Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Haematologica ; 103(2): 351-360, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29146705

RESUMEN

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.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Complemento C3/farmacología , Factor VIII/inmunología , Animales , Presentación de Antígeno/inmunología , Activación de Complemento , Células Dendríticas/fisiología , Endocitosis/efectos de los fármacos , Humanos , Inmunidad/efectos de los fármacos , Ratones
2.
Basic Res Cardiol ; 111(1): 7, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26667317

RESUMEN

Recent advances in basic cardiovascular research as well as their translation into the clinical situation were the focus at the last "New Frontiers in Cardiovascular Research meeting". Major topics included the characterization of new targets and procedures in cardioprotection, deciphering new players and inflammatory mechanisms in ischemic heart disease as well as uncovering microRNAs and other biomarkers as versatile and possibly causal factors in cardiovascular pathogenesis. Although a number of pathological situations such as ischemia-reperfusion injury or atherosclerosis can be simulated and manipulated in diverse animal models, also to challenge new drugs for intervention, patient studies are the ultimate litmus test to obtain unequivocal information about the validity of biomedical concepts and their application in the clinics. Thus, the open and bidirectional exchange between bench and bedside is crucial to advance the field of ischemic heart disease with a particular emphasis of understanding long-lasting approaches in cardioprotection.


Asunto(s)
Enfermedades Cardiovasculares , Investigación Biomédica Traslacional , Animales , Humanos
3.
Basic Res Cardiol ; 111(6): 69, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27743118

RESUMEN

In this meeting report, particularly addressing the topic of protection of the cardiovascular system from ischemia/reperfusion injury, highlights are presented that relate to conditioning strategies of the heart with respect to molecular mechanisms and outcome in patients' cohorts, the influence of co-morbidities and medications, as well as the contribution of innate immune reactions in cardioprotection. Moreover, developmental or systems biology approaches bear great potential in systematically uncovering unexpected components involved in ischemia-reperfusion injury or heart regeneration. Based on the characterization of particular platelet integrins, mitochondrial redox-linked proteins, or lipid-diol compounds in cardiovascular diseases, their targeting by newly developed theranostics and technologies opens new avenues for diagnosis and therapy of myocardial infarction to improve the patients' outcome.


Asunto(s)
Cardiología/tendencias , Enfermedades Cardiovasculares , Nanomedicina Teranóstica/tendencias , Animales , Cardiología/métodos , Humanos
4.
EMBO J ; 28(16): 2469-78, 2009 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-19574954

RESUMEN

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.


Asunto(s)
Convertasas de Complemento C3-C5/metabolismo , Factor B del Complemento/química , Factor B del Complemento/metabolismo , Venenos Elapídicos/química , Venenos Elapídicos/metabolismo , Elapidae/metabolismo , Animales , Línea Celular , Convertasas de Complemento C3-C5/química , Factor B del Complemento/genética , Cristalografía por Rayos X , Venenos Elapídicos/genética , Venenos Elapídicos/aislamiento & purificación , Activación Enzimática , Expresión Génica , Humanos , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína
5.
Blood ; 114(26): 5322-30, 2009 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-19805620

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Complemento C3/deficiencia , Linfoma/tratamiento farmacológico , Linfoma/inmunología , Animales , Anticuerpos Monoclonales de Origen Murino , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Rituximab
6.
Adv Exp Med Biol ; 703: 151-62, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20711713

RESUMEN

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.


Asunto(s)
Inactivadores del Complemento/farmacología , Venenos Elapídicos/farmacología , Degeneración Macular Húmeda/tratamiento farmacológico , Animales , Complemento C3/genética , Modelos Animales de Enfermedad , Venenos Elapídicos/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Recombinantes de Fusión/genética , Degeneración Macular Húmeda/inmunología , Degeneración Macular Húmeda/patología , Degeneración Macular Húmeda/cirugía
7.
Hawaii Med J ; 69(6 Suppl 3): 56-8, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20540006

RESUMEN

Cancer Health Disparities. The term cancer health disparities describes the unequal burden of cancer incidence, morbidity, and mortality in different population groups. By and large, the higher burden of cancer is observed in racial or ethnic minorities, immigrant communities, inner city populations, and insular, rural, or otherwise geographically isolated populations. In rare instances, however, the majority Caucasian population suffers a higher burden of cancer, e.g., melanoma. There are multiple and overlapping causes for cancer health disparities, including genetic make up, cultural norms and beliefs, behavior, environmental factors, as well as social aspects such as socio-economic status, poverty, and education. Reducing cancer health disparities is an important and integral component of the nation's effort to improve the health of all Americans. The science of cancer health disparities aims to better understand the causes of health disparities and to develop better means of prevention, diagnosis, and therapy, although some of the social aspects of cancer health disparities may be beyond the reach of the biomedical community.


Asunto(s)
Investigación Biomédica/educación , Creación de Capacidad , Investigación sobre Servicios de Salud/organización & administración , Disparidades en el Estado de Salud , Disparidades en Atención de Salud , Neoplasias , Guam , Hawaii , Educación en Salud/organización & administración , Humanos , Relaciones Interinstitucionales , Neoplasias/diagnóstico , Neoplasias/prevención & control , Apoyo a la Investigación como Asunto , Universidades
8.
Front Cell Dev Biol ; 8: 606407, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33425913

RESUMEN

This article reviews the pathogenetic role of the complement system in myocardial infarction reperfusion injury. The complement activation pathways involved in myocardial tissue injury are identified, as are the complement-derived effector molecules. The results of past anti-complement therapies are reviewed; as the more recent therapeutic concept of complement depletion with humanized CVF described.

9.
Toxicon ; 184: 68-77, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32526239

RESUMEN

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.


Asunto(s)
Convertasas de Complemento C3-C5/química , Factor B del Complemento/química , Venenos Elapídicos/química , Animales , Complemento C3 , Factor H de Complemento , Humanos , Proteínas Recombinantes de Fusión
10.
JCO Glob Oncol ; 6: 155-160, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32031449

RESUMEN

Sociocultural, geographic, and biologic factors contribute to cancer health disparities (CHDs) in indigenous Pacific peoples (IPPs) in Guam, Hawai'i, and the US Associated Pacific Islands (USAPI). IPPs experience a greater burden of CHDs that are associated with late-stage diagnosis and poor survival outcomes compared with majority populations in the United States. A 16-year partnership between the University of Guam (UOG) and University of Hawai'i Cancer Center (UHCC) aims to advance health equity in Guam, Hawai'i, and the USAPI through cancer research, training, and outreach. Investigators at collaborating institutions study issues of regional and cultural relevance in IPPs, including breast, cervical, liver, and oral cancers and use of tobacco and betel nuts (Areca nuts). Junior faculty with IPP ancestry or those who are focused on CHDs in IPPs receive mentorship and career development opportunities, academic fellowships are provided for graduate students, and Pacific Island communities are engaged through a participatory development process. The partnership has generated more than 90 peer-reviewed publications, more than 100 abstracts, and 11 grant awards. Thirty graduate scholars from under-represented minorities have been trained, including two who are now UOG faculty and are conducting independent research, contributing to the partnership, and mentoring scientists of tomorrow. Participatory community engagement has contributed to the passage of significant cancer prevention and control legislation in Hawai'i, Guam, and Saipan. Research capacity at UOG has increased significantly, and research at UHCC has expanded to address issues unique to IPPs. Graduate students from under-represented minorities are pursuing careers in cancer research. A regional research infrastructure has been established to support team science, and research findings are informing public health policy and planning.


Asunto(s)
Creación de Capacidad , Neoplasias de la Boca , Guam , Hawaii/epidemiología , Humanos , Islas del Pacífico/epidemiología , Estados Unidos/epidemiología
11.
Dev Comp Immunol ; 33(1): 105-16, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18760301

RESUMEN

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.


Asunto(s)
Complemento C3/química , Venenos Elapídicos/química , Animales , Clonación Molecular , Complemento C3/genética , Convertasas de Complemento C3-C5/química , Convertasas de Complemento C3-C5/genética , Factor H de Complemento/química , Venenos Elapídicos/genética , Fibrinógeno/química , Hemólisis , Humanos , Modelos Moleculares , Unión Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Ovinos
12.
Toxicon ; 167: 106-116, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31207349

RESUMEN

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.


Asunto(s)
Complemento C3/química , Venenos Elapídicos/química , Secuencia de Aminoácidos , Humanos , Proteínas Recombinantes de Fusión/química , Análisis de Secuencia de Proteína , Relación Estructura-Actividad
13.
Hawaii J Med Public Health ; 78(6): 205-207, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31205817

RESUMEN

This manuscript describes the efforts in research, education, and outreach of a unique partnership between the University of Hawai'i Cancer Center and the University of Guam in addressing cancer health disparities faced by Pacific Islanders in Hawai'i, Guam, and other parts of Micronesia. Significant accomplishments of this 15-year collaboration in research, training Micronesian students, and impact on the local communities are highlighted.


Asunto(s)
Investigación Biomédica , Instituciones Oncológicas , Disparidades en Atención de Salud , Nativos de Hawái y Otras Islas del Pacífico/etnología , Neoplasias/etiología , Universidades , Investigación Biomédica/educación , Conducta Cooperativa , Detección Precoz del Cáncer , Guam , Hawaii , Educación en Salud , Humanos , Neoplasias/diagnóstico , Neoplasias/prevención & control
14.
Adv Exp Med Biol ; 632: 293-307, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19025130

RESUMEN

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.


Asunto(s)
Complemento C3/química , Complemento C3/metabolismo , Complemento C3/uso terapéutico , Proteínas del Sistema Complemento/metabolismo , Animales , Artritis Experimental/tratamiento farmacológico , Complemento C3/genética , Venenos Elapídicos/química , Venenos Elapídicos/metabolismo , Humanos , Ratones , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Ingeniería de Proteínas , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico
15.
Mol Immunol ; 97: 1-7, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29525557

RESUMEN

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.


Asunto(s)
Anticuerpos Neutralizantes/metabolismo , Formación de Anticuerpos , Venenos Elapídicos/inmunología , Proteínas Recombinantes de Fusión/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Drosophila melanogaster , Venenos Elapídicos/química , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión/química
17.
Mol Immunol ; 41(2-3): 191-9, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15159065

RESUMEN

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.


Asunto(s)
Clonación Molecular/métodos , Venenos Elapídicos/genética , Animales , Activación de Complemento/efectos de los fármacos , Venenos Elapídicos/química , Venenos Elapídicos/farmacología , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes
18.
Mol Immunol ; 39(5-6): 357-65, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12220893

RESUMEN

Cobra venom factor (CVF), the anticomplementary protein in cobra venom, activates the alternative complement pathway, eventually leading to complement consumption. Here, we describe the development of a transgenic mouse model for CVF. We generated a DNA construct containing the full-length cDNA for single-chain pre-pro-CVF. Expression of CVF was controlled by the alpha(1)-antitrypsin promoter to achieve liver-specific expression. Linearized DNA was microinjected into murine ovary cells (strain CD(2)F(1) (BALB/cxDBA/2J)) and the newborn mice were analyzed for stable integration of CVF DNA. After establishing the transgene, mice were propagated in a BALB/c background. The CVF mRNA was detected in the liver and, in some animals, in the kidney. CVF protein was detected in small amounts in the serum. Serum complement hemolytic activity in CVF-transgenic mice was virtually absent. The concentration of plasma C3 was significantly reduced. The CVF-transgenic animals show no unusual phenotype. They provide an animal model to study the effect of long-term complement depletion by continued activation, as well as the role of complement in host immune response and pathogenesis of disease.


Asunto(s)
Venenos Elapídicos/genética , Animales , Complemento C3/análisis , Proteínas del Sistema Complemento/fisiología , Venenos Elapídicos/análisis , Hemólisis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Ratones Transgénicos , ARN Mensajero/análisis
19.
Thromb Haemost ; 113(3): 548-52, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25031089

RESUMEN

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.


Asunto(s)
Activación de Complemento/efectos de los fármacos , Complemento C3/farmacología , Inactivadores del Complemento/farmacología , Proteínas del Sistema Complemento/metabolismo , Venenos Elapídicos/farmacología , Factores Inmunológicos/farmacología , Daño por Reperfusión/tratamiento farmacológico , Lesión Pulmonar Inducida por Ventilación Mecánica/tratamiento farmacológico , Animales , Proteínas del Sistema Complemento/inmunología , Modelos Animales de Enfermedad , Humanos , Proteínas Recombinantes de Fusión/farmacología , Daño por Reperfusión/inmunología , Lesión Pulmonar Inducida por Ventilación Mecánica/inmunología
20.
Methods Mol Biol ; 283: 87-108, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15197304

RESUMEN

Heterobifunctional crosslinking reagents are small molecular weight chemicals containing two different reactive groups that have become important tools in generating conjugates of two different biomolecules, such as two proteins. The resulting bioconjugates are hybrid molecules or proteins, a new category of biomolecules that exhibit the combined functions of the two parent biomolecules. An important category of hybrid proteins are conjugates of antibodies with other effector molecules, such as drugs or toxins. These antibody conjugates or immunoconjugates have a variety of the applications in medicine, with particular emphasis on the treatment of cancer. The most commonly used heterobifunctional crosslinking reagents for the synthesis of antibody conjugates contain an N-hydroxysuccinimide ester moiety, which allows derivatization of amino groups in proteins. The chemical modification of a functionally important amino group in the antigen-binding region of an antibody causes impairment or loss of the antigen binding function, resulting in a defective antibody conjugate that lacks one of its component functions. Furthermore, even if the chemical derivatization does not affect the antigen binding function, the subsequent coupling of an effector protein at or near the antigen-binding region can also cause the loss of the antigen binding function for steric reasons. In this chapter, heterobifunctional crosslinking reagents are described that allow the generation of antibody conjugates where the effector proteins are coupled to the antibody carbohydrate moieties. Because antibody carbohydrate moieties are distal from the antigen-binding region, the use of carbohydrate-directed heterobifunctional crosslinking reagents, such as S-(2-thiopyridyl)-L-cysteine hydrazide (TPCH), prevents inactivation of the antigen-binding function. The synthesis of two carbohydrate-directed heterobifunctional crosslinking reagents is described. Coupling protocols for the preparation of antibody conjugates with effector proteins of different sizes using carbohydrate-directed heterobifunctional crosslinking reagents are also provided.


Asunto(s)
Anticuerpos/química , Cisteína/análogos & derivados , Inmunoconjugados/química , Oligosacáridos/química , Anticuerpos Monoclonales/química , Reactivos de Enlaces Cruzados , Cisteína/síntesis química , Humanos , Hidrazinas/síntesis química , Inmunoglobulina M/química , Proteínas/química , Piridinas/síntesis química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA