Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Anal Chem ; 91(12): 7603-7612, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31088078

RESUMO

Clinical and biomedical research seeks single-cell quantification to better understand their roles in a complex, multicell environment. Recently, quantification of vascular endothelial growth factor receptors (VEGFRs) provided important insights into endothelial cell characteristics and response in tumor microenvironments. However, existing technologies for quantifying plasma membrane receptor tyrosine kinases (RTKs) lack multiplexing capabilities, limiting detailed characterization. Here, we use the unique spectral properties of quantum dots (Qdots) to optimize and dually quantify VEGFR1 and VEGFR2 on human umbilical vein endothelial cells (HUVECs). To enable this quantification, we reduce nonspecific binding between Qdot-conjugated antibodies and cells via buffer optimization. Second, we identify optimal labeling conditions by examining Qdot-conjugated antibody binding to five receptors: VEGFRs (VEGFR1 and VEGFR2), their coreceptor neuropilin1 (NRP1), and platelet-derived growth factor receptor (PDGFRα and PDGFRß). We establish that 800-20 000 is the dynamic range where accurate Qdot-enabled quantification can be achieved. Through these optimizations, we demonstrate measurement of 1 100 VEGFR1 and 6 900 VEGFR2 per HUVEC. We induce ∼90% upregulation of VEGFR1 and ∼30% downregulation of VEGFR2 concentration via 24 h VEGF-A165 treatment. We observe no change in VEGFR1 or VEGFR2 concentration with 24 h VEGF-B167 treatment. We further apply Qdots to analyze HUVEC heterogeneity and observe that 24 h VEGF-A165 treatment induces a ∼15% decrease in VEGFR2 heterogeneity, but little to no change in VEGFR1 heterogeneity. We observe that VEGF-B167 induces little to no change in either VEGFR1 or VEGFR2 heterogeneity. Overall, we demonstrate experimental and analytical strategies for quantifying two or more RTKs at single-level using Qdots, which will help provide new insights into biological systems.


Assuntos
Pontos Quânticos/química , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Anticorpos Imobilizados/química , Anticorpos Imobilizados/imunologia , Regulação para Baixo/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Receptores do Fator de Crescimento Derivado de Plaquetas/análise , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/imunologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia
2.
Cell Metab ; 29(2): 457-474.e5, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30595478

RESUMO

The nature of obesity-associated islet inflammation and its impact on ß cell abnormalities remains poorly defined. Here, we explore immune cell components of islet inflammation and define their roles in regulating ß cell function and proliferation. Islet inflammation in obese mice is dominated by macrophages. We identify two islet-resident macrophage populations, characterized by their anatomical distributions, distinct phenotypes, and functional properties. Obesity induces the local expansion of resident intra-islet macrophages, independent of recruitment from circulating monocytes. Functionally, intra-islet macrophages impair ß cell function in a cell-cell contact-dependent manner. Increased engulfment of ß cell insulin secretory granules by intra-islet macrophages in obese mice may contribute to restricting insulin secretion. In contrast, both intra- and peri-islet macrophage populations from obese mice promote ß cell proliferation in a PDGFR signaling-dependent manner. Together, these data define distinct roles and mechanisms for islet macrophages in the regulation of islet ß cells.


Assuntos
Inflamação/imunologia , Células Secretoras de Insulina/metabolismo , Macrófagos/imunologia , Obesidade/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Animais , Linhagem Celular , Proliferação de Células , Secreção de Insulina , Células Secretoras de Insulina/patologia , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos
3.
Curr Med Chem ; 23(37): 4176-4220, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27528054

RESUMO

Molecularly targeted anticancer therapy involves the use of drugs or other substances affecting specific molecular targets that play a part in the development, progression and spread of a given neoplasm. By contrast, the majority of classical chemotherapeutics act on all rapidly proliferating cells, both healthy and cancerous ones. Target anticancer drugs are designed to achieve a particular aim and they usually act cytostatically, not cytotoxically like classical chemotherapeutics. At present, more than 300 biological molecular targets have been identified. The proteins involved in cellular metabolism include (among others) receptor proteins, signal transduction proteins, mRNA thread matrix synthesis proteins participating in neoplastic transformation, cell cycle control proteins, functional and structural proteins. The receptor proteins that are targeted by currently used anticancer drugs comprise the epithelial growth factor receptor (EGFR), platelet-derived growth factor receptor (PDGFR) and vascular endothelial growth factor receptor(VEGFR). Target anticancer drugs may affect extracellular receptor domains (antibodies) or intracellular receptor domains (tyrosine kinase inhibitors). The blocking of the mRNA thread containing information about the structure of oncogenes (signal transduction proteins) is another molecular target of anticancer drugs. That type of treatment, referred to as antisense therapy, is in clinical trials. When the synthesis of genetic material is disturbed, in most cases the passage to the next cycle phase is blocked. The key proteins responsible for the blockage are cyclines and cycline- dependent kinases (CDK). Clinical trials are focused on natural and synthetic substances capable of blocking various CDKs. The paper discusses the molecular targets and chemical structure of target anticancer drugs that have been approved for and currently applied in antineoplastic therapy together with indications and contraindications for their application.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/química , Antineoplásicos/metabolismo , Receptores ErbB/metabolismo , Humanos , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Fator A de Crescimento do Endotélio Vascular/imunologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Arthritis Rheumatol ; 68(9): 2263-73, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27111463

RESUMO

OBJECTIVE: To describe a skin-SCID mouse chimeric model of systemic sclerosis (SSc; scleroderma) fibrosis based on engraftment of ex vivo-bioengineered skin using skin cells derived either from scleroderma patients or from healthy donors. METHODS: Three-dimensional bioengineered skin containing human keratinocytes and fibroblasts isolated from skin biopsy specimens from healthy donors or SSc patients was generated ex vivo and then grafted onto the backs of SCID mice. The features of the skin grafts were analyzed by immunohistochemistry, and the functional profile of the graft fibroblasts was defined before and after treatment with IgG from healthy controls or SSc patients. Two procedures were used to investigate the involvement of platelet-derived growth factor receptor (PDGFR): 1) nilotinib, a tyrosine kinase inhibitor, was administered to mice before injection of IgG from SSc patient sera (SSc IgG) into the grafts, and 2) human anti-PDGFR monoclonal antibodies were injected into the grafts. RESULTS: Depending on the type of bioengineered skin grafted, the regenerated human skin exhibited either the typical scleroderma phenotype or the healthy human skin architecture. Treatment of animals carrying healthy donor skin grafts with SSc IgG resulted in the appearance of a bona fide scleroderma phenotype, as confirmed by increased collagen deposition and fibroblast activation markers. Results of the experiments involving administration of nilotinib or monoclonal antibodies confirmed the involvement of PDGFR. CONCLUSION: Our results provide the first in vivo demonstration of the fibrotic properties of anti-PDGFR agonistic antibodies. This bioengineered skin-humanized mouse model can be used to test in vivo the progression of the disease and to monitor response to antifibrotic drugs.


Assuntos
Autoanticorpos/administração & dosagem , Modelos Animais de Doenças , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Esclerodermia Localizada/imunologia , Escleroderma Sistêmico/imunologia , Animais , Fibrose/imunologia , Camundongos , Camundongos SCID , Esclerodermia Localizada/patologia , Escleroderma Sistêmico/patologia , Pele/imunologia
5.
Arthritis Rheumatol ; 67(12): 3234-44, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26246178

RESUMO

OBJECTIVE: To determine the relationships between systemic sclerosis (SSc)-related autoantibodies, as well as their clinical associations, in a well-characterized Australian patient cohort. METHODS: Serum from 505 Australian SSc patients were analyzed with a commercial line immunoassay (EuroLine; Euroimmun) for autoantibodies to centromere proteins CENP-A and CENP-B, RNA polymerase III (RNAP III; epitopes 11 and 155), the 90-kd nucleolar protein NOR-90, fibrillarin, Th/To, PM/Scl-75, PM/Scl-100, Ku, topoisomerase I (topo I), tripartite motif-containing protein 21/Ro 52, and platelet-derived growth factor receptor. Patient subgroups were identified by hierarchical clustering of the first 2 dimensions of a principal components analysis of quantitative autoantibody scores. Results were compared with detailed clinical data. RESULTS: A total of 449 of the 505 patients were positive for at least 1 autoantibody by immunoblotting. Heatmap visualization of autoantibody scores, along with principal components analysis clustering, demonstrated strong, mutually exclusive relationships between CENP, RNAP III, and topo I. Five patient clusters were identified: CENP, RNAP III strong, RNAP III weak, topo I, and other. Clinical features associated with CENP, RNAP III, and topo I were consistent with previously published reports concerning limited cutaneous and diffuse cutaneous SSc. A novel finding was the statistical separation of RNAP III into 2 clusters. Patients in the RNAP III strong cluster had an increased risk of gastric antral vascular ectasia, but a lower risk of esophageal dysmotility. Patients in the other cluster were more likely to be male and to have a history of smoking and a history of malignancy, but were less likely to have telangiectasia, Raynaud's phenomenon, and joint contractures. CONCLUSION: Five major autoantibody clusters with specific clinical and serologic associations were identified in Australian SSc patients. Subclassification and disease stratification using autoantibodies may have clinical utility, particularly in early disease.


Assuntos
Autoanticorpos/imunologia , Escleroderma Sistêmico/imunologia , Idoso , Antígenos Nucleares/imunologia , Austrália , Autoantígenos/imunologia , Proteína Centromérica A , Proteína B de Centrômero/imunologia , Proteínas Cromossômicas não Histona/imunologia , Estudos de Coortes , Contratura/etiologia , Contratura/imunologia , DNA Topoisomerases Tipo I/imunologia , Proteínas de Ligação a DNA/imunologia , Transtornos da Motilidade Esofágica/etiologia , Transtornos da Motilidade Esofágica/imunologia , Exorribonucleases/imunologia , Complexo Multienzimático de Ribonucleases do Exossomo/imunologia , Feminino , Ectasia Vascular Gástrica Antral/etiologia , Ectasia Vascular Gástrica Antral/imunologia , Humanos , Immunoblotting , Autoantígeno Ku , Masculino , Pessoa de Meia-Idade , Neoplasias/epidemiologia , Proteínas Pol1 do Complexo de Iniciação de Transcrição/imunologia , Análise de Componente Principal , RNA Polimerase III/imunologia , Proteínas de Ligação a RNA/imunologia , Doença de Raynaud/etiologia , Doença de Raynaud/imunologia , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Ribonucleoproteínas/imunologia , Escleroderma Sistêmico/complicações , Escleroderma Sistêmico/epidemiologia , Fatores Sexuais , Fumar/epidemiologia , Telangiectasia/etiologia , Telangiectasia/imunologia
6.
J Cancer Res Clin Oncol ; 139(8): 1397-404, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23728535

RESUMO

PURPOSE: The existence of platelet-derived growth factor (PDGF) receptor autoantibodies in systemic sclerosis is conflicting, and such antibodies were also detected in patients with chronic graft-versus-host disease (GvHD) after allogeneic peripheral blood stem cell transplantation (PBSCT). We therefore aimed to screen for PDGF receptor autoantibodies in patients with chronic GvHD. PATIENTS AND METHODS: We evaluated the existence of PDGF receptor autoantibodies in 39 patients, while 17 patients presented with a limited and 8 patients with an extensive chronic GvHD, respectively. Furthermore, 14 out of 39 patients had no chronic GvHD. RESULTS: We detected at least low levels of PDGF receptor autoantibodies in nearly all (35 of 39) patients after allogeneic PBSCT. Interestingly, only one of six patients with high levels of PDGF receptor autoantibodies presented with an extensive chronic GvHD, while the remaining six patients had no clinical signs of chronic GvHD. Thus, there was no correlation between the quantitative detection of antibodies directed against the PDGF receptor and the presence or severity of chronic GvHD. CONCLUSION: Platelet-derived growth factor receptor autoantibodies could easily be detected in patient sera. Nevertheless, we did not observe any correlation between the presence of PDGF receptor autoantibodies and the severity of chronic GvHD in patients who underwent allogeneic PBSCT.


Assuntos
Autoanticorpos/sangue , Doença Enxerto-Hospedeiro/imunologia , Transplante de Células-Tronco de Sangue Periférico/efeitos adversos , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Autoanticorpos/imunologia , Autoantígenos/imunologia , Doença Crônica , Feminino , Humanos , Masculino , Transplante Homólogo
7.
Int J Cancer ; 130(8): 1948-59, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21633954

RESUMO

Our study investigated the immunomodulatory effects of sunitinib to rationally design combinational platforms with immunotherapies for the treatment of solid tumors. Using a mouse model, we studied the effects of sunitinib given for 4 weeks at concentrations comparable to 37.5-50 mg/day in humans, followed by 2 weeks off the drug (sunitinib 4/2). We assessed the effect of differently timed combinations of sunitinib and a poxvirus-based vaccine encoding carcinoembryonic antigen (CEA) plus 3 costimulatory molecules on immune responses in CEA-transgenic (CEA-Tg) mice. Antitumor studies were performed in CEA-Tg mice bearing CEA-transfected MC38 murine colon carcinomas (MC38-CEA), treated either concurrently or sequentially with sunitinib and vaccine. In vitro, sunitinib inhibited PDGFR phosphorylation on MC38-CEA cells at concentrations similar to those biologically available during human treatment. In vivo, one cycle of sunitinib 4/2 caused bimodal immune effects: (a) decreased regulatory cells during the 4 weeks of treatment and (b) an immune-suppression rebound during the 2 weeks of treatment interruption. In a model using CEA-Tg mice bearing CEA(+) tumors, continuous sunitinib followed by vaccine increased intratumoral infiltration of antigen-specific T lymphocytes, decreased immunosuppressant T regulatory cells and myeloid-derived suppressor cells, reduced tumor volumes and increased survival. The immunomodulatory activity of continuous sunitinib administration can create a more immune-permissive environment. In combination with immunotherapies, sunitinib treatment should precede vaccine, to precondition the immune system, to maximize the response to vaccine-mediated immune enhancement.


Assuntos
Vacinas Anticâncer/imunologia , Sistema Imunitário/imunologia , Indóis/imunologia , Neoplasias Experimentais/imunologia , Pirróis/imunologia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/imunologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer/administração & dosagem , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/imunologia , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/imunologia , Neoplasias do Colo/terapia , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Citometria de Fluxo , Humanos , Sistema Imunitário/efeitos dos fármacos , Indóis/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Pirróis/administração & dosagem , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Sunitinibe , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Resultado do Tratamento
8.
Cancer ; 116(4 Suppl): 1018-26, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20127943

RESUMO

A large body of evidence suggests that the platelet-derived growth factor (PDGF) family and associated receptors are potential targets in oncology therapeutic development because of their critical roles in the proliferation and survival of various cancers and in the regulation and growth of the tumor stroma and blood vessels. Several small molecules that nonspecifically target the PDGF signaling axis are in current use or development as anticancer therapies. However, for the majority of these agents, PDGF and its receptors are neither the primary targets nor the principal mediators of anticancer activity. IMC-3G3, a fully human monoclonal antibody of the immunoglobulin G subclass 1, specifically binds to the human PDGF receptor alpha (PDGFRalpha) with high affinity and blocks PDGF ligand binding and PDGFRalpha activation. The results of preclinical studies and the frequent expression of PDGFRalpha in many types of cancer and in cancer-associated stroma support a rationale for the clinical development of IMC-3G3. Currently, IMC-3G3 is being evaluated in early clinical development for patients with several types of solid malignancies.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Animais , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Humanos , Imunoglobulina G/uso terapêutico , Camundongos , Neovascularização Patológica/tratamento farmacológico , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Receptores do Fator de Crescimento Derivado de Plaquetas/fisiologia
10.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 38(3): 504-8, 2007 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-17593843

RESUMO

OBJECTIVE: Through genetic engineering to produce the fusion protein of glutathione S-transferase (GST) linked to amino-terminal end of platelet-derived growth factor receptor (PDGFR), and to prepare the bioactive monoclonal antibody. METHODS: With taking GST-PDGFR-N fusion protein as immunogen, the anti-PDGFR monoclonal antibody was produced by using the hybridoma technique, of which then the antigen binding characteristic was identified by indirect enzyme-linked immunosorbent assay (ELISA), Western blot and immunohistochemistry methods. RESULTS: Two cell strains of hybridoma were obtained and named as 3B12F5 and 3C6H7C11 which secreted the anti-PDGFR monoclonal antibody, of which the class and subtype identification demonstrated both strains to produce all type of IgG1. The indirect ELISA result showed that the titers of ascites fluid which two hybridoma induced were 1 : 102400 and 1 : 25600. Western blot demonstrated that the two antibodies could recognize specifically the immunogen on PDGFR and U251 cell line. The cell immunohistochemistry proved that the antibody could recognize the expressed PDGFR antigens of neurogliocytoma U251 and bladder carcinoma BIU87 cell lines. CONCLUSION: We prepare successfully the PDGFR monoclonal antibody and provide a useful tool for researching on the PDGFR expression and clinical detection.


Assuntos
Anticorpos Monoclonais/análise , Anticorpos Monoclonais/genética , Engenharia Genética/métodos , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Animais , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Hibridomas , Imunoglobulina G/análise , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia
11.
Blood ; 110(1): 237-41, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17363728

RESUMO

Extensive chronic graft-versus-host disease (ecGVHD) is characterized by fibrosis similar to that of patients with systemic sclerosis (scleroderma). Since stimulatory autoantibodies against the platelet-derived growth factor (PDGF) receptor (PDGFR) have been found in patients with scleroderma and are responsible for the activation of skin fibroblasts, we tested the hypothesis that these autoantibodies are also present in patients affected by ecGVHD. Serum from 39 patients subjected to allogeneic stem cell transplantation for hematologic malignancies (22 with ecGVHD and 17 without cGVHD) and 20 healthy controls was assayed for the presence of stimulatory autoantibodies to the PDGFR by incubating purified IgG with mouse-embryo fibroblasts lacking PDGFR alpha or beta chains or with the same cells expressing PDGFR alpha. Stimulatory antibodies to the PDGFR were found selectively in all patients with ecGVHD but in none of the patients without cGVHD. Higher levels were detected in patients with generalized skin involvement and/or lung fibrosis. Antibodies recognized native PDGFR, induced tyrosine phosphorylation, accumulation of reactive oxygen species (ROS), and stimulated type 1 collagen gene expression through the Ha-Ras-ERK1/2-ROS signaling pathway. The biologic activity of these autoantibodies suggests a role in the development of fibrosis and argues for a common pathogenetic trait in ecGVDH and scleroderma phenotypes.


Assuntos
Autoanticorpos/imunologia , Doença Enxerto-Hospedeiro/imunologia , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Adolescente , Adulto , Idoso , Estudos de Casos e Controles , Doença Crônica , Feminino , Fibrose/etiologia , Fibrose/imunologia , Neoplasias Hematológicas/complicações , Neoplasias Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas , Humanos , Masculino , Pessoa de Meia-Idade , Transdução de Sinais , Transplante Homólogo
13.
N Engl J Med ; 354(25): 2667-76, 2006 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-16790699

RESUMO

BACKGROUND: Systemic sclerosis (scleroderma) is characterized by immunologic abnormalities, injury of endothelial cells, and tissue fibrosis. Abnormal oxidative stress has been documented in scleroderma and linked to fibroblast activation. Since platelet-derived growth factor (PDGF) stimulates the production of reactive oxygen species (ROS) and since IgG from patients with scleroderma reacts with human fibroblasts, we tested the hypothesis that patients with scleroderma have serum autoantibodies that stimulate the PDGF receptor (PDGFR), activating collagen-gene expression. METHODS: We analyzed serum from 46 patients with scleroderma and 75 controls, including patients with other autoimmune diseases, for stimulatory autoantibodies to PDGFR by measuring the production of ROS produced by the incubation of purified IgG with mouse-embryo fibroblasts carrying inactive copies of PDGFR alpha or beta chains or the same cells expressing PDGFR alpha or beta. Generation of ROS was assayed with and without specific PDGFR inhibitors. Antibodies were characterized by immunoprecipitation, immunoblotting, and absorption experiments. RESULTS: Stimulatory antibodies to the PDGFR were found in all the patients with scleroderma. The antibodies recognized native PDGFR, inducing tyrosine phosphorylation and ROS accumulation. Autoantibody activity was abolished by preincubation with cells expressing the PDGFR alpha chain or with recombinant PDGFR or by PDGFR tyrosine kinase inhibitors. Stimulatory PDGFR antibodies selectively induced the Ha-Ras-ERK1/2 and ROS cascades and stimulated type I collagen-gene expression and myofibroblast phenotype conversion in normal human primary fibroblasts. CONCLUSIONS: Stimulatory autoantibodies against PDGFR appear to be a specific hallmark of scleroderma. Their biologic activity on fibroblasts strongly suggests that they have a causal role in the pathogenesis of the disease.


Assuntos
Autoanticorpos/sangue , Doenças Autoimunes/imunologia , Imunoglobulina G/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Escleroderma Sistêmico/imunologia , Adulto , Idoso , Animais , Bioensaio , Estudos de Casos e Controles , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Expressão Gênica , Genes ras/fisiologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Transdução de Sinais , Tirfostinas/farmacologia
14.
Clin Exp Immunol ; 144(2): 335-41, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16634808

RESUMO

Receptors for platelet-derived growth factor (PDGF) are abundantly expressed on synovial fibroblast-like (SFL) cells from patients with rheumatoid arthritis (RA), and stimulation with PDGF enhances both the anchorage-dependent and -independent growth of RA-SFL cells. To elucidate the molecular mechanisms responsible for the excessive growth of RA-SFL cells and to seek a novel molecular-targeting therapy for RA, we examined the expression of adapter proteins and the effect of the specific inhibition of PDGF receptor activation by imatinib mesylate. Cultured SFL cells were used in the present study after 2-5 passages. The anchorage-dependent and -independent growth patterns of the SFL cells were evaluated using a tetrazolium-based assay and colony formation in 0.3% agar, respectively. Adapter proteins Gab1 and Gab2 were expressed in RA-SFL cells, and both proteins were rapidly (< 1 min) tyrosine-phosphorylated after the stimulation of RA-SFL cells with 10 ng/ml of PDGF and, to a lesser extent, after stimulation with 100 ng/ml of epidermal growth factor (EGF). The inhibition of PDGF receptor tyrosine kinase activation by 1 microM or less of imatinib mesylate specifically suppressed the PDGF-dependent, but not EGF-dependent, tyrosine phosphorylation of various proteins. Moreover, imatinib mesylate abolished both the anchorage-dependent and -independent proliferation of RA-SFL cells induced by PDGF stimulation. These results suggest that Gab adapter proteins are expressed and likely to be involved in the growth signalling of rheumatoid synovial cells and that imatinib mesylate, a key drug in the treatment of chronic myeloid leukaemia, may also be effective for the treatment of RA.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Artrite/imunologia , Fibroblastos/imunologia , Piperazinas/imunologia , Fator de Crescimento Derivado de Plaquetas/imunologia , Inibidores de Proteínas Quinases/imunologia , Pirimidinas/imunologia , Membrana Sinovial/imunologia , Artrite Reumatoide/imunologia , Benzamidas , Divisão Celular/imunologia , Células Cultivadas , Humanos , Mesilato de Imatinib , Osteoartrite/imunologia , Fosforilação , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Transdução de Sinais/imunologia , Membrana Sinovial/patologia , Tirosina/metabolismo
15.
Tidsskr Nor Laegeforen ; 125(22): 3115-9, 2005 Nov 17.
Artigo em Norueguês | MEDLINE | ID: mdl-16299568

RESUMO

BACKGROUND: Drugs that target tyrosine kinases belong to a new class of antineoplastic therapeutics, directed at cellular signalling mechanisms. Notes on the use of these agents and some of the clinical data are discussed in this review. METHODS: The article is based on a review of recent literature and the authors' personal experience. RESULTS AND INTERPRETATION: Receptor tyrosine kinases and intracellular tyrosine kinases regulate cellular events that may be involved in tumour development, such as proliferation, survival, and angiogenesis. Some of these agents are established in clinical practice, in particular the small-molecular tyrosine kinase inhibitor imatinib in the treatment of chronic myeloid leukaemia and gastrointestinal stromal tumours and the antibody trastuzumab in the treatment of breast cancer. Inhibitors of epidermal growth factor receptor (EGFR) and other tyrosine kinases are either established in clinical practice or under clinical investigation.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados , Antineoplásicos/administração & dosagem , Benzamidas , Cetuximab , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/imunologia , Cloridrato de Erlotinib , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Tumores do Estroma Gastrointestinal/enzimologia , Tumores do Estroma Gastrointestinal/imunologia , Gefitinibe , Humanos , Mesilato de Imatinib , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/enzimologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/imunologia , Neoplasias/enzimologia , Neoplasias/imunologia , Piperazinas/administração & dosagem , Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/imunologia , Pirimidinas/administração & dosagem , Pirimidinas/uso terapêutico , Quinazolinas/administração & dosagem , Quinazolinas/uso terapêutico , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/imunologia , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Transdução de Sinais/efeitos dos fármacos , Trastuzumab
16.
J Allergy Clin Immunol ; 115(3): 534-40, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15753901

RESUMO

BACKGROUND: Platelet-derived growth factor (PDGF) may have a significant role in airway remodeling in asthma, because it is a powerful inductor of many airway fibroblast activities such as collagen synthesis. OBJECTIVE: To determine whether PDGF is a significant contributor to airway remodeling in patients with asthma by enhancing airway fibroblast procollagen I expression. METHODS: Six normal controls without asthma, 10 subjects with mild to moderate asthma, and 5 subjects with severe asthma underwent bronchoscopy with endobronchial biopsy. Biopsies were placed in Dulbecco modified Eagle medium and fibroblasts cultured in the presence and absence of PDGF isoforms -AA, -BB, and -AB (1, 5, 10, 100 ng/mL) and insulin-like growth factor 1 (100 ng/mL). Fibroblast procollagen I and PDGF receptors (PDGFRs) alpha and beta expression were determined by ELISA. RESULTS: Platelet-derived growth factor BB significantly enhanced fibroblast procollagen I expression in patients with severe asthma compared with patients with mild/moderate asthma and normal controls. Furthermore, the baseline fibroblast expression of PDGFR-beta was significantly greater in patients with severe asthma compared with the other groups. CONCLUSION: This pilot study suggests that airway fibroblasts from patients with severe asthma exhibit a synthetic phenotype, which may be driven by the overexpression of PDGFR-beta.


Assuntos
Asma/genética , Fibroblastos/imunologia , Pulmão/imunologia , Fenótipo , Adulto , Asma/imunologia , Becaplermina , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Pulmão/metabolismo , Masculino , Projetos Piloto , Fator de Crescimento Derivado de Plaquetas/imunologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Pró-Colágeno/biossíntese , Proteínas Proto-Oncogênicas c-sis , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo
17.
Transplantation ; 79(2): 182-9, 2005 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-15665766

RESUMO

BACKGROUND: Perivascular inflammation and subsequent smooth muscle cell (SMC) proliferation are central in the development of cardiac allograft arteriosclerosis. We examined the effect of combined inhibition of proinflammatory vascular endothelial growth factor (VEGF) and SMC mitogen platelet-derived growth factor (PDGF) in rat cardiac allografts. METHODS: Heterotopic cardiac transplantations were performed between fully major histocompatibility mismatched rat strains receiving cyclosporine A immunosuppression. In situ hybridization and immunohistochemistry were performed to examine VEGF and PDGF ligand and receptor (R) expression. Protein tyrosine kinase inhibitors PTK787 and imatinib were used to inhibit VEGFR and PDGFR activity, respectively. Rat coronary artery SMC migration and proliferation assays were used to examine the effect of VEGF and PDGF and tyrosine kinase inhibitors in vitro. RESULTS: Both ligand and receptor expression of VEGF and PDGF were detected in chronically rejecting allografts. In vitro, PDGF-BB mediated rat coronary artery SMC migration and proliferation was completely inhibited with imatinib and partially with PTK787. In vivo, combined treatment with PTK787 and imatinib significantly reduced the formation of neointimal lesions in arteries of cardiac allografts at 8 weeks, producing a greater effect than either drug alone. PTK787, in contrast with imatinib, reduced the number of ED1 macrophages and PDGF-B immunoreactivity in the allografts at 4 weeks. CONCLUSIONS: Blocking VEGF and PDGF receptor signaling in cardiac allografts has distinctive effects on inflammation and SMC proliferation, suggesting that targeting both inflammation and pathologic vascular remodeling may be needed to inhibit cardiac allograft arteriosclerosis.


Assuntos
Transplante de Coração/imunologia , Fator de Crescimento Derivado de Plaquetas/genética , Fator A de Crescimento do Endotélio Vascular/genética , Inibidores da Angiogênese/uso terapêutico , Animais , Arteriosclerose/patologia , Arteriosclerose/prevenção & controle , Benzamidas , Rejeição de Enxerto/imunologia , Transplante de Coração/patologia , Mesilato de Imatinib , Inflamação , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Ftalazinas/uso terapêutico , Piperazinas/uso terapêutico , Complicações Pós-Operatórias/patologia , Piridinas/uso terapêutico , Pirimidinas/uso terapêutico , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos , Ratos Endogâmicos WF , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Transplante Homólogo/imunologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
18.
J Cell Sci ; 113 ( Pt 16): 2855-63, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10910770

RESUMO

In response to endovascular injury, platelet-derived growth factor-BB (PDGF-BB) and basic fibroblast growth factor (bFGF) are released locally and modulate vascular smooth muscle cells (SMC) proliferation and migration within the vascular wall. The aim of the present in vitro study was to determine how rat aorta SMC respond to the simultaneous exposure to PDGF-BB and bFGF. In a modified Boyden chamber assay bFGF exhibited a dose-dependent effect to inhibit the chemotactic action of PDGF-BB. A comparable result was observed in proliferation assays. In contrast, MIP-1 beta, epidermal growth factor (EGF), fibronectin and acidic FGF (aFGF) did not inhibit the chemotactic effect of PDGF-BB. Denatured bFGF did not exert an inhibitory effect and neutralizing antibodies either to bFGF or to bFGF-receptor abolished the inhibition observed in the presence of bFGF. The role played by PDGF receptor alpha (PDGF-Ralpha) was investigated in PDGF-Ralpha-dominant negative-transfected SMC, by selectively blocking PDGF-BB-binding to PDGF-Ralpha with neomycin, by neutralizing PDGF-Ralpha with a monoclonal antibody and by selectively stimulating PDGF-Ralpha with PDGF-AA; in all cases the effect of bFGF to inhibit PDGF-BB-directed SMC migration was abolished. These in vitro studies show that bFGF significantly inhibits PDGF-BB-induced SMC migration and proliferation and that this effect is mediated by both PDGF-Ralpha and bFGF receptor.


Assuntos
Quimiotaxia/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Músculo Liso Vascular/citologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Animais , Anticorpos/farmacologia , Aorta/citologia , Becaplermina , Divisão Celular/efeitos dos fármacos , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Neomicina/farmacologia , Testes de Neutralização , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Proto-Oncogênicas c-sis , Ratos , Ratos Wistar , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/fisiologia
19.
Am J Physiol ; 277(5): L975-82, 1999 11.
Artigo em Inglês | MEDLINE | ID: mdl-10564183

RESUMO

Respirable ambient particles [particulate matter <10 micrometer (PM(10))] are associated with both acute and chronic adverse health effects including chronic airflow obstruction. PM(10) can induce expression of inflammatory and fibrogenic mediators, but there is controversy about the types and/or sizes of particles involved and, in particular, whether ultrafine particles are the major toxic agents. To examine whether particle size affects mediator generation, we exposed rat tracheal explants, an inflammatory cell-free model of the airway wall, to various concentrations up to 500 microgram/cm(2) of fine (0.12 micrometer) or ultrafine (0.021 micrometer) titanium dioxide (anatase), maintained the explants in an organ culture in air for 1-7 days, and used RT-PCR to examine the expression of fibrogenic mediators and procollagen. No increase in gene expression was seen at 1 or 3 days, but at 5 days, ultrafine dust induced a small increase in procollagen. At 7 days, fine titanium dioxide produced significantly greater increases for platelet-derived growth factor (PDGF)-B, transforming growth factor-alpha, and transforming growth factor-beta compared with those by ultrafine dust; both dusts produced similar increases for PDGF-A; and ultrafine dust produced increases in procollagen expression, whereas fine dust had no effect. Expression levels were dose related. Both dusts produced a similar decrease in expression of PDGF receptor-alpha and a similar increase in PDGF receptor-beta. These observations suggest that ultrafine particles are intrinsically able to induce procollagen expression even in the absence of inflammatory cells; that chronic exposure to PM(10) may result in chronic airflow obstruction, in part because of ultrafine particle-mediated increases in airway wall fibrosis; and that chemically identical dusts of differing size can produce quite different patterns of gene expression in the airway wall. Differential upregulation of PDGF receptors does not appear to explain dust-induced fibrosis in this model.


Assuntos
Poluentes Atmosféricos/farmacologia , Corantes/farmacologia , Mediadores da Inflamação/imunologia , Titânio/farmacologia , Traqueia/patologia , Animais , Células Cultivadas , Primers do DNA , Densitometria , Etídio , Fibrose , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/imunologia , Indicadores e Reagentes , Pneumopatias/induzido quimicamente , Pneumopatias/imunologia , Pneumopatias/patologia , Masculino , Doenças Profissionais/induzido quimicamente , Doenças Profissionais/imunologia , Doenças Profissionais/patologia , Tamanho da Partícula , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/imunologia , Pró-Colágeno/genética , Pró-Colágeno/imunologia , Ratos , Ratos Sprague-Dawley , Receptores do Fator de Crescimento Derivado de Plaquetas/genética , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia , Traqueia/química , Traqueia/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
20.
Glycobiology ; 8(12): 1221-5, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9858644

RESUMO

Glycolipids expressed in the plasma membrane regulate a variety of cellular processes including intracellular calcium dynamics. We used flow cytometry to characterize the glycoconjugates on the plasma membrane of T51B liver epithelial cells. Antibodies against glycolipids found to be present were tested for their ability elevate intracellular calcium. An antibody against GM3 (DH2) nearly doubles intracellular calcium while an antibody against type II chains (1B2) increases calcium to nearly four times the baseline level, similar to levels obtained with epidermal growth factor (EGF). The antibodies stimulated calcium inflow but did not trigger calcium release from internal stores. In addition DH2 but not 1B2 inhibited platelet-derived growth factor beta receptor (PDGFbetar) function. This is the first demonstration of activation of calcium inflow by agents that bind GM3 and type II chains. The ganglioside-mediated calcium inflow is likely to stimulate secretion by these liver cells.


Assuntos
Anticorpos Anti-Idiotípicos/imunologia , Cálcio/metabolismo , Gangliosídeo G(M3)/imunologia , Fígado/imunologia , Fígado/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Células Cultivadas , Receptores ErbB/metabolismo , Citometria de Fluxo , Glicoconjugados/imunologia , Glicolipídeos/imunologia , Ratos , Receptor beta de Fator de Crescimento Derivado de Plaquetas , Receptores do Fator de Crescimento Derivado de Plaquetas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA