Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Haematologica ; 103(2): 351-360, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29146705

RESUMO

Development of neutralizing antibodies against therapeutic Factor VIII (FVIII) is the most serious complication of the treatment of hemophilia A. There is growing evidence to show the multifactorial origin of the anti-FVIII immune response, combining both genetic and environmental factors. While a role for the complement system on innate as well as adaptive immunity has been documented, the implication of complement activation on the onset of the anti-FVIII immune response is unknown. Here, using in vitro assays for FVIII endocytosis by human monocyte-derived dendritic cells and presentation to T cells, as well as in vivo complement depletion in FVIII-deficient mice, we show a novel role for complement C3 in enhancing the immune response against therapeutic FVIII. In vitro, complement C3 and its cleavage product C3b enhanced FVIII endocytosis by dendritic cells and presentation to a FVIII-specific CD4+ T-cell hybridoma. The C1 domain of FVIII had previously been shown to play an important role in FVIII endocytosis, and alanine substitutions of the K2092, F2093 and R2090 C1 residues drastically reduce FVIII uptake in vitro Interestingly, complement activation rescued the endocytosis of the FVIII C1 domain triple mutant. In a mouse model of severe hemophilia A, transient complement C3 depletion by humanized cobra venom factor, which does not generate anaphylatoxin C5a, significantly reduced the primary anti-FVIII immune response, but did not affect anti-FVIII recall immune responses. Taken together, our results suggest an important adjuvant role for the complement cascade in the initiation of the immune response to therapeutic FVIII.


Assuntos
Anticorpos Neutralizantes/imunologia , Complemento C3/farmacologia , Fator VIII/imunologia , Animais , Apresentação de Antígeno/imunologia , Ativação do Complemento , Células Dendríticas/fisiologia , Endocitose/efeitos dos fármacos , Humanos , Imunidade/efeitos dos fármacos , Camundongos
2.
EMBO J ; 28(16): 2469-78, 2009 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-19574954

RESUMO

Immune protection by the complement system critically depends on assembly of C3 convertases on the surface of pathogens and altered host cells. These short-lived protease complexes are formed through pro-convertases, which for the alternative pathway consist of the complement component C3b and the pro-enzyme factor B (FB). Here, we present the crystal structure at 2.2-A resolution, small-angle X-ray scattering and electron microscopy (EM) data of the pro-convertase formed by human FB and cobra venom factor (CVF), a potent homologue of C3b that generates more stable convertases. FB is loaded onto CVF through its pro-peptide Ba segment by specific contacts, which explain the specificity for the homologous C3b over the native C3 and inactive products iC3b and C3c. The protease segment Bb binds the carboxy terminus of CVF through the metal-ion dependent adhesion site of the Von Willebrand factor A-type domain. A possible dynamic equilibrium between a 'loading' and 'activation' state of the pro-convertase may explain the observed difference between the crystal structure of CVFB and the EM structure of C3bB. These insights into formation of convertases provide a basis for further development of complement therapeutics.


Assuntos
Convertases de Complemento C3-C5/metabolismo , Fator B do Complemento/química , Fator B do Complemento/metabolismo , Venenos Elapídicos/química , Venenos Elapídicos/metabolismo , Elapidae/metabolismo , Animais , Linhagem Celular , Convertases de Complemento C3-C5/química , Fator B do Complemento/genética , Cristalografia por Raios X , Venenos Elapídicos/genética , Venenos Elapídicos/isolamento & purificação , Ativação Enzimática , Expressão Gênica , Humanos , Modelos Moleculares , Mutação , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína
3.
Blood ; 114(26): 5322-30, 2009 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-19805620

RESUMO

Growing evidence indicates antibody-dependent cellular cytotoxicity (ADCC) contributes to the clinical response to monoclonal antibody (mAb) therapy of lymphoma. Recent in vitro analysis suggests C3b can inhibit mAb-induced natural killer (NK)-cell activation and ADCC. Further studies were conducted to assess the effect of C3 depletion on mAb-induced NK activation and therapy of lymphoma. Normal human serum inhibited the ability of rituximab-coated lymphoma cells to activate NK cells as previously reported. Serum did not inhibit NK-cell activation when it was preincubated with cobra venom factor (CVF) to deplete C3. Similar results were found when transudative pleural fluid or nonmalignant ascites was used as surrogates for extravascular fluid, suggesting the inhibitory effect of complement may be present in the extravascular compartment, in which many malignant lymphocytes reside. In vivo, C3 was depleted before mAb treatment in a syngeneic murine model of lymphoma. Survival of lymphoma-bearing mice after treatment with CVF plus mAb and with a human C3 derivative with CVF-like functions (HC3-1496) plus mAb was both superior to that of mAb alone. These studies show that complement depletion enhances NK-cell activation induced by rituximab-coated target cells and improves the efficacy of mAb therapy in a murine lymphoma model.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Complemento C3/deficiência , Linfoma/tratamento farmacológico , Linfoma/imunologia , Animais , Anticorpos Monoclonais Murinos , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Ativação Linfocitária/efeitos dos fármacos , Masculino , Camundongos , Rituximab
4.
Adv Exp Med Biol ; 703: 151-62, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20711713

RESUMO

The effect of complement depletion with humanized cobra venom factor (CVF) on retinal lesion development/neovascularization was determined in a mouse model of wet age-related macular degeneration (AMD). Mice were treated with the humanized CVF protein HC3-1496 prior to, and once daily for 28 days after laser coagulation surgery of the retina. CVF transgenic mice exhibiting permanently low levels of serum complement activity and PBS-treated mice served as positive and negative controls, respectively. Fluorescein isothiocyanate (FITC)-dextran funduscopy after laser surgery indicated the presence of lesions in all mice that underwent laser surgery. In HC3-1496-treated mice as well as CVF transgenic mice smaller lesions were seen after 8 days. Measurement of lesion sizes by histopathological examination of eyes after 28 days revealed a significant reduction of lesion area and volume in both HC3-1496-treated animals and CVF transgenic animals compared to PBS-treated control animals. Systemic complement depletion with a complement depletor, such as the humanized CVF protein HC3-1496, represents a promising therapeutic concept for patients with wet AMD.


Assuntos
Inativadores do Complemento/farmacologia , Venenos Elapídicos/farmacologia , Degeneração Macular Exsudativa/tratamento farmacológico , Animais , Complemento C3/genética , Modelos Animais de Doenças , Venenos Elapídicos/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Recombinantes de Fusão/genética , Degeneração Macular Exsudativa/imunologia , Degeneração Macular Exsudativa/patologia , Degeneração Macular Exsudativa/cirurgia
5.
Toxicon ; 184: 68-77, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32526239

RESUMO

Cobra venom factor (CVF) is the complement-activating protein in cobra venom. CVF is a structural and functional analog of complement component C3. CVF, like C3b, forms a convertase with factor B. This bimolecular complex CVF, Bb is an enzyme that cleaves C3 and C5. However, CVF, Bb exhibits significantly different functional properties from C3b,Bb. Whereas both, CVF, Bb and C3b, Bb exhibit spontaneous decay-dissociation into the respective subunits, thereby eliminating the enzymatic activity, the CVF, Bb convertase is physico-chemically far more stable, decaying with a half-life that is more than two orders of magnitude slower than that of C3b,Bb. In addition, CVF, Bb is completely resistant to inactivation by Factors H and I. These two properties of CVF, Bb allow continuous activation of C3 and C5, and complement depletion in serum. In order to understand the structural basis for the physico-chemical stability of CVF,Bb, we have created recombinant hybrid proteins of CVF and human C3, based on structural differences between CVF and human C3b in the C-terminal C345C domain. Here we describe three human C3/CVF hybrid proteins which differ in only one, two, or five amino acid residues from earlier described hybrid proteins. In all three cases, the hybrid proteins containing CVF residues form more stable convertases, and exhibit stronger complement-depletion activity than hybrid proteins with human C3 residues. Three bonds between CVF residues and Factor Bb residues could be identified by crystallographic modeling that contribute to the greater stability of the convertases.


Assuntos
Convertases de Complemento C3-C5/química , Fator B do Complemento/química , Venenos Elapídicos/química , Animais , Complemento C3 , Fator H do Complemento , Humanos , Proteínas Recombinantes de Fusão
6.
Dev Comp Immunol ; 33(1): 105-16, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18760301

RESUMO

Cobra venom factor (CVF) is a structural and functional analog of complement C3 isolated from cobra venom. Both CVF and C3b can bind factor B and subsequently form the bimolecular C3/C5 convertases CVF,Bb or C3b,Bb, respectively. The two homologous enzymes exhibit several differences of which the difference in physico-chemical stability is most important, allowing continuous activation of C3 and C5 by CVF,Bb, leading to serum complement depletion. Here we describe the detailed functional properties of two hybrid proteins in which the 113 or 315 C-terminal residues of C3 were replaced with corresponding CVF sequences. Both hybrid proteins formed stable convertases that exhibited C3-cleaving activity, although at different rates. Neither convertase cleaved C5. Both convertases showed partial resistance to inactivation by factors H and I, allowing them to deplete complement in human serum. These data demonstrate that functionally important structural differences between CVF and C3 are located in the very C-terminal region of both homologous proteins, and that small substitutions in human C3 with homologous CVF sequence result in C3 derivatives with CVF-like functions. Such hybrid proteins are important tools to study the structure/function relationships in both C3 and CVF, and these "humanized CVF" proteins may become reagents for therapeutic complement depletion.


Assuntos
Complemento C3/química , Venenos Elapídicos/química , Animais , Clonagem Molecular , Complemento C3/genética , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/genética , Fator H do Complemento/química , Venenos Elapídicos/genética , Fibrinogênio/química , Hemólise , Humanos , Modelos Moleculares , Ligação Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Ovinos
7.
Toxicon ; 167: 106-116, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31207349

RESUMO

Cobra venom factor (CVF) is the complement-activating protein in cobra venom. CVF is a structural and functional analog of complement component C3. CVF, like C3b, forms a convertase with factor B. This bimolecular complex CVF,Bb is an enzyme that cleaves C3 and C5. However, CVF,Bb exhibits significantly different functional properties from C3b,Bb. The CVF,Bb convertase is physico-chemically very stable, and completely resistant to an activation by Factors H and I. These two properties, in contrast to C3b,Bb, allow continuous activation of C3 and C5, and complement depletion in serum. In order to understand the structural basis for the functional differences between CVF and C3, we have created several hybrid proteins of CVF and human C3. Here we report that replacing the C-terminal 168 amino acid residues of human C3 with the corresponding residues from CVF results in a hybrid protein (HC3-1496) which is essentially a human C3 derivative exhibiting the functional properties of CVF. This result demonstrates that the important structures for the CVF-specific functions reside within the C-terminal 168 amino acid residues of CVF. We further demonstrate that reverting the 46 C-terminal CVF residues of HC3-1496 to human C3 sequence results in a hybrid protein (HC3-1496/1617) that exhibits a physico-chemically unstable convertase with only residual complement depleting activity. This result demonstrates that most, but not all, structural requirements for CVF activity reside within the 46 C-terminal amino acid residues. We also investigated the potential role of position 1633, which is an acidic residue in human C3 (glutamic acid) but a basic amino acid residue (histidine) in CVF. However, the charge at position 1633 appears to be of no functional relevance. Exchanging the neutral amino acids present in CVF at positions 1499 and 1501 with the two charged amino acids at these positions in human C3 (aspartic acid and lysine) resulted in a hybrid protein that exhibited significantly slower convertase formation although both binding to Factor B and C3 cleavage was not affected, demonstrating that the charged amino acid residues at these two positions interfere with the formation of the convertase. In conclusion, our work demonstrates that hybrid proteins of human C3 and CVF present valuable tools to identify functionally important amino acid residues in CVF.


Assuntos
Complemento C3/química , Venenos Elapídicos/química , Sequência de Aminoácidos , Humanos , Proteínas Recombinantes de Fusão/química , Análise de Sequência de Proteína , Relação Estrutura-Atividade
8.
Adv Exp Med Biol ; 632: 293-307, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19025130

RESUMO

To obtain proteins with the complement-depleting activity of Cobra Venom Factor (CVF), but with less immunogenicity, we have prepared human C3/CVF hybrid proteins, in which the C-terminus of the alpha-chain of human C3 is exchanged with homologous regions of the C-terminus of the beta-chain of CVF. We show that these hybrid proteins are able to deplete complement, both in vitro and in vivo. One hybrid protein, HC3-1496, is shown to be effective in reducing complement-mediated damage in two disease models in mice, collagen-induced arthritis and myocardial ischemia/reperfusion injury. Human C3/CVF hybrid proteins represent a novel class ofbiologicals as potential therapeutic agents in many diseases where complement is involved in the pathogenesis.


Assuntos
Complemento C3/química , Complemento C3/metabolismo , Complemento C3/uso terapêutico , Proteínas do Sistema Complemento/metabolismo , Animais , Artrite Experimental/tratamento farmacológico , Complemento C3/genética , Venenos Elapídicos/química , Venenos Elapídicos/metabolismo , Humanos , Camundongos , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Engenharia de Proteínas , Estrutura Secundária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico
9.
Mol Immunol ; 97: 1-7, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29525557

RESUMO

Cobra venom factor (CVF) is the complement-activating protein in cobra venom. Humanized CVF (hCVF) is a human C3 derivative where the C-terminal 168 amino acid residues were replaced with the homologous sequence from CVF. hCVF has been shown in multiple models of disease with complement pathology to be a promising therapeutic agent, with no observed adverse effects. Here we describe the antibody response to hCVF in two different strains of mice. hCVF was able to repeatedly decomplement the mice after four injections in weekly intervals, demonstrating the absence of a neutralizing antibody response. In contrast, natural CVF caused decomplementation in all mice only after the first administration. After two additional administrations of natural CVF, decomplementation was inconsistent and varied tremendously from mouse to mouse. After the fourth administration, natural CVF was essentially unable to deplete complement, consistent with the known generation of a neutralizing antibody response. We also analyzed the IgG antibody response to hCVF. There was great variation, with approximately one quarter of the mice exhibiting non-detectable levels of anti-hCVF IgG, and another quarter very low levels. The levels of anti-hCVF IgG did not correlate with the levels of remaining C3. The anti-hCVF antibodies cross-reacted with natural CVF, recombinant CVF, and human C3. Whereas overall the level of anti-hCVF IgG cross-reacting with human C3 was lower compared to rCVF or nCVF, mice with higher levels of anti-hCVF IgG exhibited higher binding to CVF and human C3, excluding the possibility that higher antibody levels reflect preferential immunogenicity of CVF-specific or human C3-specific epitopes.


Assuntos
Anticorpos Neutralizantes/metabolismo , Formação de Anticorpos , Venenos Elapídicos/imunologia , Proteínas Recombinantes de Fusão/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Drosophila melanogaster , Venenos Elapídicos/química , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão/química
10.
Open Pain J ; 9: 26-37, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28154610

RESUMO

Certain types of pain are major unmet medical needs that affect more than 8 percent of the population. Neuropathic pain can be caused by many pathogenic processes including injury, autoimmune disease, neurological disease, endocrine dysfunction, infection, toxin exposure, and substance abuse and is frequently resistant to available pain therapies. The same can be said of postsurgical pain, which can arise from uncontrolled inflammation around the wound site. The complement system is part of the innate immune system and can both initiate and sustain acute and chronic inflammatory pain. Here we review the complement system and original investigations that identify potential drug targets within this system. Drugs that act to inhibit the complement system could fill major gaps in our current standard of care for neuropathic pain states.

11.
Mol Immunol ; 41(2-3): 191-9, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15159065

RESUMO

Cobra venom factor (CVF) is the complement-activating protein from cobra venom. CVF is a three-chain protein that functionally resembles C3b, the activated form of complement component C3. Like C3b, CVF forms a C3/C5 convertase with factor B in the presence of factor D and Mg(2+). Although CVF exhibits functional activity of C3b, it structurally resembles the C3b degradation product C3c, which is not able to form a C3/C5 convertase. CVF has become an important research tool to decomplement laboratory animals in order to study the role of complement in host defense, immune response, and pathogenesis of disease. As the Asian cobras of the Naja species are on the list of endangered species, cobra venom as the source for CVF has become increasingly difficult to obtain. Methods have been developed to recombinantly produce active forms of CVF. This manuscript reviews the production of recombinant pro-CVF using both prokaryotic and eukaryotic expression systems. The recombinant production of pro-CVF in two insect cell expression systems (baculovirus-infected Sf9 Spodoptera frugiperda cells, stably transfected S2 Drosophila melanogaster cells) generates three forms of pro-CVF: single-chain pro-CVF resembling pro-C3, a two-chain form of pro-CVF resembling C3, and another two-chain form of pro-CVF resembling C3b. All three forms of pro-CVF exhibit functional activity of mature, natural CVF. Recombinant pro-CVF supports the activation of factor B in the presence of factor D and Mg(2+), forms a bimolecular convertase pro-CVF,Bb that exhibits cleaving activity for both C3 and C5, and depletes the serum complement activity. The activity of pro-CVF and the resulting C3/C5 convertase is indistinguishable from CVF and the CVF,Bb convertase. Recombinant production of functionally active forms of pro-CVF ensures the availability of an important research reagent for future research involving complement depletion. The experimental systems to recombinantly produce active forms of CVF will also be invaluable for studies to delineate the structure/function relationship of CVF and its differences from C3, and to generate human C3 derivatives with CVF-like function ("humanized CVF") for therapeutic complement depletion.


Assuntos
Clonagem Molecular/métodos , Venenos Elapídicos/genética , Animais , Ativação do Complemento/efeitos dos fármacos , Venenos Elapídicos/química , Venenos Elapídicos/farmacologia , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes
12.
Thromb Haemost ; 113(3): 548-52, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25031089

RESUMO

The complement system is an intrinsic part of the immune system and has important functions in both innate and adaptive immunity. On the other hand, inadvertent or misdirected complement activation is also involved in the pathogenesis of many diseases, contributing solely or significantly to tissue injury and disease development. Multiple approaches to develop pharmacological agents to inhibit complement are currently being pursued. We have developed a conceptually different approach of not inhibiting but depleting complement, based on the complement-depleting activities of cobra venom factor (CVF), a non-toxic cobra venom component with structural and functional homology to complement component C3. We developed a humanised version of CVF by creating human complement component C3 derivatives with complement-depleting activities of CVF (humanised CVF) as a promising therapeutic agent for diseases with complement pathogenesis. Here we review the beneficial therapeutic effect of humanised CVF in several murine models of vascular diseases such as reperfusion injury.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Complemento C3/farmacologia , Inativadores do Complemento/farmacologia , Proteínas do Sistema Complemento/metabolismo , Venenos Elapídicos/farmacologia , Fatores Imunológicos/farmacologia , Traumatismo por Reperfusão/tratamento farmacológico , Lesão Pulmonar Induzida por Ventilação Mecânica/tratamento farmacológico , Animais , Proteínas do Sistema Complemento/imunologia , Modelos Animais de Doenças , Humanos , Proteínas Recombinantes de Fusão/farmacologia , Traumatismo por Reperfusão/imunologia , Lesão Pulmonar Induzida por Ventilação Mecânica/imunologia
13.
Mol Immunol ; 61(2): 191-203, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25062833

RESUMO

The complement system is an integral component of both innate and adaptive immunity. However, complement is also a pathogenetic factor in many diseases. The development of agents for therapeutic complement inhibition is the topic of intense investigations by many investigators. We have developed a distinctly different therapeutic approach: complement depletion rather than inhibition. This approach is based on cobra venom factor (CVF), a C3 analog known to be able to safely deplete complement. This manuscript will briefly review the structure and activity of CVF, along with its similarities and differences to C3. Exploiting the knowledge of the structure/function relationship of CVF and C3, we created derivatives of human C3 which display the CVF-like activity of depleting complement, referred to as humanized CVF (hCVF). This review describes the structure and activity of hCVF, including the important property of not cleaving C5. The efficacy of hCVF for therapeutic complement depletion in nine preclinical models diseases with complement pathology is reviewed, including reperfusion injury, age-related macular degeneration (AMD), paroxysmal nocturnal hemoglobinuria (PNH), and immunogenicity of Factor VIII in hemophilia A. Complement depletion is characterized by the absence of toxicity, even after intra-arterial injection into the pulmonary artery of primates. No immunogenicity has been observed.


Assuntos
Venenos Elapídicos/química , Venenos Elapídicos/farmacologia , Proteínas Recombinantes de Fusão , Animais , Inativadores do Complemento/química , Inativadores do Complemento/farmacologia , Inativadores do Complemento/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Venenos Elapídicos/uso terapêutico , Humanos
14.
Toxicon ; 60(4): 632-47, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22609532

RESUMO

Cobra Venom Factor (CVF) is the complement-activating protein in cobra venom. CVF is structurally and functionally highly homologous to complement component C3. CVF, like C3b, the activated form of C3, forms a bimolecular complex with Factor B in serum, called C3/C5 convertase, an enzyme which activates complement components C3 and C5. Despite the high degree of homology, the two C3/C5 convertases exhibit significant functional differences. The most important difference is that the convertase formed with CVF (CVF,Bb) is physico-chemically far more stable than the convertase formed with C3b (C3b,Bb). In addition, the CVF,Bb convertase and CVF are completely resistant to inactivation by the complement regulatory proteins Factor H and Factor I. Furthermore, the CVF,Bb enzyme shows efficient C5-cleaving activity in fluid phase. In contrast, the C3b,Bb enzyme is essentially devoid of fluid-phase C5-cleaving activity. By taking advantage of the high degree of sequence identity at both the amino acid (85%) and DNA levels (93%) between CVF and cobra C3, we created hybrid proteins of CVF and cobra C3 where sections, or only a few amino acids, of the CVF sequence were replaced with the homologous amino acid sequence of cobra C3. In a first set of experiments, we created five hybrid proteins, termed H1 through H5, where the cobra C3 substitutions collectively spanned the entire length of the CVF protein. We also created three additional hybrid proteins where only four or five amino acid residues in CVF were exchanged with the corresponding amino acid residues from cobra C3. Collectively, these hybrid proteins, representing loss-of-function mutants of CVF, allowed the identification of regions and individual amino acid residues important for the CVF-specific functions. The results include the observation that the CVF ß-chain is crucially important for forming a stable convertase, whereas the CVF α-chain appears to harbor no CVF-specific functions. Furthermore, the CVF γ-chain is additionally important for the fluid-phase C5-cleaving activity of CVF,Bb. Interestingly, the structural changes in the individual hybrid proteins differentially affected the molecular functions of the CVF,Bb enzyme such as convertase formation, C3 cleavage, and C5 cleavage.


Assuntos
Complemento C3/química , Venenos Elapídicos/química , Proteínas Recombinantes de Fusão/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Ativação do Complemento , Complemento C3/genética , Convertases de Complemento C3-C5/metabolismo , Fator D do Complemento/química , Venenos Elapídicos/genética , Elapidae/fisiologia , Estabilidade Enzimática , Humanos , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Plasmídeos/biossíntese , Plasmídeos/genética , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética
16.
Toxicon ; 56(7): 1198-222, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20417224

RESUMO

Cobra venom factor (CVF) is the complement-activating protein in cobra venom. This manuscript reviews the structure and function of CVF, how it interacts with the complement system, the structural and functional homology to complement component C3, and the use of CVF as an experimental tool to decomplement laboratory animals to study the functions of complement in host defense and immune response as well as in the pathogenesis of diseases. This manuscript also reviews the recent progress in using the homology between CVF and C3 to study C3 structure and function, and to develop human C3 derivatives with the complement-depleting function of CVF. These human C3 derivatives represent humanized CVF, and are a conceptually different concept for pharmacological intervention of the complement system, therapeutic complement depletion. The use of humanized CVF for therapeutic complement depletion in several pre-clinical models of human diseases is also reviewed.


Assuntos
Complemento C3/química , Inativadores do Complemento/química , Venenos Elapídicos/química , Animais , Artrite/tratamento farmacológico , Artrite/patologia , Complemento C3/fisiologia , Inativadores do Complemento/imunologia , Venenos Elapídicos/genética , Venenos Elapídicos/imunologia , Hemoglobinúria Paroxística/tratamento farmacológico , Humanos , Degeneração Macular/tratamento farmacológico , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Engenharia de Proteínas , Proteínas Recombinantes de Fusão/efeitos adversos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/uso terapêutico , Homologia de Sequência de Aminoácidos
17.
Mol Immunol ; 47(2-3): 506-10, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19747734

RESUMO

Cobra venom factor (CVF) is a complement activating protein in cobra venom, which functionally resembles C3b, and has been used for decades for decomplementation of serum to investigate the role of complement in many model systems of disease. The use of CVF for clinical practice is considered impractical because of immunogenicity issues. Humanization of CVF was recently demonstrated to yield a potent CVF-like molecule. In the present study, we demonstrate that mice treated with recombinant humanized CVF (HC3-1496) are protected from myocardial ischemia-reperfusion (MI/R) injuries with resultant preservation of cardiac function. Also, C3 deposition in the myocardium following MI/R was not observed following treatment with HC3-1496. HC3-1496 led to complement activation and depletion of C3, but preserved C5 titers. These data suggest, unlike CVF, HC3-1496 does not form a C5 convertase in the mouse, similar to recent studies in human sera/plasma. These results suggest that humanized CVF (HC3-1496) protects the ischemic myocardium from reperfusion injuries induced by complement activation and represents a novel anti-complement therapy for potential clinical use.


Assuntos
Venenos Elapídicos/imunologia , Traumatismo por Reperfusão Miocárdica/imunologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Animais , Complemento C3/imunologia , Complemento C5/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/complicações , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Traumatismo por Reperfusão Miocárdica/complicações , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Volume Sistólico/fisiologia , Função Ventricular Esquerda/fisiologia
18.
Curr Pharm Des ; 13(28): 2916-26, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17979736

RESUMO

Cobra Venom Factor (CVF) is the complement-activating protein in cobra venom. CVF is a structural and functional analog of complement component C3. In serum, CVF forms a physicochemically stable and control-resistant C3/C5 convertase that continuously activates C3 and C5, ultimately leading to depletion of serum complement. As CVF can be safely administered to vertebrate animals, it has become an important tool for complement depletion to study the biological functions of complement and its role in the pathogenesis of disease. CVF has also been used for targeted complement activation by chemically coupling it to monoclonal antibodies. Complement depletion is an attractive concept for pharmacological intervention in diseases where complement activation is part of the pathogenetic mechanism. Toward that end, the structural homology of CVF and C3 has been exploited by creating hybrid proteins in which short portions of C3 sequence have been exchanged with corresponding portions of CVF that introduce the desired ability of forming a stable convertase into human C3. These human C3 derivatives are "humanized CVF" proteins that represent an attractive biopharmaceutical for therapeutic complement depletion.


Assuntos
Ativação do Complemento/fisiologia , Proteínas do Sistema Complemento/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Venenos Elapídicos/administração & dosagem , Venenos Elapídicos/metabolismo , Animais , Ativação do Complemento/efeitos dos fármacos , Venenos Elapídicos/química , Humanos
19.
J Biol Chem ; 279(29): 30836-43, 2004 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-15131128

RESUMO

Cobra venom factor (CVF) is the complement-activating protein from cobra venom. It is a structural and functional analog of complement component C3. CVF functionally resembles C3b, the activated form of C3. Like C3b, CVF binds factor B, which is subsequently cleaved by factor D to form the bimolecular complex CVF,Bb. CVF,Bb is a C3/C5 convertase that cleaves both complement components C3 and C5. CVF is a three-chain protein that structurally resembles the C3b degradation product C3c, which is unable to form a C3/C5 convertase. Both C3 and CVF are synthesized as single-chain prepro-proteins. This study reports the recombinant expression of pro-CVF in two insect cell expression systems (baculovirus-infected Sf9 Spodoptera frugiperda cells and stably transfected S2 Drosophila melanogaster cells). In both expression systems pro-CVF is synthesized initially as a single-chain pro-CVF molecule that is subsequently proteolytically processed into a two-chain form of pro-CVF that structurally resembles C3. The C3-like form of pro-CVF can be further proteolytically processed into another two-chain form of pro-CVF that structurally resembles C3b. Unexpectedly, all three forms of pro-CVF exhibit functional activity of mature, natural CVF. Recombinant pro-CVF supports the activation of factor B in the presence of factor D and Mg2+ and depletes serum complement activity like natural CVF. The bimolecular convertase pro-CVF,Bb exhibits both C3 cleaving and C5 cleaving activity. The activity of pro-CVF and the resulting C3/C5 convertase is indistinguishable from CVF and the CVF,Bb convertase. The ability to produce active forms of pro-CVF recombinantly ensures the continued availability of an important research reagent for complement depletion because cobra venom as the source for natural CVF will be increasingly difficult to obtain as the Indian cobra is on the list of endangered species. Experimental systems to express pro-CVF recombinantly will also be invaluable for studies to delineate the structure and function relationship of CVF and its differences from C3 as well as to generate human C3 derivatives with CVF-like function for therapeutic complement depletion ("humanized CVF").


Assuntos
Venenos Elapídicos/química , Proteínas Recombinantes/química , Animais , Linhagem Celular , Convertases de Complemento C3-C5/metabolismo , Fator D do Complemento/química , Proteínas do Sistema Complemento/metabolismo , DNA Complementar/metabolismo , Drosophila melanogaster , Escherichia coli/metabolismo , Vetores Genéticos , Glicosilação , Humanos , Insetos/metabolismo , Magnésio/química , Plasmídeos/metabolismo , Conformação Proteica , Relação Estrutura-Atividade , Fatores de Tempo , Tunicamicina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA