Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
4.
Methods ; 87: 3-10, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25766927

RESUMO

Viable tumor cells actively release vesicles into the peripheral circulation and other biologic fluids, which exhibit proteins and RNAs characteristic of that cell. Our group demonstrated the presence of these extracellular vesicles of tumor origin within the peripheral circulation of cancer patients and proposed their utility for diagnosing the presence of tumors and monitoring their response to therapy in the 1970s. However, it has only been in the past 10 years that these vesicles have garnered interest based on the recognition that they serve as essential vehicles for intercellular communication, are key determinants of the immunosuppressive microenvironment observed in cancer and provide stability to tumor-derived components that can serve as diagnostic biomarkers. To date, the clinical utility of extracellular vesicles has been hampered by issues with nomenclature and methods of isolation. The term "exosomes" was introduced in 1981 to denote any nanometer-sized vesicles released outside the cell and to differentiate them from intracellular vesicles. Based on this original definition, we use "exosomes" as synonymous with "extracellular vesicles." While our original studies used ultracentrifugation to isolate these vesicles, we immediately became aware of the significant impact of the isolation method on the number, type, content and integrity of the vesicles isolated. In this review, we discuss and compare the most commonly utilized methods for purifying exosomes for post-isolation analyses. The exosomes derived from these approaches have been assessed for quantity and quality of specific RNA populations and specific marker proteins. These results suggest that, while each method purifies exosomal material, there are pros and cons of each and there are critical issues linked with centrifugation-based methods, including co-isolation of non-exosomal materials, damage to the vesicle's membrane structure and non-standardized parameters leading to qualitative and quantitative variability. The down-stream analyses of these resulting varying exosomes can yield misleading results and conclusions.


Assuntos
Fracionamento Celular/métodos , Vesículas Extracelulares/química , Neoplasias Ovarianas/química , Transporte Biológico , Fracionamento Celular/instrumentação , Centrifugação com Gradiente de Concentração/instrumentação , Centrifugação com Gradiente de Concentração/métodos , Cromatografia em Gel/instrumentação , Cromatografia em Gel/métodos , Vesículas Extracelulares/metabolismo , Feminino , Filtração/instrumentação , Filtração/métodos , Floculação , Humanos , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/patologia , Tamanho da Partícula , Ultracentrifugação/instrumentação , Ultracentrifugação/métodos
5.
Front Genet ; 4: 142, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23908664

RESUMO

We have previously demonstrated that tumor cells release membranous structures into their extracellular environment, which are termed exosomes, microvesicles or extracellular vesicles depending on specific characteristics, including size, composition and biogenesis pathway. These cell-derived vesicles can exhibit an array of proteins, lipids and nucleic acids derived from the originating tumor. This review focuses of the transcriptome (RNA) of these extracellular vesicles. Based on current data, these vesicular components play essential roles as conveyers of intercellular communication and mediators of many of the pathological conditions associated with cancer development, progression and therapeutic failures. These extracellular vesicles express components responsible for angiogenesis promotion, stromal remodeling, signal pathway activation through growth factor/receptor transfer, chemoresistance, and genetic exchange. These tumor-derived extracellular vesicles not only to represent a central mediator of the tumor microenvironment, but their presence in the peripheral circulation may serve as a surrogate for tumor biopsies, enabling real-time diagnosis and disease monitoring.

6.
PLoS One ; 7(10): e46874, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23056502

RESUMO

BACKGROUND: Exosomes are small membranous vesicles secreted into body fluids by multiple cell types, including tumor cells, and in various disease conditions. Tumor exosomes contain intact and functional mRNAs, small RNAs (including miRNAs), and proteins that can alter the cellular environment to favor tumor growth. Molecular profiling may increase our understanding of the role of exosomes in melanoma progression and may lead to discovery of useful biomarkers. METHODOLOGY/PRINCIPAL FINDINGS: In the present study, we used mRNA array profiling to identify thousands of exosomal mRNAs associated with melanoma progression and metastasis. Similarly, miRNA array profiling identified specific miRNAs, such as hsa-miR-31, -185, and -34b, involved in melanoma invasion. We also used proteomic analysis and discovered differentially expressed melanoma exosomal proteins, including HAPLN1, GRP78, syntenin-1, annexin A1, and annexin A2. Importantly, normal melanocytes acquired invasion ability through molecules transported in melanoma cell-derived exosomes. CONCLUSIONS/SIGNIFICANCE: Our results indicate that melanoma-derived exosomes have unique gene expression signatures, miRNA and proteomics profiles compared to exosomes from normal melanocytes. To the best of our knowledge, this is the first in-depth screening of the whole transcriptome/miRNome/proteome expression in melanoma exosomes. These results provide a starting point for future more in-depth studies of tumor-derived melanoma exosomes, which will aid our understanding of melanoma biogenesis and new drug-targets that may be translated into clinical applications, or as non-invasive biomarkers for melanoma.


Assuntos
Exossomos/metabolismo , Perfilação da Expressão Gênica , Melanoma/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Proteômica , Linhagem Celular Tumoral , Progressão da Doença , Chaperona BiP do Retículo Endoplasmático , Espaço Extracelular/metabolismo , Humanos , Melanócitos/citologia , Melanócitos/patologia , Melanoma/genética , Melanoma/metabolismo , Invasividade Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/genética
7.
Anal Biochem ; 428(1): 44-53, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22691960

RESUMO

Cell-derived vesicles are recognized as essential components of intercellular communication, and many disease processes are associated with their aberrant composition and release. Circulating tumor-derived vesicles have major potential as biomarkers; however, the diagnostic use of exosomes is limited by the technology available for their objective characterization and measurement. In this study, we compare nanoparticle tracking analysis (NTA) with submicron particle analysis (SPA), dynamic light scattering (DLS), and electron microscopy (EM) to objectively define size distribution, number, and phenotype of circulating cell-derived vesicles from ovarian cancer patients. Using the NanoSight LM10 instrument, cell-derived vesicles were visualized by laser light scattering and analyzing Brownian motion of these vesicles captured by video. The NTA software calculates the size and total concentration of the vesicles in solution. Using vesicles isolated from ovarian cancer patients, we demonstrate that NTA can measure the size distributions of cell-derived vesicles comparable to other analysis instrumentation. Size determinations by NTA, SPA, and DLS were more objective and complete than that obtained with the commonly used EM approach. NTA can also define the total vesicle concentration. Furthermore, the use of fluorescent-labeled antibodies against specific markers with NTA allows the determination of the "phenotype" of the cell-derived vesicles.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Nanopartículas/química , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Western Blotting , Rastreamento de Células , Micropartículas Derivadas de Células/química , Micropartículas Derivadas de Células/efeitos dos fármacos , Micropartículas Derivadas de Células/ultraestrutura , Fracionamento Químico , Cromatografia em Gel , Detergentes/farmacologia , Eletroforese em Gel de Poliacrilamida , Feminino , Fluorescência , Humanos , Immunoblotting , Nanopartículas/ultraestrutura , Tamanho da Partícula , Espalhamento de Radiação
8.
Semin Immunopathol ; 33(5): 441-54, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21688197

RESUMO

Cancer cells, both in vivo and in vitro, have been demonstrated to release membranous structures, defined as microvesicles or exosomes, consisting of an array of macromolecules derived from the originating cells, including proteins, lipids, and nucleic acids. While only recently have the roles of these vesicular components in intercellular communication become elucidated, significant evidence has demonstrated that tumor exosomes can exert a broad array of detrimental effects on the immune system-ranging from apoptosis of activated cytotoxic T cells to impairment of monocyte differentiation into dendritic cells, to induction of myeloid-suppressive cells and T regulatory cells. Immunosuppressive exosomes of tumor origin can be found within neoplastic lesions and in biologic fluids from cancer patients, implying a potential role of these pathways in in vivo tumor progression and systemic paraneoplastic syndromes. Through the expression of molecules involved in angiogenesis promotion, stromal remodeling, signaling pathway activation through growth factor/receptor transfer, chemoresistance, and genetic intercellular exchange, tumor exosomes could represent a central mediator of the tumor microenvironment. By understanding the nature of these tumor-derived exosomes/microvesicles and their roles in mediating cancer progression and modulating the host immune response will significantly impact therapeutic approaches targeting exosomes.


Assuntos
Exossomos/imunologia , Exossomos/metabolismo , Tolerância Imunológica , Neoplasias/imunologia , Neoplasias/metabolismo , Microambiente Tumoral/imunologia , Animais , Comunicação Celular , Exossomos/química , Humanos , Fatores Imunológicos/imunologia , Fatores Imunológicos/metabolismo
9.
Methods Mol Biol ; 728: 235-46, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21468952

RESUMO

While the existence of exosomes has been known for over three decades, they have garnered recent interest due to their potential diagnostic and therapeutic relevance. The expression and release of specific tumor-derived proteins into the peripheral circulation has served as the centerpiece of cancer screening and diagnosis. Recently, tissue-associated microRNA (miRNA) has been shown to be characteristic of tumor type and developmental origin, as well as exhibit diagnostic potential. Tumors actively release exosomes, exhibiting proteins and RNAs derived from the originating cell, into the peripheral circulation and other biologic fluids. Recently, we have demonstrated the presence of miRNAs within the RNA fraction of circulating tumor-derived exosomes. Currently, in over 75 investigations compiled in ExoCarta, over 2,300 proteins and 270 miRNAs have been linked with exosomes derived from biologic fluids. Our previous work has indicated that these circulating exosomal proteins and miRNAs can serve as surrogates for the tumor cell-associated counterparts, extending their diagnostic potential to asymptomatic individuals. In this chapter, we compare currently utilized methods for purifying exosomes for postisolation analyses. The exosomes derived from these approaches were assessed for quantity and quality of specific RNA populations and specific marker proteins. These results suggest that, while each method purifies exosomal material, circulating exosomes isolated by ExoQuick precipitation produces exosomal RNA and protein with greater purity and quantity than chromatography, ultracentrifugation, and DynaBeads. While this precipitation approach isolates exosomes in general and does not exhibit specificity for the originating cell, the increased quantity and quality of exosomal proteins and RNA should enhance the sensitivity and accuracy of down-stream analyses, such as qRT-PCR profiling of miRNA and mass spectrometric and electrophoretic analyses of exosomal proteins.


Assuntos
Exossomos/metabolismo , Proteômica/métodos , RNA/análise , Western Blotting , Precipitação Química , Cromatografia em Gel , Humanos , Magnetismo , Microesferas , Ultracentrifugação
10.
Am J Reprod Immunol ; 66(4): 259-69, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21410811

RESUMO

PROBLEM Our previous studies demonstrated that trophoblast-derived exosomes induced synthesis and release of pro-inflammatory cytokines, including interleukin-1ß (IL-1ß) by macrophages. The objective of this study was to characterize the mechanism and receptors associated with this induction. METHOD OF STUDY Exosomes were isolated from Sw71 trophoblast-conditioned media by ultrafiltration and ultracentrifugation. Using macrophages isolated from normal donors, cytochalasin D was used to block exosome uptake. Induction of IL-1ß mRNA was investigated by qRT-PCR, pro-IL-1ß protein by western immunoblotting, and mature IL-1ß release by ELISA. RGD peptides were used to block fibronectin binding by macrophage α5ß1 integrin. RESULTS Uptake of exosomes by macrophages was completely blocked by pre-treatment with cytochalasin D. Although induction of some cytokines (such as C4A and CCL11) requires uptake, induction of IL-1ß occurred without exosome internalization. Cytochalasin D treatment did not inhibit exosome-mediated induction of IL-1ß mRNA, production of the pro-protein, or release of mature IL-1ß. Blocking of fibronectin binding using RGD peptides demonstrated the abrogation of exosome-mediated IL-1ß production. CONCLUSION Although trophoblast-derived exosomes have been demonstrated to induce IL-1ß, this is the first demonstration of IL-1ß induction by exosome-associated fibronectin. Based on this pro-inflammatory role of exosome-associated fibronectin, it may represent an important general immunoregulatory mechanism.


Assuntos
Endocitose/efeitos dos fármacos , Exossomos/metabolismo , Fibronectinas/metabolismo , Interleucina-1beta/biossíntese , Macrófagos/metabolismo , Trofoblastos/metabolismo , Linhagem Celular , Quimiocina CCL11/biossíntese , Meios de Cultivo Condicionados/química , Citocalasina D/farmacologia , Endocitose/imunologia , Ensaio de Imunoadsorção Enzimática , Exossomos/imunologia , Feminino , Fibronectinas/imunologia , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-1beta/análise , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Oligopeptídeos/farmacologia , Gravidez , Ligação Proteica/efeitos dos fármacos , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase em Tempo Real , Receptores Imunológicos/antagonistas & inibidores , Receptores de Peptídeos/antagonistas & inibidores , Trofoblastos/citologia , Trofoblastos/imunologia , Ultracentrifugação
11.
Am J Reprod Immunol ; 65(1): 65-77, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20560914

RESUMO

INTRODUCTION: trophoblast cells have been demonstrated to regulate monocyte migration and differentiation, leading to pro-inflammatory profiles. Because trophoblast cells release exosomes with immunoregulatory properties, trophoblast-derived exosomes are proposed to 'educate' monocytes, creating a pro-inflammatory environment. METHOD OF STUDY: exosomes were isolated from conditioned media of Swan71 cells by ultrafiltration and ultracentrifugation. Exosome-induced migration was assessed using a two-chamber system. Cytokine profiles were defined using cytokine arrays, and mRNA levels of affected cytokines were examined by qRT-PCR and ELISA. RESULTS: within 20 min, 8-10% of monocytes took up labeled exosomes isolated from Swan71 cells. Trophoblast-derived exosomes increased monocyte migration in a dose-dependent manner and produced significant increases in production of interleukin (IL)-1ß, IL-6, Serpin-E1, granulocyte colony-stimulating factor, granulocyte/monocyte colony-stimulating factor, and tumor necrosis factor-α. CONCLUSION: this study presents the initial demonstration that trophoblast-derived exosomes are capable of recruiting and 'educating' monocytes to produce pro-inflammatory cytokine/chemokine profiles in a cell-contact-independent manner.


Assuntos
Exossomos/fisiologia , Monócitos/fisiologia , Trofoblastos/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular , Movimento Celular/fisiologia , Quimiocinas/biossíntese , Feminino , Regulação da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos/biossíntese , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Humanos , Interleucina-1/biossíntese , Interleucina-6/biossíntese , Macrófagos/citologia , Monócitos/citologia , Inibidor 1 de Ativador de Plasminogênio/biossíntese , Gravidez , RNA Mensageiro/genética , Trofoblastos/citologia , Fator de Necrose Tumoral alfa/biossíntese
12.
Am J Pathol ; 177(4): 1606-10, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20802178

RESUMO

Exosomes released from tumor cells having been shown to induce interleukin-6 release from myeloid-derived suppressor cells in a Toll-like receptor 2/Stat3-dependent manner. In this study, we show that exosomes released from tumor cells re-isolated from syngeneic mice are capable of inducing interleukin-6 in a Toll-like receptor 2-independent manner, whereas the data generated from exosomes of tumor cells having undergone numerous in vitro passages induce interleukin-6 in a Toll-like receptor 2-dependent manner. This discrepancy may be due to the source of tumor cells used to generate the exosomes for this study. These results suggest that exosomes released from tumor cells that are not within a tumor microenvironment may not realistically represent the role of tumor exosomes in vivo. This is an important consideration since frequently passing tumor cells in vivo is an accepted practice for studying tumor exosome-mediated inflammatory responses.


Assuntos
Exossomos/metabolismo , Interleucina-6/metabolismo , Linfoma/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Melanoma Experimental/metabolismo , Células Mieloides/metabolismo , Receptor 2 Toll-Like/metabolismo , Animais , Western Blotting , Células Cultivadas , Técnicas de Cocultura , Ensaio de Imunoadsorção Enzimática , Linfoma/patologia , Neoplasias Mamárias Experimentais/patologia , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/citologia , Fator 88 de Diferenciação Mieloide/metabolismo , Baço/citologia , Baço/metabolismo , Linfócitos T/metabolismo , Microambiente Tumoral
13.
Cancer Biomark ; 8(4-5): 281-91, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-22045359

RESUMO

Tumor cells release membranous structures, defined as microvesicles or exosomes, consisting of an array of macromolecules derived from the originating cells, including proteins, lipids, and RNA. The expression of antigenic molecules recognizable by T cells originally suggested a role for these vesicles as a cell-free antigen source for anti-cancer vaccines; however, evidence demonstrates that tumor exosomes can exert a broad array of detrimental effects on the immune system - ranging from apoptosis of activated cytotoxic T cells to impairment of monocyte differentiation into dendritic cells, to induction of myeloid-suppressive cells. Immunosuppressive exosomes of tumor origin can be found in neoplastic lesions and biologic fluids from cancer patients, implying a potential role of these pathways in in vivo tumor progression and systemic paraneoplastic syndromes. Through the expression of molecules involved in angiogenesis promotion, stromal remodeling, signaling pathway activation through growth factor/receptor transfer, chemoresistance, and intercellular genetic exchange, tumor exosomes could represent a central mediator of the tumor microenvironment. Their release by tumor cells may represent the future for targeting therapeutic interventions and for development of multiplexed diagnostic biomarkers.


Assuntos
Biomarcadores Tumorais/metabolismo , Exossomos/metabolismo , Neoplasias/diagnóstico , Neoplasias/metabolismo , Animais , Comunicação Celular , Exossomos/imunologia , Humanos , Evasão Tumoral
14.
Int J Cancer ; 127(5): 1124-30, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20027629

RESUMO

Immunotherapy of colorectal carcinoma (CRC) has great promise as the presence of T lymphocytes in CRC tissues in situ is correlated with reduced recurrence and increased survival. Thus, identification of the antigens recognized by T cells of CRC patients may permit development of vaccines with potential benefit for these patients. Using expression cloning, we identified the antigen, nucleophosmin (Npm), recognized by an HLA-A1 restricted cytotoxic T lymphocyte (CTL) line derived from the peripheral blood mononuclear cells (PBMC) of a rectal cancer patient. A decamer peptide derived from the Npm sequence sensitized peptide-pulsed HLA-A1 positive cells to lysis by the CTL line. The peptide also induced proliferative and cytotoxic T lymphocytes in the PBMC of 4 of 6 CRC patients, which lysed HLA-A1 positive peptide-pulsed target cells and CRC cells endogenously expressing Npm. Overexpression of Npm by tumors of various histological types, recognition of the antigen by T cells derived from different CRC patients and association of the antigen with poor prognostic outcome make it a promising target for immunotherapeutic intervention in cancer patients.


Assuntos
Antígeno HLA-A1/imunologia , Proteínas Nucleares/imunologia , Fragmentos de Peptídeos/farmacologia , Neoplasias Retais/imunologia , Linfócitos T Citotóxicos/imunologia , Apresentação de Antígeno , Antígenos de Neoplasias/imunologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Células Cultivadas , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Feminino , Humanos , Imunoterapia , Ativação Linfocitária , Melanoma/imunologia , Melanoma/patologia , Nucleofosmina , Fragmentos de Peptídeos/imunologia , Neoplasias Retais/patologia
15.
Gynecol Oncol ; 116(2): 213-21, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19945743

RESUMO

OBJECTIVES: Current antigen-based diagnostic assays for ovarian cancers rely on intravasation of specific aberrantly expressed proteins and their achieving detectable steady-state concentrations, resulting in their inability to truly detect small early lesions. In contrast, tumor antigen immunorecognition is observed following initial transformation events. Our objective was to characterize humoral antitumor responses in terms of IgG subclasses generated and tumor antigens recognized. METHODS: For patients with benign and malignant ovarian disease, tumor-reactive IgG subclasses were characterized by Western immunoblotting. Antigen recognition patterns were analyzed by 2-dimensional electrophoresis and proteins exhibiting shared or stage-specific recognition were defined by mass spectrometry (MS) sequencing. RESULTS: Sera from ovarian cancer patients exhibited significantly greater immunoreactivities than either controls or women with benign disease. While late-stage patients recognized more proteins at greater intensity, stage-specific differential recognition patterns were observed in the IgG subclasses, with the greatest recognition appearing in IgG2 subclasses. Immunoreactivity in IgG2 and IgG3 from stage I and II patients appears to be most intense with nuclear antigens >40 kDa, while, in stage III patients, additional immunoreactivity was present in the <40 kDa components. Stage III patients also exhibited similar reaction with membrane antigens <40 kDa. Two-dimensional electrophoresis revealed 32 stage-linked antigenic differences with 11 in early-stage and 21 in late-stage ovarian cancer. CONCLUSIONS: Owing to the timing and stability of humoral responses, quantitation of IgG subclasses recognizing specific tumor antigens provides superior biomarkers for early cancer identification and allows for differentiation of benign versus malignant ovarian masses and early- and late-stage cancers.


Assuntos
Anticorpos Antineoplásicos/classificação , Antígenos de Neoplasias/classificação , Neoplasias Ovarianas/imunologia , Anticorpos Antineoplásicos/imunologia , Antígenos de Neoplasias/imunologia , Western Blotting , Carcinoma Papilar/imunologia , Estudos de Casos e Controles , Cistadenocarcinoma Seroso/imunologia , Eletroforese em Gel Bidimensional , Epitopos , Feminino , Humanos , Imunidade Humoral , Imunoglobulina G/classificação , Imunoglobulina G/imunologia , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Ovarianas/patologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
16.
Gynecol Oncol ; 115(1): 112-120, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19647308

RESUMO

OBJECTIVE: Most ovarian cancers are diagnosed at advanced stage (67%) and prospects for significant improvement in survival reside in early diagnosis. Our objective was to validate our array assay for the identification of ovarian cancer based on quantitation of tumor-reactive IgG. METHODS: The diagnostic array utilizes specific exosome-derived antigens to detect reactive IgG in patients' sera. Specific protein targets were isolated by immunoaffinity from exosomes derived from ovarian tumor cell lines. Sera were obtained from age-matched female volunteers, women with benign ovarian disease and with ovarian cancer. Immunoreactivity was also compared between exosomal proteins and their recombinant counterparts. RESULTS: Sera from ovarian cancer patients exhibited significantly greater immunoreactivities than either normal controls or women with benign disease (both considered negative to all antigens tested). Reactivities with nucleophosmin, cathepsin D, p53, and SSX common antigen for patients with all stages of ovarian cancer were significantly higher than for controls and women with benign ovarian disease. Reactivity with placental type alkaline phosphatase, TAG 72, survivin, NY-ESO-1, GRP78, and Muc16 (CA125) allowed the differentiation between Stage III/IV and early stage ovarian cancer. CONCLUSIONS: The quantitation of circulating tumor-reactive IgG can be used to identify the presence of ovarian cancer. The analyses of IgG recognition of specific exosomal antigens allows for the differentiation of women with benign ovarian masses from ovarian cancer, as well as distinguishing early and late stage ovarian cancers. Thus, the quantitative assessment of IgG reactive with specific tumor-derived exosomal proteins can be used as diagnostic markers for ovarian cancer.


Assuntos
Autoanticorpos/imunologia , Biomarcadores Tumorais/imunologia , Imunoglobulina G/imunologia , Neoplasias Ovarianas/imunologia , Adulto , Antígenos de Neoplasias/imunologia , Autoanticorpos/sangue , Biomarcadores Tumorais/sangue , Estudos de Casos e Controles , Chaperona BiP do Retículo Endoplasmático , Epitopos , Exossomos/imunologia , Feminino , Humanos , Imunoglobulina G/sangue , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/patologia
17.
Cancer Genomics Proteomics ; 6(3): 189-94, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19487548

RESUMO

BACKGROUND: Despite optimal primary treatment of ovarian cancer, overall prognosis is poor due to recurrences. While steroid hormone receptors are frequently expressed, the role of estrogen receptor (ER) in ovarian carcinogenesis, response to treatment or prognosis has not been established. We analyzed the gene-expression in response to estradiol (E2) and genistein (Gen) in ovarian cancer cells. MATERIALS AND METHODS: Cell lines (Br-1, UL-1; Oy-1), treated with E2 (10 nM) or Gen (5 microM), were used for gene expression profiling. RT-PCR and Western immunoblotting were used to further analyze gene expression data. RESULTS: Twenty-four genes were differentially regulated in ovarian cancer cell lines. C3, CLU, COL6A1, DLC1, NME1, NRIP1, PTEN, RAC2, S100A2 were down-regulated with E2 in Br-1 and UL-1 cells. MK167, SERPINB5, SLC7A5, CDK1NA, LCN2, PLAU, PHB2, CTSB, EGLN2, ERBB2, HMGB1, ID2, ITGB4, TOP2A were up-regulated in Oy-1 cells with E2 and/or genistein. ERBB2 and ID2 (E2 and Gen), LCN2, PHB2 and HMGB1 (Gen) were down-regulated in Br-1 cells. ERalpha and ERbeta were detected in all cell lines at different levels. CONCLUSION: Variable response of ovarian cancer cells to E2 and Gen was observed. Study of ERs including splice variants, co-regulatory molecules are necessary to understand the relevance of receptors.


Assuntos
Estradiol/farmacologia , Expressão Gênica/efeitos dos fármacos , Genisteína/farmacologia , Neoplasias Ovarianas/genética , Fitoestrógenos/farmacologia , Linhagem Celular Tumoral , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Ovarianas/metabolismo , Proibitinas , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
18.
Clin Lung Cancer ; 10(1): 42-6, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19289371

RESUMO

PURPOSE: To date, there is no screening test for lung cancer shown to affect overall mortality. MicroRNAs (miRNAs) are a class of small noncoding RNA genes found to be abnormally expressed in several types of cancer, suggesting a role in the pathogenesis of human cancer. PATIENTS AND METHODS: We evaluated the circulating levels of tumor exosomes, exosomal small RNA, and specific exosomal miRNAs in patients with and without lung adenocarcinoma, correlating the levels with the American Joint Committee on Cancer (AJCC) disease stage to validate it as an acceptable marker for diagnosis and prognosis in patients with adenocarcinoma of the lung. RESULTS: To date, 27 patients with lung adenocarcinoma AJCC stages I-IV and 9 controls, all aged 21-80 years, were enrolled in the study. Small RNA was detected in the circulating exosomes. The mean exosome concentration was 2.85 mg/mL (95% CI, 1.94-3.76) for the lung adenocarcinoma group versus 0.77 mg/mL (95% CI, 0.68-0.86) for the control group (P < .001). The mean miRNA concentration was 158.6 ng/mL (95% CI, 145.7-171.5) for the lung adenocarcinoma group versus 68.1 ng/mL (95% CI, 57.2-78.9) for the control group (P < .001). Comparisons between peripheral circulation miRNA-derived exosomes and miRNA-derived tumors indicated that the miRNA signatures were not significantly different. CONCLUSION: The significant difference in total exosome and miRNA levels between lung cancer patients and controls, and the similarity between the circulating exosomal miRNA and the tumor-derived miRNA patterns, suggest that circulating exosomal miRNA might be useful as a screening test for lung adenocarcinoma. No correlation between the exosomal miRNA levels and the stage of disease can be made at this point.


Assuntos
Adenocarcinoma/diagnóstico , Biomarcadores Tumorais/análise , Neoplasias Pulmonares/diagnóstico , MicroRNAs/análise , Adenocarcinoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/metabolismo , Exossomos/metabolismo , Feminino , Humanos , Neoplasias Pulmonares/patologia , Masculino , MicroRNAs/metabolismo , Análise em Microsséries , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Prognóstico , Adulto Jovem
19.
Immun Ageing ; 6: 1, 2009 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-19265538

RESUMO

BACKGROUND: Immunosenescence is an age-associated disorder occurring primarily in T cell compartments, including altered subset composition, functions, and activation. In women, evidence implicates diminished estrogen in the postmenopausal period as a contributing factor to diminished T cell responsiveness. Since hypoestrogenism is present in postmenopausal women, our objective focused on whether T cell activation, defined as signalling molecule expressions and activation, and function, identified as IL-2 production, were affected by low estrogen. METHODS: Using Jurkat 6.1 T cells, consequences of 4 pg/ml (corresponding to postmenopausal levels) or 40 pg/ml (premenopausal levels) of estradiol (E(2)) were analyzed on signalling proteins, CD3-zeta, JAK2, and JAK3, determined by Western immunoblotting. These consequences were correlated with corresponding gene expressions, quantified by real time-polymerase chain reaction. Tyrosine phosphorylation of CD3-zeta was defined by immunoprecipitation and western immunoblotting following activation by T cell receptor (TcR) cross-linking. CD3-zeta expression and modulation was also confirmed in T cells from pre- and postmenopausal women. To assess functional consequences, IL-2 production, induced by PMA and ionomycin, was determined using enzyme-linked immunosorbent spot assay (ELISpot). RESULTS: At 40 pg/ml E(2), the level of signalling protein CD3-zeta was elevated 1.57-fold, compared with cells exposed to 4 pg/ml E(2). The CD3-zeta proteins also exhibited altered levels of activation-induced phosphorylation in the presence of 40 pg/ml E(2) versus 4 pg/ml: 23 kD phosphorylated form increased 2.64-fold and the 21 kD form was elevated 2.95-fold. Examination of kinases associated with activation signalling also demonstrated that, in the presence of 40 pg/ml E(2), JAK2 protein expression was increased 1.64-fold (p < 0.001) and JAK3 enhanced 1.79-fold (p < 0.001) compared to 4 pg/ml. mRNA levels for CD3-zeta, JAK2, and JAK3 were significantly increased following exposure to 40 pg/ml E(2) (2.39, 2.01, and 2.21 fold, respectively) versus 4 pg/ml. These findings were confirmed in vivo, since T cells from postmenopausal women exhibited 7.2-fold diminished CD3-zeta expression, compared to pre-menopausal controls and this expression was elevated 3.8-fold by addition of 40 pg/ml E(2). Functionally, Jurkat cells exposed to 40 pg/ml E(2) and activated exhibited significantly elevated numbers of IL-2 producing colonies compared to 4 pg/ml (75.3 +/- 2.2 versus 55.7 +/- 2.1 colonies, p < 0.0001). CONCLUSION: Jurkat T cells exposed to 4 pg/ml E(2) expressed significantly diminished activation signalling proteins, correlating with reduced IL-2 production. Lower signalling protein levels appear to result from decreased CD3-zeta, JAK2, and JAK3 gene expressions. These findings may provide a molecular basis for immunosenescence associated with the postmenopausal state.

20.
Am J Reprod Immunol ; 61(1): 26-33, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19086989

RESUMO

PROBLEM: This study addressed the ability of genistein to reverse the loss of T-cell signaling components, induced by estrogen deficiency associated with aging. METHOD OF STUDY: Using Jurkat 6.1 T cells, genistein regulation of CD3zeta, JAK3, and NFkappaB was analysed by Western immunoblotting at 4 or 40 pg/mL (post- and pre-menopausal levels, respectively) estradiol (E(2)). Corresponding gene expressions were quantified by real-time polymerase chain reaction (RT-PCR). For functionality, levels of IL-2 were correlated with its transcription by RT-PCR. RESULTS: At 4 pg/mL E(2), signaling proteins were decreased compared with 40 pg/mL: CD3zeta, 1.58-fold; JAK3, 1.75-fold; and NFkappaB, 1.73-fold (P < 0.001). Genistein, at 0.5 and 5.0 microm, added to 4 pg/mL E(2) induced their expression to 40 pg/mL levels. While significantly diminished IL-2 mRNA levels were observed at 4 pg/mL E(2), genistein induced IL-2 mRNA to 40 pg/mL levels. CONCLUSION: Genistein restores CD3zeta, NFkappaB and JAK3 proteins and mRNAs at post-meonopausal estrogen levels, in vitro, with no significant effect at pre-menopausal estrogen levels.


Assuntos
Estrogênios/sangue , Genisteína/farmacologia , Pós-Menopausa/efeitos dos fármacos , Pós-Menopausa/imunologia , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Complexo CD3/imunologia , Feminino , Humanos , Interleucina-2/biossíntese , Janus Quinase 3/metabolismo , Células Jurkat , Ativação Linfocitária/efeitos dos fármacos , NF-kappa B/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA