Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
Front Immunol ; 10: 752, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31024572

RESUMO

The interactions of cancer cells with components of the complement system are highly complex, leading to an outcome that is either favorable or detrimental to cancer cells. Currently, we perceive only the "tip of the iceberg" of these interactions. In this review, we focus on the complement terminal C5b-9 complex, known also as the complement membrane attack complex (MAC) and discuss the complexity of its interaction with cancer cells, starting with a discussion of its proposed mode of action in mediating cell death, and continuing with a portrayal of the strategies of evasion exhibited by cancer cells, and closing with a proposal of treatment approaches targeted at evasion strategies. Upon intense complement activation and membrane insertion of sufficient C5b-9 complexes, the afflicted cells undergo regulated necrotic cell death with characteristic damage to intracellular organelles, including mitochondria, and perforation of the plasma membrane. Several pro-lytic factors have been proposed, including elevated intracellular calcium ion concentrations and activated JNK, Bid, RIPK1, RIPK3, and MLKL; however, further research is required to fully characterize the effective cell death signals activated by the C5b-9 complexes. Cancer cells over-express a multitude of protective measures which either block complement activation, thus reducing the number of membrane-inserted C5b-9 complexes, or facilitate the elimination of C5b-9 from the cell surface. Concomitantly, cancer cells activate several protective pathways that counteract the death signals. Blockage of complement activation is mediated by the complement membrane regulatory proteins CD46, CD55, and CD59 and by soluble complement regulators, by proteases that cleave complement proteins and by protein kinases, like CK2, which phosphorylate complement proteins. C5b-9 elimination and inhibition of cell death signals are mediated by caveolin and dynamin, by Hsp70 and Hsp90, by the mitochondrial stress protein mortalin, and by the protein kinases PKC and ERK. It is conceivable that various cancers and cancers at different stages of development will utilize distinct patterns of these and other MAC resistance strategies. In order to enhance the impact of antibody-based therapy on cancer, novel precise reagents that block the most effective protective strategies will have to be designed and applied as adjuvants to the therapeutic antibodies.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Neoplasias/imunologia , Animais , Sinalização do Cálcio , Morte Celular/imunologia , Membrana Celular/imunologia , Membrana Celular/metabolismo , Ativação do Complemento , Proteínas Inativadoras do Complemento/imunologia , Proteínas Inativadoras do Complemento/metabolismo , Citotoxicidade Imunológica , Humanos , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Modelos Imunológicos , Neoplasias/metabolismo , Neoplasias/patologia , Microambiente Tumoral/imunologia
2.
Nat Rev Dis Primers ; 3: 17028, 2017 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-28516949

RESUMO

Paroxysmal nocturnal haemoglobinuria (PNH) is a clonal haematopoietic stem cell (HSC) disease that presents with haemolytic anaemia, thrombosis and smooth muscle dystonias, as well as bone marrow failure in some cases. PNH is caused by somatic mutations in PIGA (which encodes phosphatidylinositol N-acetylglucosaminyltransferase subunit A) in one or more HSC clones. The gene product of PIGA is required for the biosynthesis of glycosylphosphatidylinositol (GPI) anchors; thus, PIGA mutations lead to a deficiency of GPI-anchored proteins, such as complement decay-accelerating factor (also known as CD55) and CD59 glycoprotein (CD59), which are both complement inhibitors. Clinical manifestations of PNH occur when a HSC clone carrying somatic PIGA mutations acquires a growth advantage and differentiates, generating mature blood cells that are deficient of GPI-anchored proteins. The loss of CD55 and CD59 renders PNH erythrocytes susceptible to intravascular haemolysis, which can lead to thrombosis and to much of the morbidity and mortality of PNH. The accumulation of anaphylatoxins (such as C5a) from complement activation might also have a role. The natural history of PNH is highly variable, ranging from quiescent to life-threatening. Therapeutic strategies include terminal complement blockade and bone marrow transplantation. Eculizumab, a monoclonal antibody complement inhibitor, is highly effective and the only licensed therapy for PNH.


Assuntos
Hemoglobinúria Paroxística/patologia , Hemoglobinúria Paroxística/terapia , Proteínas de Membrana/genética , Anticorpos Monoclonais Humanizados/uso terapêutico , Transplante de Medula Óssea , Antígenos CD55/metabolismo , Antígenos CD59/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Hemoglobinúria Paroxística/genética , Hemoglobinúria Paroxística/metabolismo , Humanos , Mutação , Resultado do Tratamento
3.
J Gen Virol ; 97(8): 1765-1770, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27153814

RESUMO

Newcastle disease virus (NDV) is being developed as an oncolytic virus for virotherapy. In this study we analysed the regulation of complement-mediated inactivation of a recombinant NDV in different host cells. NDV grown in human cells was less sensitive to complement-mediated virus inactivation than NDV grown in embryonated chicken eggs. Additionally, NDV produced from HeLa-S3 cells is more resistant to complement than NDV from 293F cells, which correlated with higher expression and incorporation of complement regulatory proteins (CD46, CD55 and CD59) into virions from HeLa-S3 cells. Further analysis of the recombinant NDVs individually expressing the three CD molecules showed that CD55 is the most potent in counteracting complement-mediated virus inactivation. The results provide important information on selecting NDV manufacture substrate to mitigate complement-mediated virus inactivation.


Assuntos
Antígenos CD55/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Proteínas do Sistema Complemento/metabolismo , Interações Hospedeiro-Patógeno , Fatores Imunológicos/metabolismo , Vírus da Doença de Newcastle/imunologia , Vírus da Doença de Newcastle/fisiologia , Animais , Antígenos CD59/metabolismo , Linhagem Celular , Galinhas , Humanos , Proteína Cofatora de Membrana/metabolismo
4.
Biochem Soc Trans ; 43(5): 812-8, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26517887

RESUMO

Complement control protein modules (CCPs) occur in numerous functionally diverse extracellular proteins. Also known as short consensus repeats (SCRs) or sushi domains each CCP contains approximately 60 amino acid residues, including four consensus cysteines participating in two disulfide bonds. Varying in length and sequence, CCPs adopt a ß-sandwich type fold and have an overall prolate spheroidal shape with N- and C-termini lying close to opposite poles of the long axis. CCP-containing proteins are important as cytokine receptors and in neurotransmission, cell adhesion, blood clotting, extracellular matrix formation, haemoglobin metabolism and development, but CCPs are particularly well represented in the vertebrate complement system. For example, factor H (FH), a key soluble regulator of the alternative pathway of complement activation, is made up entirely from a chain of 20 CCPs joined by short linkers. Collectively, therefore, the 20 CCPs of FH must mediate all its functional capabilities. This is achieved via collaboration and division of labour among these modules. Structural studies have illuminated the dynamic architectures that allow FH and other CCP-rich proteins to perform their biological functions. These are largely the products of a highly varied set of intramolecular interactions between CCPs. The CCP can act as building block, spacer, highly versatile recognition site or dimerization mediator. Tandem CCPs may form composite binding sites or contribute to flexible, rigid or conformationally 'switchable' segments of the parent proteins.


Assuntos
Enzimas Ativadoras do Complemento/química , Ativação do Complemento , Proteínas Inativadoras do Complemento/química , Desenho de Fármacos , Modelos Moleculares , Engenharia de Proteínas , Animais , Sítios de Ligação , Enzimas Ativadoras do Complemento/genética , Enzimas Ativadoras do Complemento/metabolismo , Fator H do Complemento/química , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Inativadores do Complemento/química , Inativadores do Complemento/metabolismo , Inativadores do Complemento/farmacologia , Proteínas Inativadoras do Complemento/genética , Proteínas Inativadoras do Complemento/metabolismo , Humanos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia
5.
Immunobiology ; 217(11): 1080-7, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22964233

RESUMO

The recent availability of eculizumab as the first complement inhibitor renewed the interest for complement-mediated damage in several human diseases. Paroxysmal nocturnal hemoglobinuria (PNH) may be considered the paradigm a disease caused by complement dysregulation specifically on erythrocytes; in fact, PNH is a clonal, non-malignant, hematological disorder characterized by the expansion of hematopoietic stem cells and progeny mature blood cells which are deficient in some surface proteins, including the two complement regulators CD55 and CD59. As a result, PNH erythrocytes are incapable to modulate on their surface physiologic complement activation, which eventually enables the terminal lytic complement leading to complement-mediated intravascular anemia - the typical clinical hallmark of PNH. In the last decade the anti-C5 monoclonal antibody has been proven effective for the treatment of PNH, resulting in a sustained control of complement-mediated intravascular hemolysis, with a remarkable clinical benefit. Since then, different diseases with a proved or suspected complement-mediated pathophysiology have been considered as candidate for a clinical complement inhibition. At the same time, the growing information on biological changes during eculizumab treatment in PNH have improved our understanding of different steps of the complement system in human diseases, as well as their modulation by current anti-complement treatment. As a result, investigators are currently working on novel strategy of complement inhibition, looking at the second generation of anti-complement agents which hopefully will be able to modulate distinct steps of the complement cascade. Here we review PNH as a disease model, focusing on the observation that led to the development of novel complement modulators; the discussion will be extended to other hemolytic disorders potentially candidate for clinical complement inhibition.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Proteínas do Sistema Complemento/imunologia , Hemoglobinúria Paroxística/tratamento farmacológico , Hemoglobinúria Paroxística/imunologia , Antígenos CD55 , Antígenos CD59 , Ativação do Complemento , Complemento C5/imunologia , Proteínas Inativadoras do Complemento/metabolismo , Eritrócitos/imunologia , Células-Tronco Hematopoéticas/fisiologia , Humanos
6.
PLoS One ; 7(7): e40489, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22792350

RESUMO

Scabies is a parasitic infestation of the skin by the mite Sarcoptes scabiei that causes significant morbidity worldwide, in particular within socially disadvantaged populations. In order to identify mechanisms that enable the scabies mite to evade human immune defenses, we have studied molecules associated with proteolytic systems in the mite, including two novel scabies mite serine protease inhibitors (SMSs) of the serpin superfamily. Immunohistochemical studies revealed that within mite-infected human skin SMSB4 (54 kDa) and SMSB3 (47 kDa) were both localized in the mite gut and feces. Recombinant purified SMSB3 and SMSB4 did not inhibit mite serine and cysteine proteases, but did inhibit mammalian serine proteases, such as chymotrypsin, albeit inefficiently. Detailed functional analysis revealed that both serpins interfered with all three pathways of the human complement system at different stages of their activation. SMSB4 inhibited mostly the initial and progressing steps of the cascades, while SMSB3 showed the strongest effects at the C9 level in the terminal pathway. Additive effects of both serpins were shown at the C9 level in the lectin pathway. Both SMSs were able to interfere with complement factors without protease function. A range of binding assays showed direct binding between SMSB4 and seven complement proteins (C1, properdin, MBL, C4, C3, C6 and C8), while significant binding of SMSB3 occurred exclusively to complement factors without protease function (C4, C3, C8). Direct binding was observed between SMSB4 and the complement proteases C1s and C1r. However no complex formation was observed between either mite serpin and the complement serine proteases C1r, C1s, MASP-1, MASP-2 and MASP-3. No catalytic inhibition by either serpin was observed for any of these enzymes. In summary, the SMSs were acting at several levels mediating overall inhibition of the complement system and thus we propose that they may protect scabies mites from complement-mediated gut damage.


Assuntos
Proteínas de Artrópodes/química , Proteínas Inativadoras do Complemento/química , Sarcoptes scabiei/metabolismo , Inibidores de Serina Proteinase/química , Serpinas/química , Sequência de Aminoácidos , Animais , Proteínas de Artrópodes/metabolismo , Proteínas de Artrópodes/farmacologia , Ativação do Complemento/efeitos dos fármacos , Proteínas Inativadoras do Complemento/metabolismo , Proteínas Inativadoras do Complemento/farmacologia , Proteínas do Sistema Complemento/química , Trato Gastrointestinal/metabolismo , Humanos , Dados de Sequência Molecular , Tempo de Tromboplastina Parcial , Peptídeo Hidrolases/química , Ligação Proteica , Escabiose/imunologia , Escabiose/parasitologia , Análise de Sequência de DNA , Inibidores de Serina Proteinase/metabolismo , Inibidores de Serina Proteinase/farmacologia , Serpinas/metabolismo , Serpinas/farmacologia
7.
J Immunol ; 186(7): 4269-77, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21339361

RESUMO

Exposure of nonself surfaces such as those of biomaterials or transplanted cells and organs to host blood frequently triggers innate immune responses, thereby affecting both their functionality and tolerability. Activation of the alternative pathway of complement plays a decisive role in this unfavorable reaction. Whereas previous studies demonstrated that immobilization of physiological regulators of complement activation (RCA) can attenuate this foreign body-induced activation, simple and efficient approaches for coating artificial surfaces with intact RCA are still missing. The conjugation of small molecular entities that capture RCA with high affinity is an intriguing alternative, as this creates a surface with autoregulatory activity upon exposure to blood. We therefore screened two variable cysteine-constrained phage-displayed peptide libraries for factor H-binding peptides. We discovered three peptide classes that differed with respect to their main target binding areas. Peptides binding to the broad middle region of factor H (domains 5-18) were of particular interest, as they do not interfere with either regulatory or binding activities. One peptide in this group (5C6) was further characterized and showed high factor H-capturing activity while retaining its functional integrity. Most importantly, when 5C6 was coated to a model polystyrene surface and exposed to human lepirudin-anticoagulated plasma, the bound peptide captured factor H and substantially inhibited complement activation by the alternative pathway. Our study therefore provides a promising and novel approach to produce therapeutic materials with enhanced biocompatibility.


Assuntos
Via Alternativa do Complemento/imunologia , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/uso terapêutico , Materiais Biocompatíveis/metabolismo , Clonagem Molecular , Complemento C3b/antagonistas & inibidores , Complemento C3b/metabolismo , Fator H do Complemento/metabolismo , Fator H do Complemento/uso terapêutico , Fator I do Complemento/antagonistas & inibidores , Fator I do Complemento/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Hemólise , Humanos , Biblioteca de Peptídeos , Ligação Proteica/imunologia , Propriedades de Superfície
8.
J Immunol ; 184(12): 7116-24, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20483772

RESUMO

Complement factor B (fB) is a key constituent of the alternative pathway (AP). Its central role in causing inflammation and tissue injury through activation of the AP urges the need for its therapeutic targeting. In the current study, we have screened phage-displayed random peptide libraries against fB and identified a novel cyclic hendecapeptide that inhibits activation of fB and the AP. Structure-activity studies revealed that: 1) the cysteine-constrained structure of the peptide is essential for its activity; 2) Ile5, Arg6, Leu7, and Tyr8 contribute significantly to its inhibitory activity; and 3) retro-inverso modification of the peptide results in loss of its activity. Binding studies performed using surface plasmon resonance suggested that the peptide has two binding sites on fB, which are located on the Ba and Bb fragments. Studies on the mechanism of inhibition revealed that the peptide does not block the interaction of fB with the activated form of C3, thereby suggesting that the peptide inhibits fB activation primarily by inhibiting its cleavage by factor D. The peptide showed a weak effect on preformed C3 and C5 convertases. Like inhibition of fB cleavage, the peptide also inhibited C2 cleavage by activated C1s and activation of the classical as well as lectin pathways. Based on its inhibitory activities, we named the peptide Complin.


Assuntos
Complemento C2/metabolismo , Fator B do Complemento/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Via Alternativa do Complemento/fisiologia , Sequência de Aminoácidos , Proteínas Inativadoras do Complemento/química , Proteínas Inativadoras do Complemento/genética , Ensaio de Imunoadsorção Enzimática , Humanos , Dados de Sequência Molecular , Biblioteca de Peptídeos , Estrutura Quaternária de Proteína , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície
9.
Nat Rev Immunol ; 9(10): 729-40, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19730437

RESUMO

The complement system is important for cellular integrity and tissue homeostasis. Complement activation mediates the removal of microorganisms and the clearance of modified self cells, such as apoptotic cells. Complement regulators control the spontaneously activated complement cascade and any disturbances in this delicate balance can result in damage to tissues and in autoimmune disease. Therefore, insights into the mechanisms of complement regulation are crucial for understanding disease pathology and for enabling the development of diagnostic tools and therapies for complement-associated diseases.


Assuntos
Ativação do Complemento/imunologia , Convertases de Complemento C3-C5/imunologia , Proteínas Inativadoras do Complemento/imunologia , Receptores de Complemento/imunologia , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Ativação do Complemento/genética , Convertases de Complemento C3-C5/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Glomerulonefrite Membranoproliferativa/imunologia , Glomerulonefrite Membranoproliferativa/metabolismo , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/metabolismo , Humanos , Infecções/imunologia , Infecções/metabolismo , Degeneração Macular/imunologia , Degeneração Macular/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Receptores de Complemento/metabolismo
10.
Trends Immunol ; 30(6): 286-92, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19428302

RESUMO

Several studies of human cancers have established that chronic and insidious inflammation promotes the process of carcinogenesis and exacerbates the growth of existing tumors. Conversely, acute inflammation seems to have the opposite effect. Recent discoveries indicate that this dualism in the role of inflammation in cancer is mirrored by the effects of the complement system on this disease process. Previous studies have suggested that complement proteins can contribute to the immune surveillance of malignant tumors. However, a very recent study has indicated that complement proteins can also promote tumor growth. Here, we describe our current understanding of the role of complement in tumor development and progression.


Assuntos
Proteínas Inativadoras do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Neoplasias/imunologia , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Proteínas Inativadoras do Complemento/metabolismo , Proteínas do Sistema Complemento/metabolismo , Progressão da Doença , Humanos , Vigilância Imunológica , Inflamação , Neoplasias/patologia , Estresse Oxidativo , Fatores de Risco , Evasão Tumoral
11.
Ann Neurol ; 65(1): 67-75, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19194881

RESUMO

OBJECTIVE: Complement mediated injury of the neuromuscular junction is considered a primary disease mechanism in human myasthenia gravis and animal models of experimentally acquired myasthenia gravis (EAMG). We utilized active and passive models of EAMG to investigate the efficacy of a novel C5 complement inhibitor rEV576, recombinantly produced protein derived from tick saliva, in moderating disease severity. METHODS: Standardized disease severity assessment, serum complement hemolytic activity, serum cytotoxicity, acetylcholine receptor (AChR) antibody concentration, IgG subclassification, and C9 deposition at the neuromuscular junction were used to assess the effect of complement inhibition on EAMG induced by administration of AChR antibody or immunization with purified AChR. RESULTS: Administration of rEV576 in passive transfer EAMG limited disease severity as evidenced by 100% survival rate and a low disease severity score. In active EAMG, rats with severe and mild EAMG were protected from worsening of disease and had limited weight loss. Serum complement activity (CH(50)) in severe and mild EAMG was reduced to undetectable levels during treatment, and C9 deposition at the neuromuscular junction was reduced. Treatment with rEV576 resulted in reduction of toxicity of serum from severe and mild EAMG rats. Levels of total AChR IgG, and IgG(2a) antibodies were similar, but unexpectedly, the concentration of complement fixing IgG(1) antibodies was lower in a group of rEV576-treated animals, suggesting an effect of rEV576 on cellular immunity. INTERPRETATION: Inhibition of complement significantly reduced weakness in two models of EAMG. C5 inhibition could prove to be of significant therapeutic value in human myasthenia gravis.


Assuntos
Complemento C5/antagonistas & inibidores , Proteínas Inativadoras do Complemento/uso terapêutico , Miastenia Gravis Autoimune Experimental/prevenção & controle , Animais , Anticorpos/efeitos adversos , Proteínas de Artrópodes , Linhagem Celular Tumoral , Complemento C9/metabolismo , Proteínas Inativadoras do Complemento/imunologia , Proteínas Inativadoras do Complemento/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Imunoglobulina G/sangue , Imunoglobulina G/classificação , Proteínas de Insetos/imunologia , Proteínas de Insetos/uso terapêutico , Força Muscular/efeitos dos fármacos , Miastenia Gravis Autoimune Experimental/etiologia , Miastenia Gravis Autoimune Experimental/imunologia , Miastenia Gravis Autoimune Experimental/patologia , Junção Neuromuscular/efeitos dos fármacos , Junção Neuromuscular/metabolismo , Ratos , Ratos Endogâmicos Lew , Receptores Colinérgicos/imunologia , Rabdomiossarcoma/metabolismo , Rabdomiossarcoma/patologia , Proteínas e Peptídeos Salivares/sangue , Proteínas e Peptídeos Salivares/imunologia , Proteínas e Peptídeos Salivares/uso terapêutico , Índice de Gravidade de Doença , Fatores de Tempo , Redução de Peso/efeitos dos fármacos
12.
J Biol Chem ; 284(1): 505-514, 2009 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-18990693

RESUMO

Rhesus rhadinovirus (RRV) is currently the closest known, fully sequenced homolog of human Kaposi sarcoma-associated herpesvirus. Both these viruses encode complement inhibitors as follows: Kaposi sarcoma-associated herpesvirus-complement control protein (KCP) and RRV-complement control protein (RCP). Previously we characterized in detail the functional properties of KCP as a complement inhibitor. Here, we performed comparative analyses for two variants of RCP protein, encoded by RRV strains H26-95 and 17577. Both RCP variants and KCP inhibited human and rhesus complement when tested in hemolytic assays measuring all steps of activation via the classical and the alternative pathway. RCP variants from both RRV strains supported C3b and C4b degradation by factor I and decay acceleration of the classical C3 convertase, similar to KCP. Additionally, the 17577 RCP variant accelerated decay of the alternative C3 convertase, which was not seen for KCP. In contrast to KCP, RCP showed no affinity to heparin and is the first described complement inhibitor in which the binding site for C3b/C4b does not interact with heparin. Molecular modeling shows a structural disruption in the region of RCP that corresponds to the KCP-heparin-binding site. This makes RRV a superior model for future in vivo investigations of complement evasion, as RCP does not play a supportive role in viral attachment as KCP does.


Assuntos
Proteínas Inativadoras do Complemento/metabolismo , Proteínas do Sistema Complemento/metabolismo , Herpesvirus Saimiriíneo 2/metabolismo , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Proteínas Virais/metabolismo , Animais , Sítios de Ligação , Células CHO , Proteínas Inativadoras do Complemento/química , Proteínas Inativadoras do Complemento/genética , Proteínas do Sistema Complemento/química , Proteínas do Sistema Complemento/genética , Cricetinae , Cricetulus , Heparina/química , Heparina/metabolismo , Herpesvirus Saimiriíneo 2/química , Herpesvirus Saimiriíneo 2/genética , Herpesvirus Humano 8/química , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Macaca mulatta , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Especificidade da Espécie , Relação Estrutura-Atividade , Proteínas Virais/química , Proteínas Virais/genética , Ligação Viral
13.
Clin Exp Immunol ; 155(2): 239-48, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19040611

RESUMO

Multi-drug resistance (MDR) is a major obstacle in cancer chemotherapy. There are contrasting data on a possible correlation between the level of expression of the drug transporter P-glycoprotein (P-gp) and susceptibility to complement-dependent cytotoxicity (CDC). We therefore investigated the sensitivity of human ovarian carcinoma cells and their P-gp expressing MDR variants to complement. Chemoselected P-gp expressing MDR cells showed increased resistance to CDC associated with overexpression of membrane-bound complement regulatory proteins (mCRP) and increased release of the soluble inhibitors C1 inhibitor and factor I. MDR1 gene transfection alone did not alter the susceptibility of P-gp expressing A2780-MDR and SKOV3-MDR cells to CDC. However, subsequent vincristine treatment conferred an even higher resistance to complement to these cells, again associated with increased expression of mCRP. Blocking the function of P-gp with verapamil, cyclosporine A or the anti-P-gp-antibody MRK16 had no impact on their complement resistance, whereas blocking of mCRP enhanced their susceptibility to complement. These results suggest that enhanced resistance of chemoselected MDR ovarian carcinoma cells to CDC is not conferred by P-gp, but is due at least partly to overexpression of mCRP, probably induced by treatment with the chemotherapeutic agents.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Proteínas do Sistema Complemento/imunologia , Resistencia a Medicamentos Antineoplásicos/imunologia , Proteínas de Neoplasias/metabolismo , Neoplasias Ovarianas/imunologia , Subfamília B de Transportador de Cassetes de Ligação de ATP , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Antígenos CD55/metabolismo , Antígenos CD59/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Citotoxicidade Imunológica , Resistência a Múltiplos Medicamentos/imunologia , Feminino , Humanos , Proteína Cofatora de Membrana/metabolismo , Neoplasias Ovarianas/metabolismo , Transdução Genética , Células Tumorais Cultivadas
14.
Infect Immun ; 76(2): 750-8, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18070905

RESUMO

As a central component of innate immunity, complement activation is a critical mechanism of containment and clearance of microbial pathogens in advance of the development of acquired immunity. Several pathogens restrict complement activation through the acquisition of host proteins that regulate complement activation or through the production of their own complement regulatory molecules (M. K. Liszewski, M. K. Leung, R. Hauhart, R. M. Buller, P. Bertram, X. Wang, A. M. Rosengard, G. J. Kotwal, and J. P. Atkinson, J. Immunol. 176:3725-3734, 2006; J. Lubinski, L. Wang, D. Mastellos, A. Sahu, J. D. Lambris, and H. M. Friedman, J. Exp. Med. 190:1637-1646, 1999). The infectious stage of the protozoan parasite Trypanosoma cruzi produces a surface-anchored complement regulatory protein (CRP) that functions to inhibit alternative and classical pathway complement activation (K. A. Norris, B. Bradt, N. R. Cooper, and M. So, J. Immunol. 147:2240-2247, 1991). This study addresses the genomic complexity of the T. cruzi CRP and its relationship to the T. cruzi supergene family comprising active trans-sialidase (TS) and TS-like proteins. The TS superfamily consists of several functionally distinct subfamilies that share a characteristic sialidase domain at their amino termini. These TS families include active TS, adhesions, CRPs, and proteins of unknown functions (G. A. Cross and G. B. Takle, Annu. Rev. Microbiol. 47:385-411, 1993). A sequence comparison search of GenBank using BLASTP revealed several full-length paralogs of CRP. These proteins share significant homology at their amino termini and a strong spatial conservation of cysteine residues. Alternative pathway complement regulation was confirmed for CRP paralogs with 58% (low) and 83% (high) identity to AAB49414. CRPs are functionally similar to the microbial and mammalian proteins that regulate complement activation. Sequence alignment of mammalian complement control proteins to CRP showed that these sequences are distinct, supporting a convergent evolutionary pathway. Finally, we show that a clonal line of T. cruzi expresses multiple unique copies of CRP that are differentially recognized by patient sera.


Assuntos
Proteínas Inativadoras do Complemento/genética , Heterogeneidade Genética , Glicoproteínas de Membrana/genética , Proteínas de Protozoários/genética , Trypanosoma cruzi/genética , Sequência de Aminoácidos , Animais , Proteínas Inativadoras do Complemento/metabolismo , Proteínas do Sistema Complemento , Sequência Conservada , Glicoproteínas/genética , Humanos , Glicoproteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Neuraminidase/genética , Proteínas de Protozoários/metabolismo , Homologia de Sequência de Aminoácidos , Trypanosoma cruzi/metabolismo
15.
Histol Histopathol ; 22(7): 781-90, 2007 07.
Artigo em Inglês | MEDLINE | ID: mdl-17455152

RESUMO

The limited ability of articular cartilage to recover from injury, remains an unsolved clinical challenge in orthopaedic surgery. Persistent injury of the articular surface can lead to the development of posttraumatic osteoarthritis. The local inflammatory response contributes to the pathogenesis of osteoarthritis by inducing chondrocyte apoptosis and the de-regulation of chondrocyte matrix remodelling. The role of the complement system in contributing to secondary inflammation-mediated cartilage degradation represents a newer field of investigation. The purpose of this review article is to summarize the known complement-mediated actions in cartilage homeostasis and injury. This article focuses on the known effects of complement on secondary chondrocyte apoptosis, and the interplay of the complement system with pro-inflammatory cytokines. Pharmacological therapies related to complement inhibition will be discussed as they potentially represent a new avenue for attenuating the effect of the complement system on cartilage repair.


Assuntos
Cartilagem Articular/lesões , Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Ativação do Complemento , Proteínas do Sistema Complemento/metabolismo , Osteoartrite/metabolismo , Animais , Apoptose , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/imunologia , Cartilagem Articular/fisiopatologia , Condrócitos/efeitos dos fármacos , Condrócitos/imunologia , Ativação do Complemento/efeitos dos fármacos , Inativadores do Complemento/farmacologia , Inativadores do Complemento/uso terapêutico , Proteínas Inativadoras do Complemento/metabolismo , Citocinas/metabolismo , Matriz Extracelular/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Articulações/imunologia , Articulações/metabolismo , Osteoartrite/tratamento farmacológico , Osteoartrite/imunologia , Osteoartrite/fisiopatologia , Osteogênese/imunologia , Líquido Sinovial/imunologia , Líquido Sinovial/metabolismo
16.
Xenotransplantation ; 14(1): 34-47, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17214703

RESUMO

BACKGROUND: Hyperacute rejection (HAR) and early graft failure (EGF) have been described in a minority of pig-to-baboon heart transplants using organs transgenic for human complement regulatory proteins (hCRP). Here we investigate the role of coagulation cascade activation in the pathogenesis of HAR and EGF in a consecutive series where a high incidence of these outcomes was observed. METHODS: Twenty-eight naïve wild-caught Papio anubis baboons received heterotopic heart transplants from pigs transgenic for hDAF (n = 23) or hMCP (n = 5). Immunosuppression consisted of cyclosporine A, cyclophosphamide and MMF (n = 18) or anti-CD154 mAb (IDEC-131) and ATG (n = 10). Eleven received anti-Gal carbohydrates (GAS914, n = 8, or NEX1285, n = 3), of which four also underwent extracorporeal immunoadsorption (EIA), and 12 also received pharmacologic complement inhibitors (C1 INH, n = 9, or APT070, n = 3). RESULTS: Excluding one technical failure, 14 of 27 transplants (11 hDAF, 3 hMCP) exhibited either HAR (n = 10) or EGF (n = 4). Surprisingly, neither complement inhibition (with C1 INH or APT070) nor anti-Gal antibody depletion with GAS914, NEX1285, or additional EIA consistently prevented HAR or EGF despite low or undetectable complement deposition. Strikingly, most grafts with HAR/EGF exhibited prominent fibrinogen and platelet deposition associated with systemic coagulation cascade activation, consistent with non-physiologic intravascular coagulation, in many instances despite little evidence for antibody-mediated complement activation. CONCLUSION: We conclude that dysregulated coagulation correlates closely with and probably causes primary failure of pig hearts transgenic for hCRP. These data support efforts to define effective strategies to prevent dysregulated coagulation in pig organ xenografts.


Assuntos
Proteínas Inativadoras do Complemento/metabolismo , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Transplante de Coração/imunologia , Coração/fisiopatologia , Miocárdio/imunologia , Miocárdio/metabolismo , Transplante Heterólogo/imunologia , Doença Aguda , Animais , Anticorpos/imunologia , Biópsia , Coagulação Sanguínea , Proteína Inibidora do Complemento C1/metabolismo , Complemento C3a/metabolismo , Proteínas Inativadoras do Complemento/genética , Regulação da Expressão Gênica , Rejeição de Enxerto/genética , Transplante de Coração/efeitos adversos , Humanos , Imuno-Histoquímica , Suínos , Fatores de Tempo , Titulometria
17.
Immunology ; 118(3): 333-42, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16827894

RESUMO

The complement regulatory proteins CD55 and CD59 are expressed on the plasma membrane of human spermatozoa, whereas CD46 is only on the inner acrosomal membrane (IAM) which becomes surfaced exposed after the acrosome reaction when sperm assume fertilisation-competence. CD55 & CD59, two glycosylphosphatidylinositol (GPI)-anchored proteins, have been detected previously in some studies also in the acrosomal region of chemically fixed spermatozoa but never demonstrated at this site on unfixed spermatozoa. Dual labelling immunofluorescence and confocal microscopy on fresh unfixed spermatozoa, with minimal subsequent time to fixation, has shown CD55 to be markedly expressed on the IAM, more than on the plasma membrane. However, unlike for CD46, CD55 displayed patchy staining over the acrosome, with some variation between individual spermatozoa. All IAM-associated CD55 was localised within GM1-containing lipid rafts. CD59 was expressed also on the IAM, but in a pronounced granular pattern with more variation observed from one spermatozoa to another. Both CD55 & CD59 were released from the IAM by PI-PLC, demonstrating them to be GPI-anchored. Analysis of acrosome-reacted spermatozoal CD55 by Western blotting revealed a novel single 55 kDa protein lacking significant oligosaccharides susceptible to glycosidases. Antibody-induced membrane rafting and release of CD55 & CD59 in vitro may have influenced previous results. Significant coexpression of CD55 & CD46 on the IAM suggests some functional cooperation at this site.


Assuntos
Acrossomo/imunologia , Antígenos CD/metabolismo , Proteínas Inativadoras do Complemento/metabolismo , Western Blotting , Antígenos CD55/metabolismo , Antígenos CD59/metabolismo , Membrana Celular/imunologia , Humanos , Masculino , Proteína Cofatora de Membrana/metabolismo , Microscopia de Fluorescência , Fosfatidilinositol Diacilglicerol-Liase/imunologia , Fosfoinositídeo Fosfolipase C , Espermatozoides/imunologia , Fatores de Tempo , Preservação de Tecido
18.
Cancer Res ; 66(12): 6258-63, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16778201

RESUMO

Neoplasms have developed strategies to protect themselves against the complement-mediated host immunity. Invasion- and metastasis-promoting membrane type-1 (MT1) matrix metalloproteinase (MMP) is strongly associated with many metastatic cancer types. The relative importance of the individual functions of MT1-MMP in metastasis was, however, unknown. We have now determined that the expression of murine MT1-MMP in murine melanoma B16F1 cells strongly increased the number of metastatic loci in the lungs of syngeneic C57BL/6 mice. In contrast, MT1-MMP did not affect the number of metastatic loci in complement-deficient C57BL/6-C3-/- mice. Our results indicated, for the first time, that the anticomplement activity of MT1-MMP played a significant role in promoting metastasis in vivo and determined the relative importance of the anticomplement activity in the total metastatic effect of this multifunctional proteolytic enzyme. We believe that our results shed additional light on the functions of MT1-MMP in cancer and clearly make this protease a promising drug target in metastatic malignancies.


Assuntos
Complemento C3/imunologia , Proteínas Inativadoras do Complemento/imunologia , Metaloproteinases da Matriz/imunologia , Neoplasias Experimentais/enzimologia , Neoplasias Experimentais/imunologia , Animais , Complemento C3/deficiência , Complemento C3/genética , Proteínas Inativadoras do Complemento/genética , Proteínas Inativadoras do Complemento/metabolismo , Fibrossarcoma/enzimologia , Fibrossarcoma/genética , Fibrossarcoma/imunologia , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Metaloproteinase 14 da Matriz , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Metaloproteinases da Matriz Associadas à Membrana , Melanoma Experimental/enzimologia , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Melanoma Experimental/secundário , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Transfecção
19.
J Natl Cancer Inst ; 97(17): 1287-96, 2005 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-16145049

RESUMO

BACKGROUND: Clusterin is a cytoprotective chaperone protein that promotes cell survival and confers broad-spectrum treatment resistance. OGX-011 is a 2'-methoxyethyl modified phosphorothioate antisense oligonucleotide that is complementary to clusterin mRNA and has been reported to inhibit clusterin expression and enhance drug efficacy in xenograft models. The primary objective of this clinical study was to determine a biologically effective dose of OGX-011 that would inhibit clusterin expression in human cancer. METHODS: Subjects (n = 25) with localized prostate cancer with high-risk features who were candidates for prostatectomy were treated with OGX-011 by 2-hour intravenous infusion on days 1, 3, and 5 and then weekly from days 8-29 combined with androgen blockade starting on day 1; prostatectomy was performed on days 30-36. Six different doses were tested, from 40 to 640 mg. OGX-011 plasma and prostate tissue concentrations were measured by an enzyme-linked immunosorbent assay method, and the pharmacokinetics of OGX-011 were determined from these data. Prostate cancer tissue, lymph nodes, and serial samples of peripheral blood mononuclear cells were assessed for clusterin expression using quantitative real-time polymerase chain reaction and immunohistochemistry. All statistical tests were two-sided. RESULTS: Only grade 1 and 2 toxicities were observed. The plasma half-life of OGX-011 was approximately 2-3 hours, and the area under the concentration versus time curve and CMAX (peak plasma concentration) increased proportionally with dose (Ptrend < .001). OGX-011 in prostate tissue increased with dose (Ptrend < .001). Dose-dependent decreases in prostate cancer and lymph node clusterin expression were observed by polymerase chain reaction of greater than 90% (Ptrend = .008 and Ptrend < .001, respectively) and by immunohistochemistry (Ptrend < .001 and Ptrend = .01, respectively). CONCLUSIONS: OGX-011 is well tolerated and reduces clusterin expression in primary prostate tumors. The optimal biologic dose for OGX-011 at the schedule used is 640 mg.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Proteínas Inativadoras do Complemento/antagonistas & inibidores , Glicoproteínas/antagonistas & inibidores , Chaperonas Moleculares/antagonistas & inibidores , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Tionucleotídeos/administração & dosagem , Tionucleotídeos/farmacologia , Idoso , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Quimioterapia Adjuvante , Clusterina , Proteínas Inativadoras do Complemento/metabolismo , Relação Dose-Resposta a Droga , Esquema de Medicação , Ensaio de Imunoadsorção Enzimática , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicoproteínas/metabolismo , Meia-Vida , Humanos , Imuno-Histoquímica , Hibridização In Situ , Infusões Intravenosas , Linfonodos/efeitos dos fármacos , Linfonodos/metabolismo , Masculino , Pessoa de Meia-Idade , Chaperonas Moleculares/metabolismo , Terapia Neoadjuvante , Estadiamento de Neoplasias , Oligonucleotídeos Antissenso/efeitos adversos , Reação em Cadeia da Polimerase , Prostatectomia , Neoplasias da Próstata/sangue , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Neoplasias da Próstata/cirurgia , Tionucleotídeos/farmacocinética
20.
J Exp Med ; 201(12): 1937-48, 2005 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-15967823

RESUMO

After cell death, via apoptosis or necrosis, the uptake of dead cells by neighboring cells or phagocytes prevents the release of intracellular content. An array of molecules, including initiation molecules of the complement system, are involved in marking dead cells for uptake. After binding of these molecules, complement activation takes place, which when uncontrolled might result in a proinflammatory state. In the current study we demonstrate that complement inhibitor, C4b-binding protein (C4BP), binds strongly to necrotic cells, irrespective of the cell type used or the method of induction. After binding of the C4BP-protein S (PS) complex to necrotic cells via PS-phosphatidylserine and C4BP-DNA interactions, C4BP-PS inhibits complement activation on these cells. C4BP binds DNA via a patch of positively charged amino acids, mainly on the second complement control domain of the C4BP alpha-chain (affinity constant: 190 nM). Furthermore, C4BP limits DNA release from necrotic cells and inhibits DNA-mediated complement activation in solution. The C4BP-necrotic cell interaction also occurs in vivo as necrotic areas of arteriosclerotic plaques and of various cancers stain strongly positive for C4BP. This study describes a novel mechanism in which C4BP limits the inflammatory potential of necrotic cells.


Assuntos
Ativação do Complemento/fisiologia , Proteínas Inativadoras do Complemento/metabolismo , DNA/metabolismo , Antígenos de Histocompatibilidade/metabolismo , Necrose/metabolismo , Proteína S/metabolismo , Anticorpos Monoclonais/metabolismo , Linhagem Celular , Proteína de Ligação ao Complemento C4b , Ensaio de Desvio de Mobilidade Eletroforética , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Fosfatidilserinas/metabolismo , Ligação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA