic determinants on HLA. In agreement with this, we observed that preventing anti-HLA-IgG hexamer formation using an IgG-Fc:Fc blocking peptide, completely inhibited C3b and C4b deposition. Our results show that HLA antibodies can induce complement activation on platelets including membrane attack complex formation, pore formation and calcium influx. We propose that these events can contribute to fast platelet clearance in vivo in patients refractory to platelet transfusions with HLA alloantibodies, who may benefit from functional-platelet matching and treatment with complement inhibitors.


Assuntos
Plaquetas/imunologia , Via Clássica do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Antígenos HLA/imunologia , Isoanticorpos/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Plaquetas/metabolismo , Cálcio/metabolismo , Via Clássica do Complemento/efeitos dos fármacos , Proteínas do Sistema Complemento/metabolismo , Relação Dose-Resposta a Droga , Citometria de Fluxo , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/farmacologia , Imunoglobulinas Intravenosas/farmacologia , Isoanticorpos/farmacologia , Modelos Biológicos , Ativação Plaquetária/efeitos dos fármacos , Ligação Proteica , Receptores de IgG/metabolismo
16.
Blood ; 132(23): 2431-2440, 2018 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-30309891

RESUMO

The mechanisms by which exposure to heparin initiates antibody responses in many, if not most, recipients are poorly understood. We recently demonstrated that antigenic platelet factor 4 (PF4)/heparin complexes activate complement in plasma and bind to B cells. Here, we describe how this process is initiated. We observed wide stable variation in complement activation when PF4/heparin was added to plasma of healthy donors, indicating a responder "phenotype" (high, intermediate, or low). Proteomic analysis of plasma from these healthy donors showed a strong correlation between complement activation and plasma immunoglobulin M (IgM) levels (r = 0.898; P < .005), but not other Ig isotypes. Complement activation response to PF4/heparin in plasma displaying the low donor phenotype was enhanced by adding pooled IgM from healthy donors, but not monoclonal IgM. Depletion of IgM from plasma abrogated C3c generation by PF4/heparin. The complement-activating features of IgM are likely mediated by nonimmune, or natural, IgM, as cord blood and a monoclonal polyreactive IgM generate C3c in the presence of PF4/heparin. IgM facilitates complement and antigen deposition on B cells in vitro and in patients receiving heparin. Anti-C1q antibody prevents IgM-mediated complement activation by PF4/heparin complexes, indicating classical pathway involvement. These studies demonstrate that variability in plasma IgM levels correlates with functional complement responses to PF4/heparin. Polyreactive IgM binds PF4/heparin, triggers activation of the classical complement pathway, and promotes antigen and complement deposition on B cells. These studies provide new insights into the evolution of the heparin-induced thrombocytopenia immune response and may provide a biomarker of risk.


Assuntos
Linfócitos B/imunologia , Via Clássica do Complemento/imunologia , Heparina/imunologia , Imunoglobulina M/imunologia , Ativação Linfocitária , Fator Plaquetário 4/imunologia , Complemento C3c/imunologia , Via Clássica do Complemento/efeitos dos fármacos , Heparina/farmacologia , Humanos , Fator Plaquetário 4/farmacologia , Proteômica
17.
PLoS One ; 13(6): e0198644, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29874282

RESUMO

The complement system is an intricate defense network that rapidly removes invading pathogens. Although many complement regulators are present to protect host cells under homeostasis, the impairment of Factor H (FH) regulatory mechanism has been associated with several autoimmune and inflammatory diseases. To understand the dynamics involved in the pivotal balance between activation and regulation, we have developed a comprehensive computational model of the alternative and classical pathways of the complement system. The model is composed of 290 ordinary differential equations with 142 kinetic parameters that describe the state of complement system under homeostasis and disorder through FH impairment. We have evaluated the state of the system by generating concentration-time profiles for the biomarkers C3, C3a-desArg, C5, C5a-desArg, Factor B (FB), Ba, Bb, and fC5b-9 that are influenced by complement dysregulation. We show that FH-mediated disorder induces substantial levels of complement activation compared to homeostasis, by generating reduced levels of C3 and FB, and to a lesser extent C5, and elevated levels of C3a-desArg, Ba, Bb, C5a-desArg, and fC5b-9. These trends are consistent with clinically observed biomarkers associated with complement-mediated diseases. Furthermore, we introduced therapy states by modeling known inhibitors of the complement system, a compstatin variant (C3 inhibitor) and eculizumab (C5 inhibitor). Compstatin demonstrates strong restorative effects for early-stage biomarkers, such as C3a-desArg, FB, Ba, and Bb, and milder restorative effects for late-stage biomarkers, such as C5a-desArg and fC5b-9, whereas eculizumab has strong restorative effects on late-stage biomarkers, and negligible effects on early-stage biomarkers. These results highlight the need for patient-tailored therapies that target early complement activation at the C3 level, or late-stage propagation of the terminal cascade at the C5 level, depending on the specific FH-mediated disease and the manifestations of a patient's genetic profile in complement regulatory function.


Assuntos
Via Alternativa do Complemento/fisiologia , Via Clássica do Complemento/fisiologia , Proteínas do Sistema Complemento/metabolismo , Modelos Biológicos , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Síndrome Hemolítico-Urêmica Atípica/imunologia , Biomarcadores/análise , Biomarcadores/metabolismo , Inativadores do Complemento/farmacologia , Inativadores do Complemento/uso terapêutico , Via Alternativa do Complemento/efeitos dos fármacos , Via Clássica do Complemento/efeitos dos fármacos , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/imunologia , Biologia Computacional , Glomerulonefrite Membranoproliferativa/diagnóstico , Glomerulonefrite Membranoproliferativa/imunologia , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/fisiologia , Degeneração Macular/diagnóstico , Degeneração Macular/imunologia
18.
Clin Pharmacol Ther ; 104(4): 655-663, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29737533

RESUMO

Aberrant activation of the classical complement pathway is the common underlying pathophysiology of orphan diseases such as bullous pemphigoid, antibody-mediated rejection of organ transplants, cold agglutinin disease, and warm autoimmune hemolytic anemia. Therapeutic options for these complement-mediated disorders are limited and sutimlimab, a humanized monoclonal antibody directed against complement factor C1s, may be potentially useful for inhibition of the classical complement pathway. A phase I, first-in-human, double-blind, randomized, placebo-controlled, dose-escalation trial of single and multiple doses of sutimlimab or placebo was conducted in 64 volunteers to evaluate safety, tolerability, pharmacokinetic, and pharmacodynamic profiles. Single and multiple infusions of sutimlimab were well tolerated without any safety concerns. sutimlimab exhibited a steep concentration-effect relationship with a Hill coefficient of 2.4, and an IC90 of 15.5 µg/mL. This study establishes the foundation for using sutimlimab as a highly selective inhibitor of the classical complement pathway in different diseases.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Complemento C1s/antagonistas & inibidores , Inativadores do Complemento/administração & dosagem , Via Clássica do Complemento/efeitos dos fármacos , Adulto , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacocinética , Áustria , Complemento C1s/imunologia , Inativadores do Complemento/efeitos adversos , Inativadores do Complemento/farmacocinética , Relação Dose-Resposta a Droga , Método Duplo-Cego , Esquema de Medicação , Feminino , Voluntários Saudáveis , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Adulto Jovem
19.
Int J Toxicol ; 36(6): 449-462, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29202623

RESUMO

ANX005 is a humanized immunoglobulin G4 recombinant antibody against C1q that inhibits its function as the initiating molecule of the classical complement cascade. The safety and tolerability of ANX005 are currently being evaluated in a phase I trial in healthy volunteers ( www.clinicaltrials.gov Identifier: NCT03010046). Inhibition of C1q can be applied therapeutically in a broad spectrum of diseases, including acute antibody-mediated autoimmune disease, such as Guillain-Barré syndrome (GBS), and in chronic diseases of the central nervous system involving complement-mediated neurodegeneration, such as Alzheimer's disease (AD). To support the clinical development of ANX005, several studies were conducted to assess the pharmacology, pharmacokinetics, and potential toxicity of ANX005. ANX-M1, the murine precursor of ANX005, functionally inhibits the classical complement cascade both in vitro and in vivo, to protect against disease pathology in mouse models of GBS and AD. Toxicology studies with ANX005, itself, showed that intravenous administration once weekly for 4 weeks was well tolerated in rats and monkeys, with no treatment-related adverse findings. Serum levels of ANX005 in monkeys correlate with a reduction in free C1q levels both in the serum and in the cerebrospinal fluid. In summary, ANX005 has shown proof of concept in in vitro and in vivo nonclinical pharmacology models, with no toxicity in the 4-week repeat-dose studies in rats and monkeys. The no observed adverse effect level was 200 mg/kg/dose, which is 200-fold higher than the first-in-human starting dose of 1 mg/kg in healthy volunteers.


Assuntos
Anticorpos Monoclonais Humanizados/toxicidade , Doenças Autoimunes/tratamento farmacológico , Complemento C1q/imunologia , Doenças Neurodegenerativas/tratamento farmacológico , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Doenças Autoimunes/imunologia , Complemento C1q/metabolismo , Via Clássica do Complemento/efeitos dos fármacos , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Injeções Intravenosas , Macaca fascicularis , Masculino , Doenças Neurodegenerativas/imunologia , Ratos Sprague-Dawley , Especificidade da Espécie
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...