Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 13: 1007102, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36330514

RESUMEN

Background: The complement system is an essential component of our innate defense and plays a vital role in the pathogenesis of many diseases. Assessment of complement activation is critical in monitoring both disease progression and response to therapy. Complement analysis requires accurate and standardized sampling and assay procedures, which has proven to be challenging. Objective: We performed a systematic analysis of the current methods used to assess complement components and reviewed whether the identified studies performed their complement measurements according to the recommended practice regarding pre-analytical sample handling and assay technique. Results are supplemented with own data regarding the assessment of key complement biomarkers to illustrate the importance of accurate sampling and measuring of complement components. Methods: A literature search using the Pubmed/MEDLINE database was performed focusing on studies measuring the key complement components C3, C5 and/or their split products and/or the soluble variant of the terminal C5b-9 complement complex (sTCC) in human blood samples that were published between February 2017 and February 2022. The identified studies were reviewed whether they had used the correct sample type and techniques for their analyses. Results: A total of 92 out of 376 studies were selected for full-text analysis. Forty-five studies (49%) were identified as using the correct sample type and techniques for their complement analyses, while 25 studies (27%) did not use the correct sample type or technique. For 22 studies (24%), it was not specified which sample type was used. Conclusion: A substantial part of the reviewed studies did not use the appropriate sample type for assessing complement activation or did not mention which sample type was used. This deviation from the standardized procedure can lead to misinterpretation of complement biomarker levels and hampers proper comparison of complement measurements between studies. Therefore, this study underlines the necessity of general guidelines for accurate and standardized complement analysis.


Asunto(s)
Activación de Complemento , Complemento C5 , Humanos , Complemento C3 , Complejo de Ataque a Membrana del Sistema Complemento , Biomarcadores
2.
Front Immunol ; 13: 1039765, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36420270

RESUMEN

The most commonly used markers to assess complement activation are split products that are produced through activation of all three pathways and are located downstream of C3. In contrast, C4d derives from the cleavage of C4 and indicates either classical (CP) or lectin pathway (LP) activation. Although C4d is perfectly able to distinguish between CP/LP and alternative pathway (AP) activation, no well-established markers are available to differentiate between early CP and LP activation. Active enzymes of both pathways (C1s/C1r for the CP, MASP-1/MASP-2 for the LP) are regulated by C1 esterase inhibitor (C1-INH) through the formation of covalent complexes. Aim of this study was to develop validated immunoassays detecting C1s/C1-INH and MASP-1/C1-INH complex levels. Measurement of the complexes reveals information about the involvement of the respective pathways in complement-mediated diseases. Two sandwich ELISAs detecting C1s/C1-INH and MASP-1/C1-INH complex were developed and tested thoroughly, and it was investigated whether C1s/C1-INH and MASP-1/C1-INH complexes could serve as markers for either early CP or LP activation. In addition, a reference range for these complexes in healthy adults was defined, and the assays were clinically validated utilizing samples of 414 COVID-19 patients and 96 healthy controls. The immunoassays can reliably measure C1s/C1-INH and MASP-1/C1-INH complex concentrations in EDTA plasma from healthy and diseased individuals. Both complex levels are increased in serum when activated with zymosan, making them suitable markers for early classical and early lectin pathway activation. Furthermore, measurements of C1-INH complexes in 96 healthy adults showed normally distributed C1s/C1-INH complex levels with a physiological concentration of 1846 ± 1060 ng/mL (mean ± 2SD) and right-skewed distribution of MASP-1/C1-INH complex levels with a median concentration of 36.9 (13.18 - 87.89) ng/mL (2.5-97.5 percentile range), while levels of both complexes were increased in COVID-19 patients (p<0.0001). The newly developed assays measure C1-INH complex levels in an accurate way. C1s/C1-INH and MASP-1/C1-INH complexes are suitable markers to assess early classical and lectin pathway activation. An initial reference range was set and first studies showed that these markers have added value for investigating and unraveling complement activation in human disease.


Asunto(s)
COVID-19 , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa , Adulto , Humanos , Proteína Inhibidora del Complemento C1 , Proteínas del Sistema Complemento , COVID-19/diagnóstico , Lectinas , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/metabolismo , Ensayo de Inmunoadsorción Enzimática
3.
J Nephrol ; 34(3): 801-810, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33306183

RESUMEN

INTRODUCTION: Various studies have reported the importance of complement regulators in preventing mesothelial damage during peritoneal dialysis (PD). Its assessment, however, is limited in clinical practice due to the lack of easy access to the peritoneal membrane. Recently, a soluble form of the complement regulatory protein CD59 (sCD59) has been described. We therefore aimed to investigate the role of sCD59 in PD. METHODS: Plasma sCD59 was measured in 48 PD patients, 41 hemodialysis patients, 15 non-dialysis patients with chronic kidney disease and 14 healthy controls by ELISA (Hycult; HK374-02). Additionally, sCD59 and sC5b-9 were assessed in the peritoneal dialysate. RESULTS: sCD59 and sC5b-9 were detectable in the peritoneal dialysate of all patients, and marginally correlated (r = 0.27, P = 0.06). Plasma sCD59 levels were significantly higher in PD patients than in patients with chronic kidney disease and healthy controls, but did not differ from hemodialysis patients. During follow-up, 19% of PD patients developed peritoneal membrane failure and 27% of PD patients developed loss of residual renal function. In adjusted models, increased sCD59 levels in the dialysate (HR 3.44, 95% CI 1.04-11.40, P = 0.04) and in plasma (HR 1.08, 95% CI 1.01-1.17, P = 0.04) were independently associated with the occurrence of peritoneal membrane failure. Higher plasma levels of sCD59 were also associated with loss of residual renal function (HR 1.10, 95% CI 1.04-1.17, P < 0.001). CONCLUSIONS: Our study suggests that sCD59 has potential as a biomarker to predict peritoneal membrane function and loss of residual renal function in PD, thereby offering a tool to improve patient management.


Asunto(s)
Fallo Renal Crónico , Diálisis Peritoneal , Biomarcadores , Antígenos CD59 , Proteínas del Sistema Complemento , Soluciones para Diálisis , Humanos , Diálisis Peritoneal/efectos adversos , Peritoneo
4.
Antioxidants (Basel) ; 8(11)2019 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-31766295

RESUMEN

Oxidative stress-induced damage of the retinal pigment epithelium (RPE) and chronic inflammation have been suggested as major contributors to a range of retinal diseases. Here, we examined the effects of oxidative stress on endogenous complement components and proinflammatory and angiogenic responses in RPE cells. ARPE-19 cells exposed for 1-48 h to H2O2 had reduced cell-cell contact and increased markers for epithelial-mesenchymal transition but showed insignificant cell death. Stressed ARPE-19 cells increased the expression of complement receptors CR3 (subunit CD11b) and C5aR1. CD11b was colocalized with cell-derived complement protein C3, which was present in its activated form in ARPE-19 cells. C3, as well as its regulators complement factor H (CFH) and properdin, accumulated in the ARPE-19 cells after oxidative stress independently of external complement sources. This cell-associated complement accumulation was accompanied by increased nlrp3 and foxp3 expression and the subsequently enhanced secretion of proinflammatory and proangiogenic factors. The complement-associated ARPE-19 reaction to oxidative stress, which was independent of exogenous complement sources, was further augmented by the poly(ADP-ribose) polymerase (PARP) inhibitor olaparib. Our results indicate that ARPE-19 cell-derived complement proteins and receptors are involved in ARPE-19 cell homeostasis following oxidative stress and should be considered as targets for treatment development for retinal degeneration.

5.
Mult Scler ; 25(4): 523-531, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-29421990

RESUMEN

BACKGROUND: CD59, a broadly expressed glycosylphosphatidylinositol-anchored protein, is the principal cell inhibitor of complement membrane attack on cells. In the demyelinating disorders, multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD), elevated complement protein levels, including soluble CD59 (sCD59), were reported in cerebrospinal fluid (CSF). OBJECTIVES: We compared sCD59 levels in CSF and matched plasma in controls and patients with MS, NMOSD and clinically isolated syndrome (CIS) and investigated the source of CSF sCD59 and whether it was microparticle associated. METHODS: sCD59 was quantified using enzyme-linked immunosorbent assay (ELISA; Hycult; HK374-02). Patient and control CSF was subjected to western blotting to characterise anti-CD59-reactive materials. CD59 was localised by immunostaining and in situ hybridisation. RESULTS: CSF sCD59 levels were double those in plasma (CSF, 30.2 ng/mL; plasma, 16.3 ng/mL). Plasma but not CSF sCD59 levels differentiated MS from NMOSD, MS from CIS and NMOSD/CIS from controls. Elimination of microparticles confirmed that CSF sCD59 was not membrane anchored. CONCLUSION: CSF levels of sCD59 are not a biomarker of demyelinating diseases. High levels of sCD59 in CSF relative to plasma suggest an intrathecal source; CD59 expression in brain parenchyma was low, but expression was strong on choroid plexus (CP) epithelium, immediately adjacent the CSF, suggesting that this is the likely source.


Asunto(s)
Antígenos CD59/líquido cefalorraquídeo , Plexo Coroideo/metabolismo , Enfermedades Desmielinizantes/líquido cefalorraquídeo , Esclerosis Múltiple/líquido cefalorraquídeo , Neuromielitis Óptica/líquido cefalorraquídeo , Adulto , Antígenos CD59/sangre , Enfermedades Desmielinizantes/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/sangre , Neuromielitis Óptica/sangre
6.
Sci Rep ; 5: 15381, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26480887

RESUMEN

Triggering receptor expressed on myeloid cells (TREM)-1 plays an important role in innate immune responses and is upregulated under infectious as well as non-infectious conditions. In addition, a soluble TREM-1 variant (sTREM-1) is detectable in sera or bronchoalveolar-lavage fluids from patients. Currently, various studies are difficult to compare, since the methods of detection by enzyme-linked immunosorbent assays (ELISA) vary among different research groups. In this study, we compared three different s-TREM-1 specific ELISAs and identified individual assay characteristics finding notable differences in sTREM-1 concentrations in part depending on the employed buffers. Investigating potential confounding factors for sTREM-1 detection, serum heat-inactivation (HI) showed improved recovery compared to non-HI (NHI) serum, reproducible by addition of complement and re-heat-inactivation. Hence we identified complement as a heat-sensitive confounder in some sTREM-1 ELISAs. We conclude that it is difficult to directly compare data of several studies, in particular if different ELISAs are engaged. Immunoassays for research use only are in general hampered by lack of standardization. Further standardization is needed until sTREM-1 ELISA is capable for better reproducibility of studies and clinical application.


Asunto(s)
Ensayo de Inmunoadsorción Enzimática , Glicoproteínas de Membrana/sangre , Receptores Inmunológicos/sangre , Biomarcadores , Proteínas del Sistema Complemento/metabolismo , Factores de Confusión Epidemiológicos , Humanos , Isoformas de Proteínas , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Receptor Activador Expresado en Células Mieloides 1
7.
Artículo en Inglés | MEDLINE | ID: mdl-26973884

RESUMEN

BACKGROUND: gC1qR is a multifunctional cellular protein that has been linked to inflammation and cancer. gC1qR is highly upregulated in adenocarcinomas as compared to normal tissue counterparts, and soluble gC1qR (sgC1qR) has been detected in vitro in the pericellular milieu of proliferating malignant cells. AIM: The present study explored the tissue expression of gC1qR in pancreatic cancer by immunohistochemistry, and the presence of sgC1qR in vivo, by examining blood and malignant effusions from patients with metastatic pancreatic adenocarcinoma. METHODS: Tissue expression of gC1qR by pancreatic adenocarcinoma was visualized by immunohistochemistry. SgC1qR was quantified in serum from healthy volunteers (n=20) and pancreatic cancer patients (n=34), as well as in malignant pleural (n=23) and peritoneal effusions (n=27), using a newly developed, sensitive immunocapture sandwich ELISA. RESULTS: Overexpression of gC1qR was confirmed in pancreatic adenocarcinoma compared to nonmalignant pancreatic tissue. Moreover, increased serum levels of sgC1qR (0.29 ± 0.22 ng/ml) were noted in patients with metastatic pancreatic cancer compared to healthy controls (0.15 ± 0.10 ng/ml) (mean ± S.D.) (p=0.035). In 11 of 16 patients for whom sequential samples were available, serum sgC1qR levels rose with disease progression, and paralleled changes in tumor biomarkers, CEA and CA19.9. In addition to blood, sgC1qR was detected in malignant pleural (0.55 ± 0.47 ng/ml) and peritoneal effusions (0.57 ± 0.38 ng/ml). CONCLUSION: This study provides the first evidence for the presence of sgC1qR in vivo. The ability to detect sgC1qR in blood and body fluids will enable further studies to elucidate its pathophysiology in malignancy.

8.
J Immunol ; 179(3): 2005-12, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17641068

RESUMEN

Based on immune reactivity in response to a whole-cell colon tumor vaccine and using serological identification of Ags by recombinant cDNA expression cloning, we here describe the molecular and functional identification of a novel human tumor Ag. By screening a cDNA expression library derived from the coloncarcinoma cell line HT-29 with pooled colorectal cancer patients' sera, 26 clones reactive with IgG Abs could be identified. Characterization of these cDNA clones by sequence analysis and alignment, and detailed serological analysis revealed cancer-related immunoreactivity for the ErbB-3-binding protein-1 (Ebp1). Immunohistochemical staining of colorectal tumors and neighboring normal colon tissue indicated the observed cancer-related immunogenicity of Ebp1 to be related to overexpression. Via reverse immunology, five potential HLA-A2-restricted T cell epitopes were identified, of which two (Ebp1(45-54) and Ebp1(59-67)) bound HLA-A2 with intermediate and high affinity, respectively. Analysis of their immunogenicity in vitro indicated that only the high-affinity Ebp1(59) epitope gave rise to CD8(+) T cells capable of recognizing both exogenously loaded Ebp1 peptide and endogenously expressed Ebp1 on target cells. In addition, in vivo CD8(+) T cell responsiveness against the Ebp1(59) epitope could be detected in two of nine and three of six cancer patients PBMC and tumor draining lymph nodes, respectively, but not in nine of nine healthy donors tested. These data confirm that Ebp1 is an immunogenic protein, capable of eliciting CD8-mediated responses in vivo and in vitro, providing a rationale for further exploration of Ebp1 as a possible target for anticancer immunotherapy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Antígenos de Neoplasias/química , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/terapia , Inmunización Pasiva , Proteínas de Unión al ARN/química , Receptor ErbB-3/metabolismo , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Clonación Molecular , Neoplasias Colorrectales/patología , Pruebas Inmunológicas de Citotoxicidad , ADN Complementario/biosíntesis , Sistemas de Liberación de Medicamentos , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Biblioteca de Genes , Antígeno HLA-A2/inmunología , Células HT29 , Humanos , Inmunización Pasiva/métodos , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/genética , Linfocitos T Citotóxicos/inmunología
9.
Trends Mol Med ; 13(5): 200-9, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17379575

RESUMEN

Gene therapy is one of the promising strategies in cancer treatment. Recent studies identified molecular targets on angiogenically activated endothelial cells that can be used to deliver gene-transfer vehicles to the tumor site specifically. Furthermore, non-viral vehicles are emerging as an alternative for traditional viral gene-therapy approaches. Here, we describe how viral and non-viral gene-transfer vehicles have been and can be modified to target tumor endothelial cells for anti-angiogenesis gene therapy. Improving the specificity and safety of existing gene-therapy vehicles will make angiogenesis-targeted cancer gene therapy a valuable tool in the clinical setting.


Asunto(s)
Marcación de Gen/métodos , Terapia Genética/métodos , Neoplasias/terapia , Neovascularización Patológica/terapia , Animales , Portadores de Fármacos/uso terapéutico , Vectores Genéticos/uso terapéutico , Humanos , Modelos Biológicos , Neoplasias/irrigación sanguínea , Virus/genética
10.
Bioconjug Chem ; 18(3): 785-90, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17378601

RESUMEN

Identification of a tumor angiogenesis specific ligand would allow targeting of tumor vasculature. Lipidic vehicles can be used to deliver therapeutic agents for treatment of disease or contrast agents for molecular imaging. A targeting ligand would allow specific delivery of such formulations to angiogenic sites, thereby reducing side effects and gaining efficiency. Anginex, a synthetic 33-mer angiostatic peptide, has been described to home angiogenically activated endothelium, suggesting an ideal candidate as targeting ligand. To investigate this application of anginex, fluorescently labeled paramagnetic liposomes were conjugated with anginex. Using phase contrast and fluorescence microscopy as well as magnetic resonance imaging (MRI), we demonstrate that anginex-conjugated liposomes bind specifically to activated endothelial cells, suggesting application as an angiogenesis targeting agent for molecular targeting and molecular imaging of angiogenesis-dependent disease.


Asunto(s)
Endotelio Vascular/metabolismo , Vehículos Farmacéuticos/metabolismo , Proteínas/metabolismo , Secuencia de Aminoácidos , Células Cultivadas , Células Endoteliales/metabolismo , Endotelio Vascular/química , Humanos , Liposomas/química , Liposomas/metabolismo , Espectroscopía de Resonancia Magnética , Datos de Secuencia Molecular , Péptidos , Vehículos Farmacéuticos/análisis , Vehículos Farmacéuticos/química , Proteínas/análisis , Proteínas/química
11.
Proc Natl Acad Sci U S A ; 103(43): 15975-80, 2006 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-17043243

RESUMEN

We describe that galectin-1 (gal-1) is a receptor for the angiogenesis inhibitor anginex, and that the protein is crucial for tumor angiogenesis. gal-1 is overexpressed in endothelial cells of different human tumors. Expression knockdown in cultured endothelial cells inhibits cell proliferation and migration. The importance of gal-1 in angiogenesis is illustrated in the zebrafish model, where expression knockdown results in impaired vascular guidance and growth of dysfunctional vessels. The role of gal-1 in tumor angiogenesis is demonstrated in gal-1-null mice, in which tumor growth is markedly impaired because of insufficient tumor angiogenesis. Furthermore, tumor growth in gal-1-null mice no longer responds to antiangiogenesis treatment by anginex. Thus, gal-1 regulates tumor angiogenesis and is a target for angiostatic cancer therapy.


Asunto(s)
Galectina 1/metabolismo , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Galectina 1/deficiencia , Galectina 1/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Trasplante de Neoplasias , Neoplasias/genética , Neoplasias/terapia , Péptidos , Unión Proteica , Proteínas/metabolismo , Pez Cebra
12.
Biochem Biophys Res Commun ; 349(3): 1073-8, 2006 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-16970922

RESUMEN

Anginex, a synthetic 33-mer angiostatic peptide, specifically inhibits vascular endothelial cell proliferation and migration along with induction of apoptosis in endothelial cells. Here we report on the in vivo characterization of recombinant anginex and use of the artificial anginex gene for gene therapy approaches. Tumor growth of human MA148 ovarian carcinoma in athymic mice was inhibited by 80% when treated with recombinant anginex. Histological analysis of the tumors showed an approximate 2.5-fold reduction of microvessel density, suggesting that angiogenesis inhibition is the cause of the anti-tumor effect. Furthermore, there was a significant correlation between the gene expression patterns of 16 angiogenesis-related factors after treatment with both recombinant and synthetic anginex. To validate the applicability of the anginex gene for gene therapy, stable transfectants of murine B16F10 melanoma cells expressing recombinant anginex were made. Supernatants of these cells inhibited endothelial cell proliferation in vitro. Furthermore, after subcutaneous injection of these cells in C57BL/6 mice, an extensive delay in tumor growth was observed. These data show that the artificial anginex gene can be used to produce a recombinant protein with similar activity as its synthetic counterpart and that the gene can be applied in gene therapy approaches for cancer treatment.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Neoplasias/irrigación sanguínea , Neoplasias/patología , Proteínas/farmacología , Inhibidores de la Angiogénesis/genética , Inhibidores de la Angiogénesis/metabolismo , Animales , Antineoplásicos/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Péptidos , Proteínas/genética , Proteínas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Nano Lett ; 6(1): 1-6, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16402777

RESUMEN

MRI detectable and targeted quantum dots were developed. To that aim, quantum dots were coated with paramagnetic and pegylated lipids, which resulted in a relaxivity, r(1), of nearly 2000 mM(-1)s(-1) per quantum dot. The quantum dots were functionalized by covalently linking alphavbeta3-specific RGD peptides, and the specificity was assessed and confirmed on cultured endothelial cells. The bimodal character, the high relaxivity, and the specificity of this nanoparticulate probe make it an excellent contrast agent for molecular imaging purposes.


Asunto(s)
Aumento de la Imagen , Integrina alfaVbeta3/metabolismo , Sondas Moleculares , Oligopéptidos/metabolismo , Puntos Cuánticos , Células Cultivadas , Células Endoteliales/metabolismo , Gadolinio DTPA , Humanos , Liposomas , Imagen por Resonancia Magnética , Microscopía Fluorescente , Fosfatidiletanolaminas , Polietilenglicoles , Venas Umbilicales/citología
14.
Biochem Biophys Res Commun ; 333(4): 1261-8, 2005 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-15979575

RESUMEN

Anginex, a designed peptide 33-mer, is a potent angiogenesis inhibitor and anti-tumor agent in vivo. Anginex functions by inhibiting endothelial cell (EC) proliferation and migration leading to detachment and apoptosis of activated EC's. To better understand tumor endothelium targeting properties of anginex and enable its use in gene therapy, we constructed an artificial gene encoding the biologically exogenous peptide and produced the protein recombinantly in Pichia pastoris. Mass spectrometry shows recombinant anginex to be a dimer and circular dichroism shows the recombinant protein folds with beta-strand structure like the synthetic peptide. Moreover, like parent anginex, the recombinant protein is active at inhibiting EC growth and migration, as well as inhibiting angiogenesis in vivo in the chorioallantoic membrane of the chick embryo. This study demonstrated that it is possible to produce a functionally active protein version of a rationally designed peptide, using an artificial gene and the recombinant protein approach.


Asunto(s)
Inhibidores de la Angiogénesis/biosíntesis , Inhibidores de la Angiogénesis/química , Clonación Molecular/métodos , Genes Sintéticos/genética , Ingeniería de Proteínas/métodos , Proteínas/química , Proteínas/metabolismo , Inhibidores de la Angiogénesis/análisis , Inhibidores de la Angiogénesis/genética , Inhibidores de la Angiogénesis/farmacología , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Técnicas de Transferencia de Gen , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Péptidos , Pichia/genética , Pichia/metabolismo , Proteínas/genética , Proteínas/farmacología , Proteínas Recombinantes/análisis , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química
15.
Exp Cell Res ; 299(2): 286-93, 2004 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15350528

RESUMEN

The present study describes a method to simultaneously obtain the angiogenic expression profile in tumor cells and vascular cells of a single tumor. Human- and mouse-specific primers were used for quantitative real-time RT-PCR to determine the expression of vascular endothelial growth factors A, B, C, and D, vascular endothelial growth factor receptors 1, 2, and 3, neuropilin 1 and 2, angiopoietin 1, 2, 3/4, tyrosine kinase receptors 1 and 2, basic fibroblast growth factor (bFGF) in xenograft tumors obtained by injection of human ovarian carcinoma cells in nude mice. In addition, the effect of treatment with anginex and taxol on the expression profile was analyzed. Most factors were expressed higher in vascular cells as compared to tumor cells. In response to treatment, tumor cells significantly upregulated bFGF expression and downregulated VEGF receptor expression. This was accompanied by downregulation of VEGF-B and -D, and upregulation of angiopoietin-3 as well as angiopoetin receptors in nontumor cells. In conclusion, real-time qRT-PCR combined with xenograft tumor models presents a sensitive method to monitor angiogenesis and to analyze interactions between tumor cells and nontumor cells in vivo. The approach can be applied to different research fields in which xenograft models are used.


Asunto(s)
Proteínas Angiogénicas/genética , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Melanoma/irrigación sanguínea , Neovascularización Patológica/genética , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Cutáneas/irrigación sanguínea , Inhibidores de la Angiogénesis/farmacología , Animales , Antineoplásicos/farmacología , Secuencia de Bases , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/metabolismo , Melanoma/patología , Ratones , Datos de Secuencia Molecular , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Paclitaxel/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Ácido Nucleico , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Trasplante Heterólogo
16.
J Immunol ; 169(5): 2772-80, 2002 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12193752

RESUMEN

In the last few years it has been shown that the humoral immune response in cancer patients is a rich source of putative cancer vaccine candidates. To fully explore the complex information present within the Ab repertoire of cancer patients, we have applied a method, serological Ag selection, to molecularly define tumor Ags recognized by the humoral immune response in colorectal cancer (CRC). First, we built a cDNA display library by cloning a cDNA library from CRC cell line HT-29 for expression as a fusion protein with a filamentous phage minor coat protein, pVI. This cDNA display library was then enriched on pooled sera from CRC patients who had undergone active specific immunization with autologous tumor. We identified a panel of 19 clones reactive with the serum pool. Seventeen of 19 (89%) clones showed reactivity with one or more of the eight Ag-reactive sera, conversely six of eight (75%) sera were reactive with at least one of the 19 clones. Sequencing revealed that these 19 clones represented 13 different Ags. A detailed serological analysis of the 13 different Ags showed preferential reactivity to sera of cancer patients for six different Ags. Four of these Ags displayed increased serum reactivity after the active specific immunization procedure. Furthermore, one of the six Ags, a novel Ag homologous to HSPC218, showed restricted expression in normal testis, suggesting that it belongs to the cancer-testis Ag family. Some of the Ags we have identified may be candidates for tumor vaccination, for sero-diagnosis of cancer, as prognostic markers, or as probes for monitoring tumor cell-based vaccination trials.


Asunto(s)
Antígenos de Neoplasias/genética , Antígenos de Neoplasias/aislamiento & purificación , Neoplasias Colorrectales/inmunología , ADN Complementario/biosíntesis , Perfilación de la Expresión Génica/métodos , Biblioteca de Genes , Inovirus/genética , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Antineoplásicos/sangre , Reacciones Antígeno-Anticuerpo/genética , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/sangre , Clonación Molecular/métodos , Neoplasias Colorrectales/sangre , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Células HT29 , Humanos , Masculino , Persona de Mediana Edad , Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/genética , Análisis de Secuencia de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...