Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Expert Rev Mol Diagn ; 18(3): 259-277, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29431533

RESUMEN

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.


Asunto(s)
Biomarcadores de Tumor/genética , Hibridación in Situ/métodos , Técnicas de Diagnóstico Molecular/métodos , Neoplasias/genética , Medicina de Precisión/métodos , Biomarcadores de Tumor/metabolismo , ADN Viral/genética , Reordenamiento Génico , Humanos , Neoplasias/patología , Neoplasias/virología
2.
Hum Pathol ; 74: 32-42, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28993274

RESUMEN

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.


Asunto(s)
Carcinoma de Células Escamosas/virología , Papillomavirus Humano 16/aislamiento & purificación , Proteínas Oncogénicas Virales/metabolismo , Neoplasias Orofaríngeas/virología , Infecciones por Papillomavirus/diagnóstico , Proteínas Represoras/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Femenino , Humanos , Inmunohistoquímica , Hibridación in Situ , Masculino , Neoplasias Orofaríngeas/metabolismo , Neoplasias Orofaríngeas/patología , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/patología , ARN Viral
3.
Expert Rev Hematol ; 10(6): 505-514, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28468596

RESUMEN

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.


Asunto(s)
Transformación Celular Viral , Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 4 , Herpesvirus Humano 8 , Linfoma de Efusión Primaria , Sarcoma de Kaposi , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/metabolismo , Infecciones por Virus de Epstein-Barr/patología , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Linfoma de Efusión Primaria/metabolismo , Linfoma de Efusión Primaria/patología , Linfoma de Efusión Primaria/virología , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/patología
4.
Oncotarget ; 8(20): 32492-32504, 2017 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-28415643

RESUMEN

To gain new insights into desmoplastic small round cell tumors (DSRCTs) by means of gene expression profiling (GEP). Formalin-fixed, paraffin-embedded surgical specimens obtained from seven pretreated DSRCT patients were interrogated using GEP complemented by immunohistochemistry, a cancer stem cell array, and miRNA in situ hybridisation, including the combined chimera modules miRNA-200/ZEB1 and miRNA-34/SLUG. The chimera modules divided the cases into three classes that respectively recapitulated the traits of mesenchymal epithelial reverse transition (MErT), epithelial mesenchymal transition (EMT), and hybrid/partial EMT. This indicates a close correlation between the reprogramming governed by EMT regulators and DSRCT biology, which was further confirmed by miRNA-21 and is consistent with the broad morphological spectrum of DSRCTs. Starting from the miRNA-200/ZEB1 axis, we also found that DSRCTs carry a signature of immunological ignorance that is not responsive to PD--L1 blockade. Evidence that the up-regulation of miRNA-200 and E-cadherin, and quite a high level of miRNA-21 expression segregate with the MErT supports the idea that, in addition to the hybrid/partial state, MErT is also enriched in stemness: the androgen-positive cases, whose stemness traits were confirmed by stem cell arrays, all fell into these two classes. Our findings also confirmed that tumoral cell PDGFRA expression correlates with desmoplasia, and demonstrated the co-expression of PDGFRA and ISLR/Meflin, another marker of pluripotency. Despite the limited number of cases, these findings provide unexpectedly relevant information concerning the pathogenesis of DSRCTs, and prove the validity of miRNA-based chimera circuit modelling in the clinico-pathological setting.


Asunto(s)
Tumor Desmoplásico de Células Pequeñas Redondas/genética , Tumor Desmoplásico de Células Pequeñas Redondas/patología , Perfilación de la Expresión Génica/métodos , Femenino , Humanos , Inmunohistoquímica
5.
Curr Opin HIV AIDS ; 12(1): 39-46, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27755151

RESUMEN

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.


Asunto(s)
Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por VIH/complicaciones , Linfoma/epidemiología , Humanos , Linfoma/mortalidad
6.
Cancer Res ; 76(18): 5562-72, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27402080

RESUMEN

Organization of cancer cells into endothelial-like cell-lined structures to support neovascularization and to fuel solid tumors is a hallmark of progression and poor outcome. In triple-negative breast cancer (TNBC), PDGFRß has been identified as a key player of this process and is considered a promising target for breast cancer therapy. Thus, we aimed at investigating the role of miRNAs as a therapeutic approach to inhibit PDGFRß-mediated vasculogenic properties of TNBC, focusing on miR-9 and miR-200. In MDA-MB-231 and MDA-MB-157 TNBC cell lines, miR-9 and miR-200 promoted and inhibited, respectively, the formation of vascular-like structures in vitro Induction of endogenous miR-9 expression, upon ligand-dependent stimulation of PDGFRß signaling, promoted significant vascular sprouting of TNBC cells, in part, by direct repression of STARD13. Conversely, ectopic expression of miR-200 inhibited this sprouting by indirectly reducing the protein levels of PDGFRß through the direct suppression of ZEB1. Notably, in vivo miR-9 inhibition or miR-200c restoration, through either the generation of MDA-MB-231-stable clones or peritumoral delivery in MDA-MB-231 xenografted mice, strongly decreased the number of vascular lacunae. Finally, IHC and immunofluorescence analyses in TNBC specimens indicated that PDGFRß expression marked tumor cells engaged in vascular lacunae. In conclusion, our results demonstrate that miR-9 and miR-200 play opposite roles in the regulation of the vasculogenic ability of TNBC, acting as facilitator and suppressor of PDGFRß, respectively. Moreover, our data support the possibility to therapeutically exploit miR-9 and miR-200 to inhibit the process of vascular lacunae formation in TNBC. Cancer Res; 76(18); 5562-72. ©2016 AACR.


Asunto(s)
MicroARNs/genética , Neovascularización Patológica/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Neoplasias de la Mama Triple Negativas/patología , Animales , Western Blotting , Diferenciación Celular , Células Endoteliales/patología , Femenino , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones SCID , Neovascularización Patológica/patología , Reacción en Cadena de la Polimerasa , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Neoplasias de la Mama Triple Negativas/genética
7.
Oncotarget ; 7(29): 45015-45026, 2016 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-27304187

RESUMEN

Sunitinib improves the outcomes of patients with solitary fibrous tumours (SFTs). The aim of this study was to investigate and contextualise sunitinib-induced morpho-functional changes in order to gain insights into the drug's mechanism of action.To this end, four surgical specimens obtained from two sunitinib-responsive patients with malignant SFT, and one primary cell culture obtained from fresh tumoral tissue and its stabilised cell line, were studied by means of immunohistochemistry, bright field in situ hybridisation, immunofluorescence/confocal microscopy, and biochemistry.The post-sunitinib surgical samples were characterised by two biologically relevant morpho-functional changes: clear areas and necrotic foci. The first were associated with the attenuation/loss of PDGFRB expression and decreased mTOR signalling, and corresponded to a pathological response. The second were associated with the over-expression of PDGFRB and VEGFA, strong mTOR signalling activation, and the appearance of HIF1α expression, hallmarks of pathological progression. The analysis clearly showed that sunitinib reduces the vascular supply network and inhibits tumoral cells. It also either induces autophagy, thus favouring drug response, or impairs autophagy as a result of lysosome sequestration, thus favouring disease progression. These distinct autophagic events were associated with different myeloid immune contextures. Finally, we also found that PDGFRB is one of the components of a complex that includes Beclin 1 and VPS34.The results of these tissue-based analyses provide new insights into sunitinib's mechanism of action in SFT patients.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Indoles/farmacología , Pirroles/farmacología , Tumores Fibrosos Solitarios/patología , Inhibidores de la Angiogénesis/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Masculino , Tumores Fibrosos Solitarios/tratamiento farmacológico , Sunitinib , Transcriptoma/efectos de los fármacos
9.
Endocr Relat Cancer ; 22(5): 759-75, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26206776

RESUMEN

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.


Asunto(s)
Carcinoma/patología , Indoles/farmacología , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Metaboloma , Mitosis/efectos de los fármacos , Proteoma/análisis , Neoplasias de la Tiroides/patología , Apoptosis , Biomarcadores de Tumor/metabolismo , Western Blotting , Carcinoma/tratamiento farmacológico , Carcinoma/metabolismo , Carcinoma Papilar , Ciclo Celular , Proliferación Celular , Cromatografía Liquida/métodos , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Receptores Inmunológicos/metabolismo , Espectrometría de Masas en Tándem/métodos , Cáncer Papilar Tiroideo , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/metabolismo , Células Tumorales Cultivadas
10.
J Oral Pathol Med ; 44(9): 734-45, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25495427

RESUMEN

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.


Asunto(s)
Carcinoma de Células Escamosas/virología , Neoplasias Orofaríngeas/enzimología , Neoplasias Orofaríngeas/virología , Papillomaviridae/aislamiento & purificación , Infecciones por Papillomavirus/enzimología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Anfirregulina/genética , Anfirregulina/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/patología , Humanos , Neoplasias de la Boca/patología , Mutación , Neoplasias Orofaríngeas/patología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Papillomaviridae/genética , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/virología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Tirosina Quinasas Receptoras/análisis , Proteínas Tirosina Quinasas Receptoras/genética , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Factor de Crecimiento Transformador alfa/genética , Factor de Crecimiento Transformador alfa/metabolismo
11.
Am J Pathol ; 184(3): 618-30, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24521760

RESUMEN

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.


Asunto(s)
Biomarcadores de Tumor/aislamiento & purificación , Herpesvirus Humano 8/fisiología , Linfoma de Efusión Primaria/metabolismo , Sarcoma de Kaposi/metabolismo , Linfocitos B/metabolismo , Linfocitos B/patología , Línea Celular Tumoral , Biología Computacional , Humanos , Linfoma de Efusión Primaria/patología , Proteoma/química , Sarcoma de Kaposi/patología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Espectrometría de Masas en Tándem
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA