Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cancer Drug Resist ; 4(1): 223-232, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35582012

RESUMO

Aim: Ligand-targeted therapeutics are experiencing increasing use for treatment of human diseases due to their ability to concentrate a desired drug at a pathologic site while reducing accumulation in healthy tissues. For many ligand-targeted drug conjugates, a critical aspect of conjugate design lies in engineering release of the therapeutic payload to occur only after its internalization by targeted cells. Because disulfide bond reduction is frequently exploited to ensure intracellular drug release, an understanding of the redox properties of endocytic compartments can be critical to ligand-targeted drug design. While the redox properties of folate receptor trafficking endosomes have been previously reported, little is known about the trafficking of prostate-specific membrane antigen (PSMA), a receptor that is experiencing increasing use for drug targeting in humans. Methods: To obtain this information, we have constructed a PSMA-targeted fluorescence resonance energy transfer pair that reports on disulfide bond reduction by changing fluorescence from red to green. Results: We show here that this reporter exhibits rapid and selective uptake by PSMA-positive cells, and that reduction of its disulfide bond proceeds steadily but incompletely following internalization. The fact that maximal disulfide reduction reaches only ~50%, even after 24 h incubation, suggests that roughly half of the conjugates must traffic through endosomes that display no reducing capacity. Conclusion: As the level of disulfide reduction differs between PSMA trafficked and previously published folate trafficked conjugates, it also follows that not all internalizing receptors are translocated through similar intracellular compartments. Taken together, these data suggest that the efficiency of disulfide bond reduction must be independently analyzed for each receptor trafficking pathway when disulfide bond reduction is exploited for intracellular drug release.

2.
Arthritis Res Ther ; 21(1): 143, 2019 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-31174578

RESUMO

OBJECTIVES: Most therapies for autoimmune and inflammatory diseases either neutralize or suppress production of inflammatory cytokines produced by activated macrophages (e.g., TNFα, IL-1, IL-6, IL-17, GM-CSF). However, no approved therapies directly target this activated subset of macrophages. METHODS: First, we undertook to examine whether the folate receptor beta (FR-ß) positive subpopulation of macrophages, which marks the inflammatory subset in animal models of rheumatoid arthritis, might constitute the prominent population of macrophages in inflamed lesions in humans. Next, we utilized anti-FR-ß monoclonal antibodies capable of mediating antibody-dependent cell cytotoxicity (ADCC) to treat animal models of rheumatoid arthritis and peritonitis. RESULTS: Human tissue samples of rheumatoid arthritis, Crohn's disease, ulcerative colitis, idiopathic pulmonary fibrosis, nonspecific interstitial pneumonia, chronic obstructive pulmonary disease, systemic lupus erythematosus, psoriasis, and scleroderma are all characterized by dramatic accumulation of macrophages that express FR-ß, a protein not expressed on resting macrophages or any other healthy tissues. A monoclonal antibody to FR-ß accumulates specifically in inflamed lesions of murine inflammatory disease models and successfully treats such models of rheumatoid arthritis and peritonitis. More importantly, elimination of FR-ß-positive macrophages upon treatment with an anti-FR-ß monoclonal antibody promotes the departure of other immune cells, including T cells, B cells, neutrophils, and dendritic cells from the inflamed lesions. CONCLUSIONS: These data suggest that specific elimination of FR-ß-expressing macrophages may constitute a highly specific therapy for multiple autoimmune and inflammatory diseases and that a recently developed human anti-human FR-ß monoclonal antibody (m909) might contribute to suppression of this subpopulation of macrophages.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Artrite Reumatoide/imunologia , Receptor 2 de Folato/imunologia , Imunidade Celular , Ativação de Macrófagos , Macrófagos/metabolismo , Animais , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/patologia , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Macrófagos/imunologia , Macrófagos/patologia , Camundongos
3.
Biosci Trends ; 12(3): 298-308, 2018 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-29899195

RESUMO

Our goal is to develop a switch-controlled approach to enable better control of reactivity and safety of chimeric antigen receptor (CAR)-T therapy for non-small-cell lung cancer (NSCLC). Lentiviral transduction was performed to generate anti-FITC CAR-T cells and target cells stably expressing either isoform of the folate receptor. Colorimetric-based cytotoxic assay, enzyme-linked immunosorbent assay, and multiparametric flow cytometry analysis were used to evaluate the specificity and activity of CAR-T cells in vitro. Human primary T cells stably expressing the fully human anti-FITC CAR were generated. Anti-FITC CAR-T cells displayed antigen-specific and folate-FTIC dependent reactivity against engineered A549-FRα and THP-1-FRß. The selective activation and proliferation of anti-FITC CAR-T cells in vitro stringently relied on the co-existence of folate-FITC and FR- expressing target cells and was dose-titratable with the folate-FITC switch. The excellent in vitro efficacy and specificity of an adaptor-controlled CAR-T therapy to target both tumor cells and tumor-associated macrophages in NSCLCs were validated.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/terapia , Neoplasias Pulmonares/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/transplante , Células A549 , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Citometria de Fluxo , Receptor 1 de Folato/imunologia , Receptor 1 de Folato/metabolismo , Células HEK293 , Células HL-60 , Humanos , Células Jurkat , Lentivirus/genética , Ligantes , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Macrófagos/imunologia , Linfócitos T/imunologia , Transdução Genética , Microambiente Tumoral/imunologia
4.
Leuk Lymphoma ; 59(12): 2982-2989, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29616859

RESUMO

Receptor-targeted delivery of imaging and therapeutic agents has emerged as an attractive strategy to diagnosis and treat many diseases including cancer. One of the most well-studied receptors for targeted therapies is the folate receptor (FR) family. FR-α and FR-ß are present on many cancers with little expression in normal tissues; leading to the testing of at least six folate-targeted drugs in human clinical trials for various cancers. However, the expression of FR in blood cancers has not been fully explored with no reports of FR expression in myelomas. Herein, we report the expression of both FR-α and FR-ß on CD138 + plasma cells isolated from patients with multiple myeloma. In addition, all-trans retinoic acid was shown to increase the levels of FR-α and FR-ß in two myeloma cell lines. Altogether, this data suggests that folate-targeted therapies for the treatment of multiple myeloma warrants further investigation.


Assuntos
Antineoplásicos/farmacologia , Receptor 1 de Folato/metabolismo , Receptor 2 de Folato/metabolismo , Mieloma Múltiplo/patologia , Plasmócitos/efeitos dos fármacos , Tretinoína/farmacologia , Antineoplásicos/uso terapêutico , Medula Óssea/patologia , Linhagem Celular Tumoral , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Mieloma Múltiplo/tratamento farmacológico , Plasmócitos/metabolismo , Sindecana-1/metabolismo , Regulação para Cima/efeitos dos fármacos
5.
Oncotarget ; 9(4): 4485-4495, 2018 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-29435118

RESUMO

A number of folate receptor (FR) targeted small molecular drugs and monoclonal antibodies have been introduced into clinical trials to treat FR positive cancers. Because the therapeutic efficacy of these drugs depends prominently on the level of FR-α expression on the cancer cells, patients have been commonly selected for FR-targeted therapies based on the intensity of a folate-targeted radioimaging agent. Unfortunately, uptake of such imaging agents can be mediated by both major isoforms of the folate receptor, FR-α and FR-ß. Logically, if the FR positive cell population in a tumor mass is dominated by FR-ß positive macrophages, patients could be selected for therapy that have few FR-expressing cancer cells. Although several IHC studies have examined expression of either FR-α or FR-ß, no study to date has investigated expression of both FR-α and FR-ß in the same tumor mass. Herein, we utilize monoclonal antibodies specific for FR-α (mAb343) and FR-ß (m909) to query each isoform's expression in a range of cancers. We show that lung and pancreatic adenocarcinomas express the full spectrum of FR-α and FR-ß combinations with ~76% of lung adenocarcinomas expressing both FR-α and FR-ß while pancreatic cancers express primarily FR-ß. Thus, while folate-targeted imaging of lung cancer patients might accurately reflect the expression of FR-α on lung cancer cells, imaging of pancreatic cancer patients could mislead a physician into treating a nonresponding patient. Overall, these data suggest that an independent analysis of both FR-α and FR-ß should be obtained to predict the potential efficacy of a folate-targeted drug.

6.
Cell Mol Immunol ; 14(9): 783-791, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27665946

RESUMO

Interleukin-37 (IL-37) is an inhibitory member of the IL-1 family of cytokines. We previously found that balanced selection maintains common variations of the human IL37 gene. However, the functional consequences of this selection have yet to be validated. Here, using cells expressing exogenous IL-37 variants, including IL-37 Ref and IL-37 Var1 and Var2, we found that the three variants of IL-37 exhibited different immunoregulatory potencies in response to immune stimulation. The protein level of IL-37 Var2 was found to be significantly less than that of IL-37 Ref or Var1, despite the comparable mRNA levels of all three variants. Further study showed that IL-37 Var2 was rapidly degraded by a proteasome-dependent mechanism mediated by enhanced polyubiquitination, leading to a transient upregulation of IL-37 Var2 after immune stimulation. Finally, when ectopically expressed in cells, human IL-37 Var2 exerted less inhibition on proinflammatory cytokine production than did other IL-37 variants. Conversely, purified extracellular IL-37 variant proteins demonstrated comparable inhibitory abilities in vitro. In conclusion, our study reveals that common genetic variants of IL37 lead to different immune-inhibitory potencies, primarily as a result of differences in IL-37 protein stability, suggesting the possible involvement of these variants in various human diseases.


Assuntos
Interleucina-1/genética , Interleucina-1/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Heterogeneidade Genética , Heterozigoto , Humanos , Interleucina-1/química , Interleucina-6/metabolismo , Fenótipo , Poliubiquitina/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Ubiquitinação , Regulação para Cima
7.
Mol Ther ; 24(12): 2078-2089, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27731313

RESUMO

Phosphodiesterase 4 (PDE4) inhibitors are approved for the treatment of some moderate to severe inflammatory conditions. However, dose-limiting side effects in the central nervous system and gastrointestinal tract, including nausea, emesis, headache, and diarrhea, have impeded the broader therapeutic application of PDE4 inhibitors. We sought to exploit the wealth of validation surrounding PDE4 inhibition by improving the therapeutic index through generation of an antibody-drug conjugate (ADC) that selectively targets immune cells through the CD11a antigen. The resulting ADC consisted of a human αCD11a antibody (based on efalizumab clone hu1124) conjugated to an analog of the highly potent PDE4 inhibitor GSK256066. Both the human αCD11a ADC and a mouse surrogate αCD11a ADC (based on the M17 clone) rapidly internalized into immune cells and suppressed lipololysaccharide (LPS)-induced TNFα secretion in primary human monocytes and mouse peritoneal cells, respectively. In a carrageenan-induced air pouch inflammation mouse model, treatment with the ADC significantly reduced inflammatory cytokine production in the air pouch exudate. Overall, these results provide compelling evidence for the feasibility of delivering drugs with anti-inflammatory activity selectively to the immune compartment via CD11a and the development of tissue-targeted PDE4 inhibitors as a promising therapeutic modality for treating inflammatory diseases.


Assuntos
Aminoquinolinas/metabolismo , Antígenos CD11/metabolismo , Imunoconjugados/administração & dosagem , Inflamação/imunologia , Inibidores da Fosfodiesterase 4/metabolismo , Sulfonas/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imunoconjugados/farmacologia , Lipopolissacarídeos/efeitos adversos , Camundongos , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Peritônio/efeitos dos fármacos , Peritônio/imunologia , Fator de Necrose Tumoral alfa/metabolismo
8.
Medicine (Baltimore) ; 95(21): e3802, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27227959

RESUMO

To investigate the frequency and the spectrum of major opportunistic infections (OIs), evaluate the major clinical factors associated with each specific OI, and identify the risk factors for in-hospital death among HIV patients in East China.A retrospective cohort study was made including all the HIV-infected patients who were admitted for the first time to the Shanghai Public Health Clinical Center during June 1, 2013 to June 1, 2015. The demographic and clinical data were collected. Comparison of continuous variables was analyzed by one-way ANOVA and rank sum test. Person χ test and Fisher exact test were applied to analyze the categorical variables. A Cox proportional hazards regression model was used to determine the risk for the occurrence of in-hospital death.In total, 920 patients were enrolled with age of 41.59 ± 13.36 years and 91% male. Median CD4 was 34 (IQR, 13-94) cells/µL. Among these patients, 94.7% acquired OIs while the rest developed malignancies. Pneumocystis pneumonia and bacterial coinfection (42.1%) was found to be the most common OIs, followed by tuberculosis (31.4%), CMV (20.9%), Cryptococcosis (9.0%), and MAC infection (5.2%). Of the above 5 major OIs, CMV-infected patients had the lowest median CD4 cell count 22.50 (IQR, 7.50-82.00) while the patients with tuberculosis infection had the highest count 61.00 (IQR, 27.00-176.00). In-hospital death rate was 4.2 per 100 person-years among these patients. Of note, admitted patients with 2 types of OIs (2.20, 95% CI 1.39-3.48) and those patients who were 40-year old or older (1.75, 95% CI 1.10-2.78) had a higher risk of such death.Pneumocystis pneumonia and tuberculosis were still the leading causes for the admission of HIV-infected patients in East China, and these patients tended to have very low CD4 cell counts. It is believed that expanding the HIV screening test and pushing the infected ones get ART earlier is required for generating a more successful HIV management strategy.


Assuntos
Infecções Oportunistas Relacionadas com a AIDS/mortalidade , Síndrome da Imunodeficiência Adquirida/complicações , Adulto , Contagem de Linfócito CD4 , China/epidemiologia , Coinfecção , Feminino , Infecções por HIV/complicações , Mortalidade Hospitalar , Humanos , Masculino , Pessoa de Meia-Idade , Pneumonia por Pneumocystis/complicações , Estudos Retrospectivos , Fatores de Risco , Tuberculose/complicações
9.
Proc Natl Acad Sci U S A ; 113(4): E459-68, 2016 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-26759369

RESUMO

Chimeric antigen receptor T (CAR-T) cell therapy has produced impressive results in clinical trials for B-cell malignancies. However, safety concerns related to the inability to control CAR-T cells once infused into the patient remain a significant challenge. Here we report the engineering of recombinant antibody-based bifunctional switches that consist of a tumor antigen-specific Fab molecule engrafted with a peptide neo-epitope, which is bound exclusively by a peptide-specific switchable CAR-T cell (sCAR-T). The switch redirects the activity of the bio-orthogonal sCAR-T cells through the selective formation of immunological synapses, in which the sCAR-T cell, switch, and target cell interact in a structurally defined and temporally controlled manner. Optimized switches specific for CD19 controlled the activity, tissue-homing, cytokine release, and phenotype of sCAR-T cells in a dose-titratable manner in a Nalm-6 xenograft rodent model of B-cell leukemia. The sCAR-T-cell dosing regimen could be tuned to provide efficacy comparable to the corresponding conventional CART-19, but with lower cytokine levels, thereby offering a method of mitigating cytokine release syndrome in clinical translation. Furthermore, we demonstrate that this methodology is readily adaptable to targeting CD20 on cancer cells using the same sCAR-T cell, suggesting that this approach may be broadly applicable to heterogeneous and resistant tumor populations, as well as other liquid and solid tumor antigens.


Assuntos
Antígenos CD19/imunologia , Antígenos de Neoplasias/imunologia , Imunoterapia Adotiva/métodos , Leucemia de Células B/terapia , Receptores de Antígenos de Linfócitos T/imunologia , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Especificidade do Receptor de Antígeno de Linfócitos T , Subpopulações de Linfócitos T/imunologia , Animais , Azidas , Linfócitos B/imunologia , Linfócitos B/patologia , Fatores de Transcrição de Zíper de Leucina Básica/imunologia , Linhagem Celular Tumoral , Citocinas/metabolismo , Citotoxicidade Imunológica , Relação Dose-Resposta Imunológica , Feminino , Genes Reporter , Vetores Genéticos , Humanos , Imunoterapia Adotiva/efeitos adversos , Ativação Linfocitária , Linfopenia/etiologia , Linfopenia/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Fenilalanina/análogos & derivados , Engenharia de Proteínas/métodos , Receptores de Antígenos de Linfócitos T/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas de Saccharomyces cerevisiae/imunologia , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Relação Estrutura-Atividade , Subpopulações de Linfócitos T/transplante , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Oncotarget ; 6(16): 14700-9, 2015 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-25909292

RESUMO

Over-expression of folate receptor alpha on cancer cells has been frequently exploited for delivery of folate-targeted imaging and therapeutic agents to tumors. Because limited information exists on expression of the beta isoform of the folate receptor in human cancers (FR-ß), we have evaluated the immunohistochemical staining pattern of FR-ß in 992 tumor sections from 20 different human cancer types using a new anti-human FR-ß monoclonal antibody. FR-ß expression was shown to be more pronounced in cells within the stroma, primarily macrophages and macrophage-like cells than cancer cells in every cancer type studied. Moreover, FR-ß expression in both cancer and stromal cells was found to be statistically more prominent in females than males. A significant positive correlation was also observed between FR-ß expression on stromal cells and both the stage of the cancer and the presence of lymph node metastases. Based on these data we conclude FR-ß may constitute a good target for specific delivery of therapeutic agents to activated macrophages and that accumulation of FR-ß positive macrophages in the stroma could serve as a useful indicator of a tumor's metastatic potential.


Assuntos
Receptor 2 de Folato/metabolismo , Macrófagos/metabolismo , Neoplasias/metabolismo , Feminino , Humanos , Masculino
11.
J Leukoc Biol ; 96(4): 563-70, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25015955

RESUMO

Activated macrophages are commonly involved in the pathogenesis of inflammatory and autoimmune diseases and have been frequently reported to overexpress FR-ß. Although FR-targeted therapies aimed at eliminating activated macrophages have shown promise for treating inflammatory diseases, little work has been performed to evaluate whether other hematopoietic cells might also express FR-ß. Analysis of peripheral blood cells with a mAb to human FR-ß reveals that only monocytes express FR-ß. Molecular characterization of these circulating monocytes further demonstrates that solely the classic/proinflammatory subset (CD14(high)CD16(-)) expresses the FR and that only CD14(high)CD16(-) FR-ß(+) monocytes also display the ability to bind folate-linked molecules. Confirmation that this subset of monocytes indeed constitutes the proinflammatory subpopulation was obtained by demonstrating coexpression of FR-ß with other proinflammatory markers, including CCR2 and HLA-DR. Synovial monocytes from the joints of patients with RA were also shown to express FR-ß. As inhibition of the chemotaxis of proinflammatory monocytes into sites of inflammation has been explored frequently as a means of controlling autoimmune diseases, demonstration that FR-ß is uniquely expressed on this proinflammatory subpopulation offers a new strategy to suppress migration of inflammatory monocytes into sites of inflammation.


Assuntos
Receptor 2 de Folato/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Biomarcadores/metabolismo , Humanos , Imunofenotipagem , Imunoterapia , Receptores de Lipopolissacarídeos/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/terapia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Fenótipo , Líquido Sinovial/imunologia , Líquido Sinovial/metabolismo
12.
Laryngoscope ; 124(8): E312-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24448885

RESUMO

OBJECTIVES/HYPOTHESIS: Folate receptor (FR) expression, although known to be elevated in many types of cancer and inflammatory cells, has not been well characterized in head and neck squamous cell carcinoma (HNSCC). We hypothesized that tumor infiltrating inflammatory cells expressing FR-ß could allow fluorescent visualization of HNSCC tumors using folate conjugated dyes even when FR expression in cancer cells is low. STUDY DESIGN: Retrospective review of clinical pathologic specimens and in vivo animal study. METHODS: A tissue microarray with tumor and tumor-free tissue from 22 patients with HNSCC was stained with antibodies to FR-α and FR-ß. We characterized FR-ß(+) cells by examining CD45, CD68, CD206, and transforming growth factor (TGF)-ß expression. To investigate fluorescent imaging, mice with orthotopic tumor xenografts were imaged in vivo after intravenous injections of folate conjugated fluorescein isothiocyanate (folate-FITC) and were histologically evaluated ex vivo. RESULTS: All tumor samples demonstrated significant FR-ß staining and negligible FR-α staining. FR-ß(+) cells found in tumors coexpressed CD68 and had increased expression of CD206 and TGF-ß characteristic of tumor-associated macrophages. In the xenograft models, tumors showed strong in vivo fluorescence after folate-FITC injection in contrast to surrounding normal tissues. Histologic examination of the xenograft tissue similarly showed folate-FITC uptake in areas of inflammatory cellular infiltrate. CONCLUSIONS: Although HNSCC tumor cells do not express FR, HNSCC tumors contain a significant population of FR-ß-expressing macrophages. Folate conjugated fluorescent dye is able to specifically target and label tumor xenografts to permit macroscopic fluorescence imaging due to FR-ß expression on the infiltrating inflammatory cells.


Assuntos
Carcinoma de Células Escamosas/patologia , Receptor 2 de Folato/análise , Neoplasias de Cabeça e Pescoço/patologia , Proteínas de Neoplasias/análise , Imagem Óptica , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Carcinoma de Células Escamosas/química , Feminino , Receptor 1 de Folato , Neoplasias de Cabeça e Pescoço/química , Humanos , Camundongos , Pessoa de Meia-Idade , Estudos Retrospectivos , Carcinoma de Células Escamosas de Cabeça e Pescoço
13.
Mol Pharm ; 10(5): 1918-27, 2013 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-23641923

RESUMO

Pro-inflammatory macrophages play a prominent role in such autoimmune diseases as rheumatoid arthritis, Crohn's disease, psoriasis, sarcoidosis, and atherosclerosis. Because pro-inflammatory macrophages have also been shown to overexpress a receptor for the vitamin folic acid (i.e., folate receptor beta; FR-ß), folate-linked drugs have been explored for use in imaging and treatment of these same diseases. To determine whether allergic inflammatory disorders might be similarly targeted with folate-linked drugs, we have examined the characteristics of macrophages that are prominent in the pathogenesis of asthma. We report here that macrophages from the lungs of mice with experimental allergic asthma express FR-ß. We further document that these FR-ß(+) macrophages coexpress markers of alternatively activated (M2-type) macrophages, including the mannose receptor and arginase-1. Finally, we demonstrate that folate-conjugated fluorescent dyes and radioimaging agents can be specifically targeted to these asthmatic lung macrophages, with little uptake by macrophages present in healthy lung tissue. These data suggest strategies for the development of novel diagnostic agents for the imaging of asthma and other diseases involving alternatively activated macrophages.


Assuntos
Asma/diagnóstico , Asma/imunologia , Ácido Fólico , Ativação de Macrófagos , Animais , Arginase/metabolismo , Asma/diagnóstico por imagem , Diagnóstico por Imagem/métodos , Modelos Animais de Doenças , Feminino , Corantes Fluorescentes , Receptor 2 de Folato/metabolismo , Ácido Fólico/análogos & derivados , Ácido Fólico/metabolismo , Humanos , Lectinas Tipo C/metabolismo , Pulmão/diagnóstico por imagem , Pulmão/imunologia , Pulmão/metabolismo , Macrófagos/diagnóstico por imagem , Macrófagos/imunologia , Macrófagos/metabolismo , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Oligopeptídeos , Cintilografia , Compostos Radiofarmacêuticos , Receptores de Superfície Celular/metabolismo , Tecnécio
14.
Arthritis Res Ther ; 13(2): R59, 2011 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-21477314

RESUMO

INTRODUCTION: Folate receptor beta (FRß) is only detectable in placenta and limited to some hematopoietic cells of myeloid lineage in healthy people. Studies have indicated that FRß is over-expressed in activated macrophages in autoimmune diseases and some cancer cells. In this study we aimed to develop an FRß-specific human monoclonal antibody (mAb) that could be used as a therapeutic agent to treat rheumatoid arthritis and other autoimmune diseases, as well as FRß positive cancers. METHODS: Functional recombinant FRß protein was produced in insect cells and used as antigen to isolate a mAb, m909, from a human naïve Fab phage display library. Binding of Fab and IgG1 m909 to FRß was measured by ELISA, surface plasmon resonance, immune fluorescence staining, and flow cytometry. Antibody-dependent cell-mediated cytotoxicity (ADCC) was evaluated with FRß positive CHO cells as target cells and isolated peripheral blood monocytes as effector cells in an in vitro assay. RESULTS: Fab m909 bound with relatively high affinity (equilibrium dissociation constant 57 nM) to FRß. The IgG1 m909 showed much higher (femtomolar) avidity as measured by ELISA, and it bound to FRß positive cells in a dose-dependent manner, but not to parental FRß negative cells. m909 did not compete with folate for the binding to FRß on cells. m909 was not only able to select FRß positive, activated macrophages from synovial fluid cells of arthritis patients as efficiently as folate, but also able to mediate ADCC in FRß positive cells. CONCLUSIONS: Unlike folate-drug conjugates, m909 selectively binds to FRß, does not recognize FRα, and has at least one effector function. m909 alone has potential to eliminate FRß positive cells. Because m909 does not compete with folate for receptor binding, it can be used with folate-drug conjugates in a combination therapy. m909 can also be a valuable research reagent.


Assuntos
Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Artrite Reumatoide/imunologia , Receptor 2 de Folato/imunologia , Macrófagos/imunologia , Separação Celular , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunofluorescência , Humanos , Ativação de Macrófagos/imunologia , Microscopia Confocal , Biblioteca de Peptídeos , Proteínas Recombinantes/imunologia , Ressonância de Plasmônio de Superfície
15.
Cell Signal ; 21(1): 143-50, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18950704

RESUMO

Heat shock protein 27 (HSP27) is an ubiquitiously expressed protein, which has been mediated in various biological functions. Here, we present a novel mechanism utilized by HSP27 in regulating IL-1beta induced NF-small ka, CyrillicB activation. Both over-expression and RNAi experiments indicate that HSP27 physically interacts with tumor necrosis factor receptor-associated factor 6 (TRAF6) and promotes TRAF6 ubiquitination. Over-expressed HSP27 augments IL-1beta induced TRAF6 ubiquitination and Ismall ka, CyrillicB kinase (IKK) activation. On the other hand, IL-1beta stimulation reduces endogenous HSP27/TRAF6 association, but inhibiting HSP27 phosphorylation by using SB202190, an inhibitor of p38, and MAPKAPK2 RNAi increases HSP27/TRAF6 association and thereby enhances TRAF6 ubiquitination, IKK phosphorylation as well as NF-small ka, CyrillicB activation. Furthermore, co-transfection study shows that HSP27 S78/82A, two phosphorylated serine site deficient mutants, but not wild-type HSP27 (HSP27 WT) and HSP27 S15A mutant increases TRAF6 ubiquitination and thereby mediates IL-1beta triggered IKK phosphorylation. Taken together, our data indicate that HSP27 regulates IL-1beta triggered NF-small ka, CyrillicB activation via a feedback loop which includes the interaction between HSP27 phosphorylation and ability of HSP27 to bind with TRAF6. The findings of this study reveal a novel mechanism by which HSP27 controls cytokine stimulation.


Assuntos
Proteínas de Choque Térmico HSP27/metabolismo , Quinase I-kappa B/metabolismo , Interleucina-1beta/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Ubiquitinação , Linhagem Celular , Proteínas de Choque Térmico HSP27/genética , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mutagênese Sítio-Dirigida , NF-kappa B/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Mol Immunol ; 46(4): 541-50, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18950863

RESUMO

Heat shock protein 90 (Hsp90) is an abundantly and ubiquitously expressed chaperone with majority of client proteins which act as signal molecules. Transforming growth factor beta-activated kinase 1 (TAK1) is a mitogen-activated protein kinase kinase kinase (MAPKKK), and is essential in interleukin-1beta (IL-1beta) triggered signaling pathways. In the present study, we found that Hsp90 plays an important role in regulating IL-1beta signaling by keeping TAK1 stability. The results showed that the specific inhibitor geldanamycin (GA) of Hsp90 dramatically inhibited IL-1beta stimulated TAK1-MAPKs and TAK1-nuclear factor-kappaB (NF-kappaB) activation, resulting in the decrease of cyclooxygenase-2 (COX-2) protein expression. Silencing Hsp90 expression through RNA interference (RNAi) also down-regulated TAK1, as well as attenuated IL-1beta induced phosphorylation of c-Jun NH(2)-terminal kinase (JNK) and p38 MAPKs, and degradation of IkappaBalpha. The same results were obtained in T6RZC stable cells which initiated IL-1beta-induced cell signaling at the level of the oligomerization and ubquitination of TNF receptor-associated factor 6 (TRAF6). We further found that Hsp90 formed a complex with TAK1 via its N-terminal domain and GA destabilized TAK1 and induced TAK1 degradation through proteasome pathway. Taken together our results demonstrate that Hsp90 regulates IL-1beta-induced signaling by interacting with TAK1 and maintaining the stability of TAK1, suggesting that Hsp90 might act as the chaperone of TAK1 in immune and inflammatory responses related with IL-1 signal cascades.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Interleucina-1beta/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Benzoquinonas/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/imunologia , Inibidores Enzimáticos/farmacologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/imunologia , Células HeLa , Humanos , Interleucina-1beta/imunologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lactamas Macrocíclicas/farmacologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/imunologia , NF-kappa B/antagonistas & inibidores , NF-kappa B/imunologia , NF-kappa B/metabolismo , Complexo de Endopeptidases do Proteassoma/imunologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Estabilidade Proteica , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Fator 6 Associado a Receptor de TNF/antagonistas & inibidores , Fator 6 Associado a Receptor de TNF/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
Mol Immunol ; 46(5): 848-57, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18962899

RESUMO

We have reported that intracellular glutathione S-transferases P1 (GSTP1) suppresses LPS (lipopolysaccharide)-induced excessive production of pro-inflammatory factors by inhibiting LPS-stimulated MAPKs (mitogen-activated protein kinases) as well as NF-kappaB activation. But under pathogenic circumstances, physiologic levels of GSTP1 are insufficient to stem pro-inflammatory signaling. Here we show that LPS-induced up-regulation of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in RAW246.7 cells is significantly reduced by incubating cells with recombinant GSTP1 protein. In vivo study demonstrates that treatment of mice (i.p.) with recombinant GSTP1 protein effectively suppresses the devastating effects of acute inflammation, which includes reduction of mortality rate of endotoxic shock, alleviation of LPS-induced acute lung injury and abrogation of thioglycolate (TG)-induced peritoneal deposition of leukocytes and polymorphonuclear cells (PMNs). Meanwhile, GSTP1 prevented LPS-induced TNF-alpha, IL-1beta, MCP-1 and NO production. Further investigation by using confocal microscopy and flow cytometry shows that recombinant GSTP1 protein can be delivered into RAW246.7 cells, mouse peritoneal macrophages and HEK 293 cells suggesting that extracellular GSTP1 protein could be transported across plasma membrane and act as a cytosolic protein. In conclusion our research demonstrates a new finding that increasing cellular GSTP1 level by supplement of recombinant GSTP1 effectively suppresses the devastating effects of acute inflammation.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Glutationa S-Transferase pi/farmacologia , Choque Séptico/tratamento farmacológico , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/imunologia , Animais , Linhagem Celular , Quimiocina CCL2/imunologia , Glutationa S-Transferase pi/imunologia , Humanos , Inflamação/dietoterapia , Inflamação/imunologia , Interleucina-1beta/imunologia , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/imunologia , Óxido Nítrico/imunologia , Óxido Nítrico Sintase Tipo II/imunologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/imunologia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Choque Séptico/induzido quimicamente , Choque Séptico/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/imunologia
18.
Protein J ; 26(6): 359-70, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17587159

RESUMO

The bacterial expression and purification of human glutathione S-transferase P1-1(hGST P1-1), as a hexahistidine-tagged polypeptide was performed. Site-directed mutagenesis was used to construct mutants in which alanine replaced two (C47A/C101A), three (C14A/C47A/C101A) or all four (C14A/C47A/ C101A/C169A) cysteine residues using the plasmid for the wild type enzyme. Analysis of their catalytic activities and kinetic parameters suggested that cysteins are not essential for the catalytic activity but may contribute to some extent to the catalytic efficiency. Moreover, on SDS-polyacrylamide gel electrophoresis (SDS-PAGE) under nonreducing conditions, hexahistidine-tagged hGST P1-1 (His(6)-hGST P1-1) treated with 1 mM H(2)O(2) showed at least three extra bands, in addition to the native His(6)-hGST P1-1 subunit band. These extra bands were not detected in the cysteinyl mutants. Thus, it indicated that disulfide bonds were formed mainly within subunits between cysteine residues, causing an apparent reduction in molecular weight, only small amounts of binding between subunits being observed.


Assuntos
Cisteína/química , Glutationa Transferase/metabolismo , Histidina/química , Oligopeptídeos/química , Sequência de Bases , Catálise , Dicroísmo Circular , Primers do DNA , DNA Complementar , Eletroforese em Gel de Poliacrilamida , Fluorescência , Glutationa Transferase/antagonistas & inibidores , Glutationa Transferase/química , Glutationa Transferase/genética , Humanos , Cinética , Mutagênese Sítio-Dirigida , Oxirredução , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
Appl Microbiol Biotechnol ; 76(1): 141-9, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17562041

RESUMO

The extracellular portion (amino acids 95-281 or 114-281) of the human tumor necrosis factor-related apoptosis-inducing ligand (sTRAIL) was genetically linked to the C terminus of the fluoresce-enhanced green fluorescent protein variant (EGFP) to generate two versions of EGFP-sTRAIL fusion proteins, designated EGFP-sTR95 and EGFP-sTR114, respectively. The two versions of EGFP-sTRAIL fusion proteins both induce extensive apoptosis in lymphoid as well as nonlymphoid tumor cell lines. In addition, the two versions of fusion proteins retain similar fluorescence spectra to those of EGFP and have shown the specific binding to TRAIL receptor-positive cells; thus, the stained cells could be analyzed with flow cytometry. Hence, the two versions of fusion proteins represent a readily obtainable source of biologically active sTRAIL that may prove useful in exploit fully the characteristics of both the soluble TRAIL and its receptor system.


Assuntos
Proteínas de Fluorescência Verde/biossíntese , Engenharia de Proteínas , Proteínas Recombinantes de Fusão/biossíntese , Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Apoptose , Escherichia coli/metabolismo , Proteínas de Fluorescência Verde/genética , Células HeLa , Humanos , Células Jurkat/efeitos dos fármacos , Células Jurkat/fisiologia , Ligação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Solubilidade , Ligante Indutor de Apoptose Relacionado a TNF/genética
20.
Appl Microbiol Biotechnol ; 73(5): 1114-22, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17021878

RESUMO

A fusion protein of enhanced green fluorescent protein (EGFP) and soluble domain of human a proliferation-inducing ligand (sAPRIL) was efficiently expressed in Escherichia coli BL 21 (DE3). The soluble EGFP/sAPRIL, around 43 kDa, was purified in milligram amounts using metal chellate affinity chromatography and detected with anti-His(6) and anti-hsAPRIL monoclonal antibody. The chimeric protein exhibited similar fluorescence spectra with free EGFP. In vitro, purified EGFP/sAPRIL specifically bound receptor B cell maturation antigen (BCMA) detected by enzyme linked immunosorbent assay (ELISA) and receptors [including heparan sulfate proteoglycan (HSPGs)]-positive cell lines analyzed by fluorescence-activated cell sorting (FACS). Confocal laser microscopy images visibly showed the HSPGs'-dependent binding of EGFP/sAPRIL to NIH-3T3 cell. In addition, the chimera retained the bioactivity to stimulate/co-stimulate proliferation of NIH-3T3 and Jurkat cell/human B cell in vitro. Therefore, the fusion protein shows a readily obtainable source of biologically active sAPRIL which has considerable potential for single-step fluorescence detection assay in the study of APRIL and its receptors.


Assuntos
Proteínas de Fluorescência Verde/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Antígeno de Maturação de Linfócitos B/metabolismo , Células Cultivadas , Cromatografia de Afinidade , Ensaio de Imunoadsorção Enzimática , Escherichia coli/genética , Citometria de Fluxo , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteoglicanas de Heparan Sulfato/metabolismo , Humanos , Células Jurkat , Leucócitos Mononucleares , Ativação Linfocitária , Camundongos , Microscopia Confocal , Células NIH 3T3 , Ligação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Espectrometria de Fluorescência , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA