Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
1.
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
2.
Bioessays ; 41(10): e1900074, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31452228

RESUMO

The human immune system can directly lyse invading micro-organisms and aberrant host cells by generating pores in the cell envelope, called membrane attack complexes (MACs). Recent studies using single-particle cryoelectron microscopy have revealed that the MAC is an asymmetric, flexible pore and have provided a structural basis on how the MAC ruptures single lipid membranes. Despite these insights, it remains unclear how the MAC ruptures the composite cell envelope of Gram-negative bacteria. Recent functional studies on Gram-negative bacteria elucidate that local assembly of MAC pores by surface-bound C5 convertase enzymes is essential to stably insert these pores into the bacterial outer membrane (OM). These convertase-generated MAC pores can subsequently efficiently damage the bacterial inner membrane (IM), which is essential for bacterial killing. This review summarizes these recent insights of MAC assembly and discusses how MAC pores kill Gram-negative bacteria. Furthermore, this review elaborates on how MAC-dependent OM damage could lead to IM destabilization, which is currently not well understood. A better understanding on how MAC pores kill bacteria could facilitate the future development of novel strategies to treat infections with Gram-negative bacteria.


Assuntos
Membrana Externa Bacteriana , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Bactérias Gram-Negativas , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Humanos
3.
Exp Eye Res ; 184: 266-277, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31082363

RESUMO

Data from human dry and wet age-related macular degeneration (AMD) eyes support the hypothesis that constant 'tickover' of the alternative complement pathway results in chronic deposition of the complement membrane attack complex (MAC) on the choriocapillaris and the retinal pigment epithelium (RPE). Sub-lytic levels of MAC lead to cell signaling associated with tissue remodeling and the production of cytokines and inflammatory molecules. Lytic levels of MAC lead to cell death. CD59 is a naturally occurring inhibitor of the assembly of MAC. CD59 may thus be therapeutically efficacious against the pathophysiology of dry and wet AMD. The first gene therapy clinical trial for geographic atrophy - the advanced form of dry AMD has recently completed recruitment. This trial is studying the safety and tolerability of expressing CD59 from an adeno-associated virus (AAV) vector injected once into the vitreous. A second clinical trial assessing the efficacy of CD59 in wet AMD patients is also under way. Herein, the evidence for the role of MAC in the pathophysiology of dry as well as wet AMD and the scientific rationale underlying the use of AAV- delivered CD59 for the treatment of dry and wet AMD is discussed.


Assuntos
Antígenos CD59/uso terapêutico , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Terapia Genética , Atrofia Geográfica/fisiopatologia , Degeneração Macular Exsudativa/fisiopatologia , Animais , Antígenos CD59/genética , Ensaios Clínicos como Assunto , Dependovirus/genética , Vetores Genéticos , Atrofia Geográfica/terapia , Humanos , Injeções Intravítreas , Degeneração Macular Exsudativa/terapia
4.
J Leukoc Biol ; 106(1): 187-192, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30821872

RESUMO

In this mini review, we describe the molecular mechanisms in polymicrobial sepsis that lead to a series of adverse events including activation of inflammatory and prothrombotic pathways, a faulty innate immune system, and multiorgan dysfunction. Complement activation is a well-established feature of sepsis, especially involving generation of C5a and C5b-9, along with engagement of relevant receptors for C5a. Activation of neutrophils by C5a leads to extrusion of DNA, forming neutrophil extracellular traps that contain myeloperoxidase and oxidases, along with extracellular histones. Generation of the distal complement activation product, C5b-9 (known as the membrane attack complex, MAC), also occurs in sepsis. C5b-9 activates the NLRP3 inflammasome, which damages mitochondria, together with appearance in plasma of IL-1ß and IL-18. Histones are strongly proinflammatory as well as being prothrombotic, leading to activation of platelets and development of venous thrombosis. Multiorgan dysfunction is also a feature of sepsis. It is well known that septic cardiomyopathy, which if severe, can lead to death. This complication in sepsis is linked to reduced levels in cardiomyocytes of three critical proteins (SERCA2, NCX, Na+ /K+ -ATPase). The reductions in these three key proteins are complement- and histone-dependent. Dysfunction of these ATPases is linked to the cardiomyopathy of sepsis. These data suggest novel targets in the setting of sepsis in humans.


Assuntos
Doenças Transmissíveis/imunologia , Sepse/imunologia , Animais , Cardiomiopatias/etiologia , Doenças Transmissíveis/tratamento farmacológico , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Proteínas do Sistema Complemento/fisiologia , Histonas/fisiologia , Humanos , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Sepse/complicações , Sepse/tratamento farmacológico
5.
Transplant Proc ; 50(6): 1697-1700, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30056884

RESUMO

Complement activation is considered one of the mediators of renal ischemia-reperfusion injury. Elevated levels of C5b-9, C3a, and C5a are detected in sera of deceased kidney donors. The goal of the study was to characterize the functional activity of complement pathways in donor sera and to assess their influence on transplant outcome. MATERIALS AND METHODS: Sixty-four deceased kidney donors (age 45 ± 16 years; 28 female, 36 male) and 27 healthy controls (age 42 ± 12 years; 14 female, 13 male) were enrolled in the study. The results of transplantation for the respective 122 kidney recipients were included in the analysis. The functional activities of classical (CP), lectin (LP), and alternative (AP) pathways were measured using Wielisa-kit (reference normal level = 100%). In most cases, decreased functional activity reflects the activation status of the pathway. RESULTS: The median (interquartile range) functional activities of the pathways in donor sera were CP 118 (89-150)%, LP 80 (20-127)%, and AP 74 (50-89)%, and did not differ from the control values CP 110 (102-115)%, LP 81 (26-106)%, AP 76 (61-88)%. The frequency of pathway activation observed in controls was CP 0%, LP 11%, and AP 0%. Deceased donors did not differ in activation of classical (11%) and lectin (13%) pathways, but presented a higher rate of alternative pathway activation (19%, P = .03). No significant influence of any pathway functional activity or its activation was proved to influence the transplant outcome. CONCLUSION: Complement activation via alternative pathway was observed in diseased donor sera. No predictive potential of donor complement functional activity on the transplant outcome could be proved.


Assuntos
Aloenxertos/metabolismo , Complemento C3a/fisiologia , Complemento C5a/fisiologia , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Rim/metabolismo , Doadores de Tecidos , Adulto , Ativação do Complemento , Feminino , Humanos , Transplante de Rim , Lectinas/sangue , Masculino , Pessoa de Meia-Idade , Traumatismo por Reperfusão/etiologia
6.
Curr Hypertens Rep ; 19(11): 87, 2017 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-29046976

RESUMO

PURPOSE OF REVIEW: Preeclampsia affects 3-4% of pregnancies with few treatment options to reduce maternal and fetal harm. Recent evidence that targeting the complement system may be an effective therapeutic strategy in prevention or treatment of preeclampsia will be reviewed. RECENT FINDINGS: Studies in humans confirm the safety and efficacy of C5 blockade in complement-mediated disorders of pregnancy, including preeclampsia. Animal models mimic the placental abnormalities and/or the maternal symptoms which characterize preeclampsia. These models in mouse and rat have defined a role for complement and its regulators in placental dysfunction, hypertension, proteinuria, endothelial dysfunction, fetal growth restriction, and angiogenic imbalance, thus informing future human studies. Targeting excessive complement activation, particularly the terminal complement complex (C5b-9) and C5a may be an effective strategy to prolong pregnancy in women with preeclampsia. Continued research is needed to identify the initiator(s) of activation, the pathways involved, and the key component(s) in the pathophysiology to allow development of safe and effective therapeutics to target complement without compromising its role in homeostasis and host defense.


Assuntos
Ativação do Complemento/fisiologia , Placenta/fisiopatologia , Pré-Eclâmpsia/fisiopatologia , Animais , Complemento C5a/fisiologia , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Modelos Animais de Doenças , Endotélio Vascular/fisiopatologia , Feminino , Retardo do Crescimento Fetal/fisiopatologia , Homeostase , Humanos , Hipertensão/fisiopatologia , Camundongos , Neovascularização Patológica/fisiopatologia , Gravidez , Proteinúria/fisiopatologia , Ratos
7.
Nat Rev Nephrol ; 13(5): 311-318, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28262777

RESUMO

The development of type 1 and type 2 diabetes mellitus has a substantial negative impact on morbidity and mortality and is responsible for substantial individual and socioeconomic costs worldwide. One of the most serious consequences of diabetes mellitus is the development of diabetic angiopathy, which manifests clinically as microvascular and macrovascular complications. One microvascular complication, diabetic nephropathy, is the most common cause of end-stage renal disease in developed countries. Although several available therapeutic interventions can delay the onset and progression of diabetic nephropathy, morbidity associated with this disease remains high and new therapeutic approaches are needed. In addition, not all patients with diabetes mellitus will develop diabetic nephropathy and thus new biomarkers are needed to identify individuals who will develop this life-threatening disease. An increasing body of evidence points toward a role of the complement system in the pathogenesis of diabetic nephropathy. For example, circulating levels of mannose-binding lectin (MBL), a pattern recognition molecule of the innate immune system, have emerged as a robust biomarker for the development and progression of this disease, and evidence suggests that MBL, H-ficolin, complement component C3 and the membrane attack complex might contribute to renal injury in the hyperglycaemic mileu. New approaches to modulate the complement system might lead to the development of new agents to prevent or slow the progression of diabetic nephropathy.


Assuntos
Proteínas do Sistema Complemento/fisiologia , Nefropatias Diabéticas/etiologia , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/epidemiologia , Glicoproteínas/fisiologia , Humanos , Lectinas/fisiologia , Lectina de Ligação a Manose/sangue
8.
Invest Ophthalmol Vis Sci ; 57(14): 5910-5918, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27802521

RESUMO

PURPOSE: Age-related macular degeneration (AMD) commonly causes blindness in the elderly. Yet, it is untreatable in the large fraction of all AMD patients that develop the early dry form. Dry AMD is marked by the deposition of membrane attack complex (MAC) on choriocapillaris (CC), which is implicated in CC degeneration and subsequent atrophy of overlying retinal pigment epithelium. Since MAC is also found on the CC of young eyes, here we investigated whether and how aging increases choroidal endothelial susceptibility to MAC injury. METHODS: Monkey chorioretinal endothelial cells (ECs, RF/6A) were cultured to high passages (>P60) to achieve replicative senescence. We treated ECs with complement-competent human serum to promote MAC deposition and injury, which were assessed by flow cytometry and trypan blue exclusion assay, respectively. Stiffness of EC was measured by atomic force microscopy indentation while Rho GTPase activity was quantified by Rho G-LISA assay. RESULTS: Our findings reveal that senescent ECs are significantly stiffer than their normal counterparts, which correlates with higher cytoskeletal Rho activity in these cells and their greater susceptibility to complement (MAC) injury. Importantly, inhibition of Rho activity in senescent ECs significantly reduced cell stiffness and MAC-induced lysis. CONCLUSIONS: By revealing an important role of senescence-associated choroidal EC stiffening in complement injury, these findings implicate CC stiffening as an important determinant of age-related CC atrophy seen in dry AMD. Future studies are needed to validate these findings in appropriate animal models so new therapeutic targets can be identified for treatment of dry AMD.


Assuntos
Senescência Celular/fisiologia , Corioide/efeitos dos fármacos , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Proteínas do Sistema Complemento/farmacologia , Células Endoteliais/efeitos dos fármacos , Degeneração Macular/patologia , Degeneração Macular/fisiopatologia , Idoso , Animais , Células Cultivadas , Corioide/citologia , Corioide/fisiologia , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Células Endoteliais/fisiologia , Haplorrinos , Humanos , Masculino , Microscopia de Força Atômica , Retina/citologia
9.
J Dent Res ; 94(12): 1765-72, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26464397

RESUMO

Complement system activation has been shown to be involved in inflammation and regeneration processes that can be observed within the dental pulp after moderate carious decay. Studies simulating carious injuries in vitro have shown that when human pulp fibroblasts are stimulated by lipoteichoic acid (LTA), they synthetize all complement components. Complement activation leads to the formation of the membrane attack complex (MAC), which is known for its bacterial lytic effect. This work was designed to find out whether human pulp fibroblasts can kill Streptococcus mutans and Streptococcus sanguinis via complement activation. First, histological staining of carious tooth sections showed that the presence of S. mutans correlated with an intense MAC staining. Next, to simulate bacterial infection in vitro, human pulp fibroblasts were incubated in serum-free medium with LTA. Quantification by an enzymatic assay showed a significant increase of MAC formation on bacteria grown in this LTA-conditioned medium. To determine whether the MAC produced by pulp fibroblasts was functional, bacteria sensitivity to LTA-conditioned medium was evaluated using agar well diffusion assay and succinyl dehydrogenase (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide [MTT]) assay. Both assays showed that S. mutans and S. sanguinis were sensitive to LTA-conditioned medium. Finally, to evaluate whether MAC formation on cariogenic bacteria, by pulp fibroblasts, can be directly induced by the presence of these bacteria, a specific coculture model of human pulp fibroblasts and bacteria was developed. Immunofluorescence revealed an intense MAC labeling on bacteria after direct contact with pulp fibroblasts. The observed MAC formation and its lethal effects were significantly reduced when CD59, an inhibitor of MAC formation, was added. Our findings demonstrate that the MAC produced by LTA-stimulated pulp fibroblasts is functional and can kill S. mutans and S. sanguinis. Taken together, these data clearly highlight the function of pulp fibroblasts in destroying cariogenic bacteria.


Assuntos
Ativação do Complemento/fisiologia , Cárie Dentária/microbiologia , Polpa Dentária/citologia , Fibroblastos/fisiologia , Células Cultivadas , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Polpa Dentária/fisiologia , Fibroblastos/imunologia , Imunofluorescência , Humanos , Streptococcus mutans/imunologia , Streptococcus sanguis/imunologia
10.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 46(2): 173-8, 227, 2015 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-25924424

RESUMO

OBJECTIVE: To explore the protective effect of Cordycepin (3'-deoxyadenosine), a bioactive compound of Cordyceps Sinensis, on injury of podocytes. METHODS: C5b-9-induced podocyte injury was used as a model of membranous nephropathy in vitro. This model was established using mouse podocyte cell line--MPC5. Cordycepin was given as an intervention. Ultra-micro morphological changes were observed by electron microscope. F-actin cytoskeleton and expression of nephrin were observed by fluorescence microscope. The phosphorylation of mitogen-activated protein kinase (MAPK) was measured by Western blot. RESULTS: Stimulated by C5b-9 for 3 h, MPC5 showed secondary foot processes, with cytoskeleton structure damaged, nephrin relocated from the cell surface to the cytoplasm, and cell signal pathway-p38, JNK and ERK activated. Cordycepin protected foot processes and cytoskeleton structures of podocytes, suppressed the redistribution of nephrin, and inhibited p38/JNK action. CONCLUSION: Cordycepin can protect podocyte from C5b-9-induced injury partly through inhibiting the activation of p38/JNK signaling pathway.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Desoxiadenosinas/farmacologia , Podócitos/efeitos dos fármacos , Citoesqueleto de Actina/ultraestrutura , Animais , Linhagem Celular , Sistema de Sinalização das MAP Quinases , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Substâncias Protetoras/farmacologia
11.
Semin Thromb Hemost ; 40(5): 544-50, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24967890

RESUMO

Molecular linkages between von Willebrand factor (VWF) and the alternative complement pathway (AP) have recently been discovered. Endothelial cell (EC)-anchored ultra-large (UL) VWF multimeric strings function as an activating surface for the AP. C3 (in active C3b form) binds to the EC-anchored ULVWF strings, and promotes the assembly of C3bBb (C3 convertase) and C3bBbC3b (C5 convertase). These linkages may help to explain enigmatic clinical problems related to thrombotic microangiopathies, including some cases of refractory thrombotic thrombocytopenic purpura (TTP), TTP associated with only mild-modest deficiencies of ADAMTS-13, the provocation (or exacerbation) of acute episodes in patients with the atypical hemolytic uremic syndrome, and thrombosis in paroxysmal nocturnal hemoglobinuria. Recent experiments have also demonstrated that complement factor H performs a dual role: participating in regulation of the AP by binding to EC-anchored ULVWF strings; and functioning as a reductase to decrease the size of soluble VWF multimers.


Assuntos
Via Alternativa do Complemento/fisiologia , Microangiopatias Trombóticas/fisiopatologia , Fator de von Willebrand/fisiologia , Síndrome Hemolítico-Urêmica Atípica/fisiopatologia , Fator H do Complemento/fisiologia , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Hemoglobinúria Paroxística/fisiopatologia , Humanos , Púrpura Trombocitopênica Trombótica/fisiopatologia , Trombose/fisiopatologia , Microangiopatias Trombóticas/genética
12.
Subcell Biochem ; 80: 241-53, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24798015

RESUMO

Apicomplexans are eukaryotic parasites of major medical and veterinary importance. They have complex life cycles through frequently more than one host, interact with many cell types in their hosts, and can breach host cell membranes during parasite traversal of, or egress from, host cells. Some of these parasites make a strikingly heavy use of the pore-forming MACPF domain, and encode up to 10 different MACPF domain-containing proteins. In this chapter, we focus on the two most studied and medically important apicomplexans, Plasmodium and Toxoplasma, and describe the known functions of their MACPF polypeptide arsenal. Apicomplexan MACPF proteins appear to be involved in a variety of membrane-damaging events, making them an attractive model to dissect the structure-function relationships of the MACPF domain.


Assuntos
Apicomplexa/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Perforina/fisiologia , Plasmodium falciparum/metabolismo , Animais , Apicomplexa/crescimento & desenvolvimento , Complexo de Ataque à Membrana do Sistema Complemento/química , Humanos , Estágios do Ciclo de Vida , Malária/parasitologia , Perforina/química , Plasmodium falciparum/crescimento & desenvolvimento , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo
13.
Subcell Biochem ; 80: 255-69, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24798016

RESUMO

Chlamydiae are obligate intracellular bacterial parasites that infect a wide range of metazoan hosts. Some Chlamydia species are important causes of chronic inflammatory diseases of the ocular, genital and respiratory tracts in humans. Genes located in a variable region of chlamydial genomes termed the plasticity zone are known to be key determinants of pathogenic diversity. The plasticity zone protein CT153, present only in select species, contains a membrane attack complex/perforin (MACPF) domain, which may mediate chlamydial interactions with the host cell. CT153 is present throughout the C. trachomatis developmental cycle and is processed into polypeptides that interact with membranes differently than does the parent protein. Chlamydiae interact extensively with membranes from the time of invasion until they eventually exit host cells, so numerous roles for a MACPF protein in pathogenesis of these pathogens are conceivable. Here, we present an overview of what is known about CT153 and highlight potential roles of a MACPF family protein in a group of pathogens whose intracellular development is marked by a series of interactions with host cell membranes and organelles. Finally, we identify new strategies for identifying CT153 functions made feasible by the recent development of a basic toolset for genetic manipulation of chlamydiae.


Assuntos
Proteínas de Bactérias/fisiologia , Chlamydia trachomatis , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Perforina/fisiologia , Animais , Proteínas de Bactérias/química , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/genética , Chlamydia trachomatis/crescimento & desenvolvimento , Chlamydia trachomatis/patogenicidade , Complexo de Ataque à Membrana do Sistema Complemento/química , Interações Hospedeiro-Patógeno , Humanos , Perforina/química
14.
Subcell Biochem ; 80: 271-91, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24798017

RESUMO

Proteins with membrane-attack complex/perforin (MACPF) domains are found in almost all kingdoms of life, and they have a variety of biological roles, including defence and attack, organism development, and cell adhesion and signalling. The distribution of these proteins in fungi appears to be restricted to some Pezizomycotina and Basidiomycota species only, in correlation with another group of proteins with unknown biological function, known as aegerolysins. These two protein groups coincide in only a few species, and they might operate in concert as cytolytic bi-component pore-forming agents. Representative proteins here include pleurotolysin B, which has a MACPF domain, and the aegerolysin-like protein pleurotolysin A, and the very similar ostreolysin A, which have been purified from oyster mushroom (Pleurotus ostreatus). These have been shown to act in concert to perforate natural and artificial lipid membranes with high cholesterol and sphingomyelin content. The aegerolysin-like proteins provide the membrane cholesterol/sphingomyelin selectivity and recruit oligomerised pleurotolysin B molecules, to create a membrane-inserted pore complex. The resulting protein structure has been imaged with electron microscopy, and it has a 13-meric rosette-like structure, with a central lumen that is ~4-5 nm in diameter. The opened transmembrane pore is non-selectively permeable for ions and smaller neutral solutes, and is a cause of cytolysis of a colloid-osmotic type. The biological significance of these proteins for the fungal life-style is discussed.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Proteínas Fúngicas/fisiologia , Proteínas Hemolisinas/fisiologia , Perforina/fisiologia , Proteínas Citotóxicas Formadoras de Poros/fisiologia , Sequência de Aminoácidos , Animais , Complexo de Ataque à Membrana do Sistema Complemento/química , Proteínas Fúngicas/química , Proteínas Hemolisinas/química , Humanos , Dados de Sequência Molecular , Perforina/química , Filogenia , Pleurotus/genética , Pleurotus/patogenicidade , Proteínas Citotóxicas Formadoras de Poros/química , Multimerização Proteica/fisiologia , Homologia de Sequência de Aminoácidos
16.
Infect Immun ; 82(6): 2378-89, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24664511

RESUMO

Haemophilus influenzae type b (Hib) escapes the host immune system by recruitment of the complement regulator vitronectin, which inhibits the formation of the membrane attack complex (MAC) by inhibiting C5b-C7 complex formation and C9 polymerization. We reported previously that Hib acquires vitronectin at the surface by using Haemophilus surface fibrils (Hsf). Here we studied in detail the interaction between Hsf and vitronectin and its role in the inhibition of MAC formation and the invasion of lung epithelial cells. The vitronectin-binding region of Hsf was defined at the N-terminal region comprising Hsf amino acids 429 to 652. Moreover, the Hsf recognition site on vitronectin consisted of the C-terminal amino acids 352 to 374. H. influenzae was killed more rapidly in vitronectin-depleted serum than in normal human serum (NHS), and increased MAC deposition was observed at the surface of an Hsf-deficient H. influenzae mutant. In parallel, Hsf-expressing Escherichia coli selectively acquired vitronectin from serum, resulting in significant inhibition of the MAC. Moreover, when vitronectin was bound to Hsf, increased bacterial adherence and internalization into epithelial cells were observed. Taking our findings together, we have defined a fine-tuned protein-protein interaction between Hsf and vitronectin that may contribute to increased Hib virulence.


Assuntos
Adesinas Bacterianas/fisiologia , Atividade Bactericida do Sangue/imunologia , Adesão Celular/fisiologia , Haemophilus influenzae tipo b/imunologia , Vitronectina/metabolismo , Análise de Variância , Ligação Competitiva , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Relação Dose-Resposta Imunológica , Haemophilus influenzae tipo b/patogenicidade , Heparina/fisiologia , Humanos , Soro/imunologia , Soro/microbiologia
17.
Biochemistry ; 53(12): 1908-15, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24597946

RESUMO

The complement terminal pathway clears pathogens by generating cytotoxic membrane attack complex (MAC) pores on target cells. For more than 40 years, biochemical and cellular assays have been used to characterize the lytic nature of the MAC and to define its protein composition. Although models for pore formation have been inferred from structures of bacterial cytolysins, it was only recently that we were able to visualize how complement components come together during MAC assembly. This review highlights structural analyses of terminal pathway complexes to explore molecular mechanisms underlying MAC formation.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/química , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Substâncias Macromoleculares/química , Animais , Humanos , Substâncias Macromoleculares/metabolismo , Substâncias Macromoleculares/uso terapêutico , Neoplasias/química , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
18.
FASEB J ; 28(3): 1511-25, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24344329

RESUMO

CCAAT/enhancer-binding protein (C/EBPß)-enhanced IL-6 and TGF-ß1 promoter activity and p300-mediated C/EBPß acetylation were involved in up-regulation of IL-6 and TGF-ß1 expression in GMCs attacked by sublytic C5b-9. In detail, the elements of C/EBPß binding to rat IL-6 and TGF-ß1 promoter and 3 acetylated sites of rat C/EBPß protein were first revealed. Furthermore, silencing the p300 or C/EBPß gene in rat kidney significantly reduced the production of IL-6 and TGF-ß1 and renal lesions in Thy-1N rats. Together, these data indicate that the mechanism of IL-6 and TGF-ß1 production in renal tissue of Thy-1N rats is associated with sublytic C5b-9 up-regulated p300 and p300-mediated C/EBPß acetylation as well as C/EBPß-activated IL-6 and TGF-ß1 genes.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Mesângio Glomerular/metabolismo , Interleucina-6/biossíntese , Nefrite/metabolismo , Fator de Crescimento Transformador beta1/biossíntese , Fatores de Transcrição de p300-CBP/metabolismo , Acetilação , Animais , Sequência de Bases , Células Cultivadas , Primers do DNA , Mesângio Glomerular/citologia , Interleucina-17/biossíntese , Masculino , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real
19.
J Clin Immunol ; 33(1): 162-71, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22926405

RESUMO

PURPOSE: Acute Hemorrhagic Leukoencephalitis (AHLE) is a rare demyelinating disorder of acute onset, rapid deterioration and significant morbidity and mortality. Most often described as a post-infectious complication of an upper respiratory illness, its precise pathophysiology remains unclear. We describe two pediatric patients with AHLE with partial complement factor I (FI) deficiency whose successful treatment included the interleukin-1 (IL-1) receptor antagonist, anakinra, implicating a role for FI and IL-1 in this disorder. METHODS: Extensive clinical workup of two patients presenting with AHLE revealed complement abnormalities, specifically related to the alternative pathway and its regulator, FI. Aggressive management with steroids, immunoglobulin, and anakinra ultimately led to improvement of clinical status and near return to neurologic baseline in both patients. Genetic sequencing of the FI coding regions of the patients and their families was performed. In vitro protein expression studies and immunohistochemistry of fixed brain tissue was used to investigate pathogenic mechanisms. RESULTS: Two novel mutations in FI were identified in our patients, which result in failure to secrete FI. Immunohistochemical evaluation of brain tissue demonstrated positive staining for C3, membrane attack complex (MAC) and IL-1. CONCLUSIONS: We propose AHLE is an unreported, rare phenotype for partial FI deficiency. The upregulation of C3, MAC and IL-1 with subsequent demyelination support a pathologic role for complement activation in AHLE, and suggest anakinra as an important adjunctive therapy in this disease.


Assuntos
Fator I do Complemento/genética , Leucoencefalite Hemorrágica Aguda/genética , Leucoencefalite Hemorrágica Aguda/imunologia , Mutação de Sentido Incorreto/imunologia , Neurônios/imunologia , Neurônios/patologia , Adolescente , Adulto , Criança , Ativação do Complemento/genética , Ativação do Complemento/imunologia , Complemento C3/fisiologia , Fator I do Complemento/deficiência , Fator I do Complemento/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Feminino , Células HEK293 , Humanos , Imunofenotipagem , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Interleucina-1/fisiologia , Leucoencefalite Hemorrágica Aguda/patologia , Masculino , Neurônios/metabolismo , Linhagem
20.
Immunol Res ; 56(1): 109-21, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23247987

RESUMO

Complement system activation plays an important role in both innate and acquired immunity, with the activation of complement and the subsequent formation of C5b-9 terminal complement complex on cell membranes inducing target cell death. Recognition of this role for C5b-9 leads to the assumption that C5b-9 might play an antitumor role. However, sublytic C5b-9 induces cell cycle progression by activating signal transduction pathways and transcription factors in cancer cells, indicating a role in tumor promotion for this complement complex. The induction of the cell cycle by C5b-9 is dependent upon the activation of the phosphatidylinositol 3-kinase (PI3K)/Akt/FOXO1 and ERK1 pathways in a Gi protein-dependent manner. C5b-9 also induces response gene to complement (RGC)-32, a gene that plays a role in cell cycle promotion through activation of Akt and the CDC2 kinase. RGC-32 is expressed by tumor cells and plays a dual role in cancers, in that it has both a tumor suppressor role and tumor-promoting activity. Thus, through the activation of tumor cells, the C5b-9-mediated induction of the cell cycle plays an important role in tumor proliferation and oncogenesis.


Assuntos
Carcinogênese/genética , Proteínas de Ciclo Celular/genética , Complexo de Ataque à Membrana do Sistema Complemento/fisiologia , Regulação Neoplásica da Expressão Gênica , Proteínas Musculares/genética , Proteínas do Tecido Nervoso/genética , Animais , Ciclo Celular , Morte Celular , Citotoxicidade Imunológica , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Genes Supressores de Tumor , Humanos , Sistema de Sinalização das MAP Quinases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA