Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Int J Mol Sci ; 20(22)2019 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-31752449

RESUMO

Background-There are currently no effective therapies for diffuse malignant peritoneal mesothelioma (DMPM) patients with disease recurrence. In this study, we investigated the biology of DMPM by analyzing the EGFR family, Axl, and MET, in order to assess the presence of cross-talk between these receptors, suggesting the effectiveness of combined targeted treatments in DMPM. Method-We analyzed a series of 22 naïve epithelioid DMPM samples from a single institute, two of which showed higher-grade malignancy ("progressed"). EGFR, HER2, HER3, Axl, and MET activation and expression were investigated by biochemical analysis, real-time PCR immunofluorescence, immunohistochemistry, next-generation sequencing, miRNA, and mRNA in situ hybridization. Results-In most DMPMs, a strong EGFR activation was associated with HER2, HER3, Axl, and MET co-activation, mediated mainly by receptor heterodimerization and autocrine-paracrine loops induced by the expression of their cognate ligands. Axl expression was downregulated by miRNA34a. Mutations in MET Sema domain were exclusively found in two "progressed" DMPMs, and the combined Axl and MET inhibition reduced cellular motility in a DMPM cell line obtained from a "progressed" DMPM. Conclusion-The results indicate that the coordinated activity of multiple cross-talks between RTKs is directly involved in the biology of DMPM, suggesting the combined inhibition of PIK3 and mTOR as an effective strategy that may be easily implemented in clinical practice, and indicating that the combined inhibition of EGFR/HER2 and HER3 and of Axl and MET deserves further investigation.


Assuntos
Neoplasias Pulmonares/genética , Mesotelioma/genética , Neoplasias Peritoneais/genética , Adulto , Idoso , Linhagem Celular Tumoral , Terapia Combinada/métodos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Masculino , Mesotelioma/tratamento farmacológico , Mesotelioma Maligno , MicroRNAs/genética , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/genética , Neoplasias Peritoneais/tratamento farmacológico , Peritônio/efeitos dos fármacos , Inibidores de Proteínas Quinases/uso terapêutico , RNA Mensageiro/genética
3.
Eur Urol Focus ; 5(4): 689-692, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28855072

RESUMO

Pan-fibroblast growth-factor receptor (FGFR) inhibitors hold promise in FGFR-altered patients, but such alterations are rare in advanced urothelial carcinoma. In order to assess whether we may increase the number of eligible patients by using different molecular techniques for detecting alterations, we pooled the results of the centralised FGFR mutation/translocation assays that were performed in Clinical Laboratory Improvement Amendments-certified laboratories within multiple phase 2 trials. At our centre, the same tissue blocks were used to analyse FGFR1-3 messenger RNA expression through messenger RNA in situ hybridisation (ISH; RNAscope 2.5 assay). From October 2016 to March 2017, 52 cases were analysed. Seventeen patients (32.7%) had an upper tract primary tumour. Ten patients (19.2%) had FGFR DNA alterations. Twenty-nine (55.8%) had positive ISH analysis: N=17 score 3, N=12 score 4. Of note, concordance between the two tests was obtained in seven out of 10 patients. Sixty percent of mutated patients had an upper tract primary tumour versus 31% of ISH-positive patients. PATIENT SUMMARY: We found three-fold higher frequency of fibroblast growth-factor receptor alterations at the RNA versus DNA level in advanced urothelial carcinoma, with a different distribution according to the method used and the site of the primary tumour. The evaluation of the therapeutic response to pan-fibroblast growth-factor receptor inhibitors according to the method of assessment is warranted.


Assuntos
Carcinoma de Células de Transição/genética , DNA de Neoplasias/genética , RNA Mensageiro/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética , Neoplasias Urológicas/genética , Idoso , Carcinoma de Células de Transição/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Estudos Retrospectivos , Neoplasias Urológicas/patologia
4.
Expert Rev Mol Diagn ; 18(3): 259-277, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29431533

RESUMO

INTRODUCTION: Bright-field in situ hybridization (ISH) methods detect gene alterations that may improve diagnostic precision and personalized management of cancer patients. Areas covered: This review focuses on some bright-field ISH techniques for detection of gene amplification or viral infection that have already been introduced in tumor pathology, research and diagnostic practice. Other emerging ISH methods, for the detection of translocation, mRNA and microRNA have recently been developed and need both an optimization and analytical validation. The review also deals with their clinical applications and implications on the management of cancer patients. Expert commentary: The technology of bright-field ISH applications has advanced significantly in the last decade. For example, an automated dual-color assay was developed as a clinical test for selecting cancer patients that are candidates for personalized therapy. Recently an emerging bright-field gene-protein assay has been developed. This method simultaneously detects the protein, gene and centromeric targets in the context of tissue morphology, and might be useful in assessing the HER2 status particularly in equivocal cases or samples with heterogeneous tumors. The application of bright-field ISH methods has become the gold standard for the detection of tumor-associated viral infection as diagnostic or prognostic factors.


Assuntos
Biomarcadores Tumorais/genética , Hibridização In Situ/métodos , Técnicas de Diagnóstico Molecular/métodos , Neoplasias/genética , Medicina de Precisão/métodos , Biomarcadores Tumorais/metabolismo , DNA Viral/genética , Rearranjo Gênico , Humanos , Neoplasias/patologia , Neoplasias/virologia
5.
Hum Pathol ; 74: 32-42, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28993274

RESUMO

The aim of this study is to compare 2 in situ hybridization (ISH) detection methods for human papilloma virus (HPV) 16 E6/E7 mRNA, that is, the RNAscope 2.0 High Definition (HD) and the upgraded RNAscope 2.5 HD version. The RNAscope 2.5 HD has recently replaced the RNAscope 2.0 HD detection kit. Therefore, this investigation starts from the need to analytically validate the new mRNA ISH assay and, possibly, to refine the current algorithm for HPV detection in oropharyngeal squamous cell carcinoma with the final goal of applying it to daily laboratory practice. The study was based on HPV status and on generated data, interpreted by a scoring algorithm. The results highlighted that the compared RNAscope HPV tests had a good level of interchangeability and enabled to identify oropharyngeal squamous cell carcinoma that are truly driven by high-risk HPV infection. This was also supported by the comparison of the RNAscope HPV test with HPV E6/E7 mRNA real-time reverse-transcription polymerase chain reaction in a fraction of cases where material for HPV E6/E7 mRNA real-time reverse-transcription polymerase chain reaction was available. Furthermore, the algorithm that associates p16 immunohistochemistry with the identification of HPV mRNA by RNAscope was more effective than the one that associated p16 immunohistochemistry with the identification of HPV DNA by ISH.


Assuntos
Carcinoma de Células Escamosas/virologia , Papillomavirus Humano 16/isolamento & purificação , Proteínas Oncogênicas Virais/metabolismo , Neoplasias Orofaríngeas/virologia , Infecções por Papillomavirus/diagnóstico , Proteínas Repressoras/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Feminino , Humanos , Imuno-Histoquímica , Hibridização In Situ , Masculino , Neoplasias Orofaríngeas/metabolismo , Neoplasias Orofaríngeas/patologia , Infecções por Papillomavirus/metabolismo , Infecções por Papillomavirus/patologia , RNA Viral
6.
Sci Rep ; 7(1): 15992, 2017 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-29167573

RESUMO

Metformin is a widely used and well-tolerated anti-diabetic drug that can reduce cancer risk and improve the prognosis of certain malignancies. However, the mechanism underlying its anti-cancer effect is still unclear. We studied the anti-cancer activity of metformin on colorectal cancer (CRC) by using the drug to treat HT29, HCT116 and HCT116 p53-/- CRC cells. Metformin reduced cell proliferation and migration by inducing cell cycle arrest in the G0/G1 phase. This was accompanied by a sharp decrease in the expression of c-Myc and down-regulation of IGF1R. The anti-proliferative action of metformin was mediated by two different mechanisms: AMPK activation and increase in the production of reactive oxygen species, which suppressed the mTOR pathway and its downstream targets S6 and 4EBP1. A reduction in CD44 and LGR5 expression suggested that the drug had an effect on tumour cells with stem characteristics. However, a colony formation assay showed that metformin slowed the cells' ability to form colonies without arresting cell growth, as confirmed by absence of apoptosis, autophagy or senescence. Our finding that metformin only transiently arrests CRC cell growth suggests that efforts should be made to identify compounds that combined with the biguanide can act synergistically to induce cell death.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Neoplasias Colorretais/metabolismo , Metformina/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Antineoplásicos/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Células HCT116 , Humanos , Hibridização In Situ , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
7.
Expert Rev Hematol ; 10(6): 505-514, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28468596

RESUMO

INTRODUCTION: Primary effusion lymphoma (PEL) is a rare B-cell lymphoid neoplasm mainly associated with HIV infection, presenting as pleural, peritoneal, and pericardial effusions. A defining property of PEL is its consistent association with Kaposi sarcoma associated herpesvirus (KSHV) infection, and, in most cases, Epstein Barr virus (EBV) co-infection. On these grounds, a review of the literature related to viral cooperation and lymphomagenesis can help to understand the complex interplay between KSHV and EBV in PEL pathogenesis. Areas covered: In this review, the authors highlight clinical, pathologic, genetic and proteomic features of PEL, in the context of viral cooperation in PEL lymphomagenesis. Expert commentary: Tumour cells are characterized by the overexpression of genes that are involved in inflammation and invasion. Coherently, PEL secretomes are enriched in proteins probably responsible for the particular tropism (cell adhesion and migration) of PEL cells. The development of PEL in HIV+ patients is multifactorial and involves a complex interplay among co-infection with oncogenic viruses (EBV and KSHV), inflammatory factors, and environmental conditions.


Assuntos
Transformação Celular Viral , Infecções por Vírus Epstein-Barr , Herpesvirus Humano 4 , Herpesvirus Humano 8 , Linfoma de Efusão Primária , Sarcoma de Kaposi , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/metabolismo , Infecções por Vírus Epstein-Barr/patologia , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Linfoma de Efusão Primária/metabolismo , Linfoma de Efusão Primária/patologia , Linfoma de Efusão Primária/virologia , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/patologia
8.
J Exp Clin Cancer Res ; 36(1): 16, 2017 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-28114961

RESUMO

BACKGROUND: Strategies aimed at obtaining a complete cytoreduction are needed to improve long-term survival for patients with colorectal cancer peritoneal carcinomatosis (CRC-pc). METHODS: We established organoid models from peritoneal metastases of two naïve CRC patients. A standard paraffin inclusion was conducted to compare their 3D structure and immunohistochemical profile with that of the corresponding surgical samples. RNA expression levels of the CRC stem cell marker LGR5 was measured by in situ hybridization. The secretome of organoids was profiled by mass spectrometry. Energy homeostasis of organoids was interfered with 4-IPP and metformin. Biochemical and metabolic changes after drug treatments were investigated by western blot and mass spectrometry. Mitochondria impairment was evaluated by electron microscopy and mitotraker staining. RESULTS: The two organoids recapitulated their corresponding clinical samples in terms of 3D structure and immmunoistochemical profile and were positive for the cancer stem cells marker LGR5. Proteomic analyses of organoids highlighted their strong dependence on energy producing pathways, which suggest that their targeting could be an effective therapeutic approach. To test this hypothesis, we treated organoids with two drugs that target metabolism acting on AMP-activated protein kinase (AMPK), the main regulator of cellular energy homeostasis, which may act as metabolic tumour suppressor in CRC. Organoids were treated with 4-IPP, an inhibitor of MIF/CD74 signalling axis which activates AMPK function, or metformin that inhibits mitochondrial respiratory chain complex I. As a new finding we observed that treatment with 4-IPP downregulated AMPK signalling activity, reduced AKT phosphorylation and activated a JNK-mediated stress-signalling response, thus generating mitochondrial impairment and cell death. Metformin treatment enhanced AMPK activation, decreasing the activity of the anabolic factors ribosomal protein S6 and p4EBP-1 and inducing mitochondrial depolarization. CONCLUSION: We provide evidence that the modulation of AMPK activity may be a strategy for targeting metabolism of CRC-pc organoids.


Assuntos
Antígenos de Diferenciação de Linfócitos B/metabolismo , Neoplasias do Colo/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Metformina/farmacologia , Neoplasias Peritoneais/secundário , Pirimidinas/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , Metabolismo Energético/efeitos dos fármacos , Humanos , Terapia de Alvo Molecular , Neoplasias Peritoneais/tratamento farmacológico , Neoplasias Peritoneais/genética , Proteômica , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas
9.
Curr Opin HIV AIDS ; 12(1): 39-46, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27755151

RESUMO

PURPOSE OF REVIEW: The present review summarizes the association of the different histotypes of Epstein-Barr virus (EBV)-associated lymphomas with known genetic lesions and/or oncogenic viruses. A more comprehensive understanding of the complex interplay existing between genetic abnormalities of tumor cells and the viral contribution to the development of EBV-associated lymphomas is pivotal for the development of more effective treatments. RECENT FINDINGS: Recent evidence indicates that HIV may contribute to lymphomagenesis by acting directly on B lymphocytes as a critical microenvironmental factor. The pathogenesis of EBV-associated lymphomas in patients with HIV infection is considered the result of the concerted action of different factors, mainly including impaired immune surveillance, genetic alterations, and concomitant viral infection (EBV and HIV). SUMMARY: Immunodeficiency states usually increase susceptibility to cancer as a result of reduced immune surveillance and enhanced chances for virus-driven oncogenesis. Lymphoma remains the most frequent neoplastic cause of death among patients infected with HIV. Several of the HIV-associated lymphomas are related to EBV infection. EBV-associated lymphomas in patients infected with HIV are heterogeneous, not only pathologically but also in terms of pathogenetic pathways and cellular derivation.


Assuntos
Infecções por Vírus Epstein-Barr/complicações , Infecções por HIV/complicações , Linfoma/epidemiologia , Humanos , Linfoma/mortalidade
10.
Int J Biol Markers ; 31(2): 0, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27040135

RESUMO

PURPOSE: HPV-related locally advanced head and neck cancers (LA-HNCs) show a good prognosis. This study aimed to investigate the HPV prevalence in LA-HNCs and compare the prognostic value of E1, E6 and L1 genomic viral fragments and p16, individually and in combination, in order to find the best prognosticator in terms of overall survival (OS) and progression-free survival (PFS). PATIENTS AND METHODS: HPV16 was searched in 255 LA-HNC formalin-fixed paraffin-embedded tumor tissues, 89 oropharyngeal cancers (OPCs), and 166 non-OPCs by DNA-PCR with 3 primer pairs. p16 was analyzed by immunohistochemistry in 235 patients. RESULTS: The prevalence of positive samples decreased constantly from E6 to L1 and E1 in both OPCs and non-OPCs. Each LA-HNC patient highlighted variable positivity for each fragment. OPCs showed a higher prevalence of positive samples compared to non-OPCs.Positive coexistence of all the fragments was more common in OPCs (31.5%) than non-OPCs (4.2%), and E1 detection was always associated with E6 and L1. E1-positive OPCs showed improved OS (p = 0.012) and PFS (p = 0.036), while L1- or E6-positive ones did not. p16-positive patients were more prevalent in the OPC (29.8%) than the non-OPC group (7.3%) (p<0.0001) and its prognostic value was not superior to that of E1. However, the multivariate Cox analysis which included E1, L1, E6 status and p16 expression did not show a significant p value. CONCLUSIONS: Though HPV16 positivity measured by DNA-PCR was higher for L1 and E6, they performed weakly as prognosticators; E1 might become a strong prognostic marker for OS and PFS in OPCs.


Assuntos
Neoplasias de Cabeça e Pescoço/virologia , Proteínas Oncogênicas Virais/análise , Neoplasias Orofaríngeas/virologia , Papillomaviridae/isolamento & purificação , Infecções por Papillomavirus/virologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/análise , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Papillomaviridae/genética , Prognóstico , Estudos Retrospectivos , Adulto Jovem
12.
Endocr Relat Cancer ; 22(5): 759-75, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26206776

RESUMO

Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine that is over-expressed in several human neoplastic cells. When MIF binds its receptor (CD74) and co-receptor (CD44), it initiates signaling cascades that orchestrate cell proliferation and survival, and it can directly modulate the activity of AMPK. These activities indicate that MIF potentially regulates cell survival and metabolism. We found that MIF was primarily co-expressed with CD74 in 16 out of 23 papillary thyroid carcinoma (PTC) and in all the 27 available anaplastic thyroid carcinoma (ATC) biopsy samples. MIF and CD74 were co-expressed in TPC-1 and HTC-C3 cell lines. The selective MIF inhibitor, 4-iodo-6-phenylpyrimidine (4-IPP), blocked MIF/CD74 internalization, activated JNK, and dose-dependently inhibited proliferation inducing apoptosis and mitotic cell death. In two CD74-negative cell lines, NIM-1 and K1, 4-IPP treatment partially reduced proliferation. Coordinated MIF and CD74 expression appeared to confer in tumor cells the plasticity necessary to escape cell cycle regulation, metabolic changes, and stress conditions. MIF/CD74 signaling removal made cells susceptible to apoptosis and mitotic cell death. This finding suggests a possible avenue for targeting DNA endoreduplication, thus preventing the proliferation of therapy-resistant cell subpopulations. This study highlights MIF/CD74 axis as an important player in the biology of aggressive thyroid neoplasms.


Assuntos
Carcinoma/patologia , Indóis/farmacologia , Oxirredutases Intramoleculares/antagonistas & inibidores , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Metaboloma , Mitose/efeitos dos fármacos , Proteoma/análise , Neoplasias da Glândula Tireoide/patologia , Apoptose , Biomarcadores Tumorais/metabolismo , Western Blotting , Carcinoma/tratamento farmacológico , Carcinoma/metabolismo , Carcinoma Papilar , Ciclo Celular , Proliferação de Células , Cromatografia Líquida/métodos , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Receptores Imunológicos/metabolismo , Espectrometria de Massas em Tandem/métodos , Câncer Papilífero da Tireoide , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/metabolismo , Células Tumorais Cultivadas
13.
J Oral Pathol Med ; 44(9): 734-45, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25495427

RESUMO

BACKGROUND: Human papillomavirus (HPV)-positive and HPV-negative oropharyngeal squamous cell carcinomas (OSCCs) are two distinct entities. We defined the molecular profiles of druggable receptor tyrosine kinases (RTKs) in both groups. MATERIALS AND METHODS: E5 expression and RTK alterations were studied in 17 HPV-positive and 59 HPV-negative formalin-fixed OSCCs. RTK activation was explored in further 12 frozen OSCCs. RESULTS: The HPV-positive OSCCs showed E5 expression and 33.3% expressed low level of HER2. The HPV-negative OSCCs showed HER2 expression (31.2%), increased HER2 gene copy number (46.51%, P = 0.045) and HER2 activation through HER2/EGFR heterodimerisation; HER3 (51.06%, P = 0.008) and neuregulin (65.63%; P = 0.03) expression, HER3 activation and HER3/EGFR heterodimerisation; and increased IGF-1R copy number (40.50%, P = 0.021), high IGF-1R cDNA values (P = 0.002), IGF-1R activation and expression of IGF1/2 and amphiregulin. PI3KCA mutations/expression/increased gene copy number and PTEN mutations were found in both groups, whereas PTEN gene loss was only observed in the HPV-positive cases. CONCLUSION: Human papillomavirus-positive and HPV-negative OSCC showed different RTK profiles. In HPV-positive cases, it would be interesting to study the expression of E5, which may modulate EGFR turnover and activate VEGF and PDGFRß. In HPV-negative cases, HER3 may be a promising druggable biomarker that deserves further investigation. PI3KCA and PTEN alterations encourage the promising clinical evaluation of PI3K/mTOR inhibitor activity in OSCC, particularly in HPV-positive/PI3KCA-mutated OSCCs because they may be driven by PI3KCA mutation alone.


Assuntos
Carcinoma de Células Escamosas/virologia , Neoplasias Orofaríngeas/enzimologia , Neoplasias Orofaríngeas/virologia , Papillomaviridae/isolamento & purificação , Infecções por Papillomavirus/enzimologia , Receptores Proteína Tirosina Quinases/metabolismo , Anfirregulina/genética , Anfirregulina/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/patologia , Humanos , Neoplasias Bucais/patologia , Mutação , Neoplasias Orofaríngeas/patologia , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Papillomaviridae/genética , Infecções por Papillomavirus/patologia , Infecções por Papillomavirus/virologia , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Receptores Proteína Tirosina Quinases/análise , Receptores Proteína Tirosina Quinases/genética , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Fator de Crescimento Transformador alfa/genética , Fator de Crescimento Transformador alfa/metabolismo
14.
Am J Pathol ; 184(3): 618-30, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24521760

RESUMO

Primary effusion lymphoma (PEL) is a rare B-cell neoplasm in which tumor cells are consistently infected by Kaposi's sarcoma-associated herpesvirus and usually grow in body cavities without tumor mass formation. To detect new proteins related to pathogenesis, four established cell lines from PEL (CRO-AP2, CRO-AP3, CRO-AP5, and CRO-AP6) were characterized by proteomics analysis of the secretome. The secretomes were analyzed using two complementary mass spectrometry platforms: liquid chromatography-mass spectrometry and matrix-assisted laser desorption/ionization time-of-flight-based approaches. Among 266 proteins identified from the proteomics analysis, 139 were considered as predicted secreted. Twenty proteins were specifically secreted by PEL cell lines after comparison with secretomes of human cell lines representative of diverse solid tumors and leukemias. More important, 27 additional proteins were shared by all CRO-AP PEL cell lines. The presence of these proteins was confirmed by IHC in CRO-AP cell lines and in six other PEL cell lines, four PEL clinical samples, and three extracavitary Kaposi's sarcoma-associated herpesvirus-positive solid lymphomas included for comparative analysis. Functional classification showed that PEL cell secretomes were enriched in proteins specifically involved in inflammation/immune response, growth/cell cycle, and mRNA processing, in addition to structural/matrix proteins and proteins with enzymatic activity.


Assuntos
Biomarcadores Tumorais/isolamento & purificação , Herpesvirus Humano 8/fisiologia , Linfoma de Efusão Primária/metabolismo , Sarcoma de Kaposi/metabolismo , Linfócitos B/metabolismo , Linfócitos B/patologia , Linhagem Celular Tumoral , Biologia Computacional , Humanos , Linfoma de Efusão Primária/patologia , Proteoma/química , Sarcoma de Kaposi/patologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Espectrometria de Massas em Tandem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...