Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Int J Mol Sci ; 25(7)2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38612777

RESUMEN

High-grade gliomas (HGGs) and glioblastoma multiforme (GBM) are characterized by a heterogeneous and aggressive population of tissue-infiltrating cells that promote both destructive tissue remodeling and aberrant vascularization of the brain. The formation of defective and permeable blood vessels and microchannels and destructive tissue remodeling prevent efficient vascular delivery of pharmacological agents to tumor cells and are the significant reason why therapeutic chemotherapy and immunotherapy intervention are primarily ineffective. Vessel-forming endothelial cells and microchannel-forming glial cells that recapitulate vascular mimicry have both infiltration and destructive remodeling tissue capacities. The transmembrane protein TMEM230 (C20orf30) is a master regulator of infiltration, sprouting of endothelial cells, and microchannel formation of glial and phagocytic cells. A high level of TMEM230 expression was identified in patients with HGG, GBM, and U87-MG cells. In this study, we identified candidate genes and molecular pathways that support that aberrantly elevated levels of TMEM230 play an important role in regulating genes associated with the initial stages of cell infiltration and blood vessel and microchannel (also referred to as tumor microtubule) formation in the progression from low-grade to high-grade gliomas. As TMEM230 regulates infiltration, vascularization, and tissue destruction capacities of diverse cell types in the brain, TMEM230 is a promising cancer target for heterogeneous HGG tumors.


Asunto(s)
Glioblastoma , Glioma , Enfermedad de Parkinson , Humanos , Glioblastoma/genética , Proteínas de la Membrana/genética , Células Endoteliales , Angiogénesis , Glioma/genética , Neuroglía , Neovascularización Patológica/genética
2.
Front Cell Neurosci ; 15: 703431, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34867197

RESUMEN

Glioblastomas (GBM) are the most aggressive tumors originating in the brain. Histopathologic features include circuitous, disorganized, and highly permeable blood vessels with intermittent blood flow. These features contribute to the inability to direct therapeutic agents to tumor cells. Known targets for anti-angiogenic therapies provide minimal or no effect in overall survival of 12-15 months following diagnosis. Identification of novel targets therefore remains an important goal for effective treatment of highly vascularized tumors such as GBM. We previously demonstrated in zebrafish that a balanced level of expression of the transmembrane protein TMEM230/C20ORF30 was required to maintain normal blood vessel structural integrity and promote proper vessel network formation. To investigate whether TMEM230 has a role in the pathogenesis of GBM, we analyzed its prognostic value in patient tumor gene expression datasets and performed cell functional analysis. TMEM230 was found necessary for growth of U87-MG cells, a model of human GBM. Downregulation of TMEM230 resulted in loss of U87 migration, substratum adhesion, and re-passaging capacity. Conditioned media from U87 expressing endogenous TMEM230 induced sprouting and tubule-like structure formation of HUVECs. Moreover, TMEM230 promoted vascular mimicry-like behavior of U87 cells. Gene expression analysis of 702 patients identified that TMEM230 expression levels distinguished high from low grade gliomas. Transcriptomic analysis of patients with gliomas revealed molecular pathways consistent with properties observed in U87 cell assays. Within low grade gliomas, elevated TMEM230 expression levels correlated with reduced overall survival independent from tumor subtype. Highest level of TMEM230 correlated with glioblastoma and ATP-dependent microtubule kinesin motor activity, providing a direction for future therapeutic intervention. Our studies support that TMEM230 has both glial tumor and endothelial cell intracellular and extracellular functions. Elevated levels of TMEM230 promote glial tumor cell migration, extracellular scaffold remodeling, and hypervascularization and abnormal formation of blood vessels. Downregulation of TMEM230 expression may inhibit both low grade glioma and glioblastoma tumor progression and promote normalization of abnormally formed blood vessels. TMEM230 therefore is both a promising anticancer and antiangiogenic therapeutic target for inhibiting GBM tumor cells and tumor-driven angiogenesis.

3.
Oncol Lett ; 21(6): 468, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33907578

RESUMEN

In total, ~25% of familial breast cancer (BC) is attributed to germline mutations of the BRCA1 and BRCA2 genes, while the rest of the cases are included in the BRCAX group. BC is also known to affect men, with a worldwide incidence of 1%. Epigenetic alterations, including DNA methylation, have been rarely studied in male breast cancer (MBC) on a genome-wide level. The aim of the present study was to examine the global DNA methylation profiles of patients with BC to identify differences between familial female breast cancer (FBC) and MBC, and according to BRCA1, BRCA2 or BRCAX mutation status. The genomic DNA of formalin-fixed paraffin-embedded tissues from 17 women and 7 men with BC was subjected to methylated DNA immunoprecipitation and hybridized on human promoter microarrays. The comparison between FBC and MBC revealed 2,846 significant differentially methylated regions corresponding to 2,486 annotated genes. Gene Ontology enrichment analysis revealed molecular function terms, such as the GTPase superfamily genes (particularly the GTPase Rho GAP/GEF and GTPase RAB), and cellular component terms associated with cytoskeletal architecture, such as 'cytoskeletal part', 'keratin filament' and 'intermediate filament'. When only FBC was considered, several cancer-associated pathways were among the most enriched KEGG pathways of differentially methylated genes when the BRCA2 group was compared with the BRCAX or BRCA1+BRCAX groups. The comparison between the BRCA1 and BRCA2+BRCAX groups comprised the molecular function term 'cytoskeletal protein binding'. Finally, the functional annotation of differentially methylated genes between the BRCAX and BRCA1+BRCA2 groups indicated that the most enriched molecular function terms were associated with GTPase activity. In conclusion, to the best of our knowledge, the present study was the first to compare the global DNA methylation profile of familial FBC and MBC. The results may provide useful insights into the epigenomic subtyping of BC and shed light on a possible novel molecular mechanism underlying BC carcinogenesis.

4.
Int J Mol Med ; 44(2): 768, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31173160

RESUMEN

After the publication of the above paper, the authors noted that the names of a couple of the authors listed on the paper were associated with the wrong affliation: Specifically, the eighth and ninth listed authors, Francesca Antonaros and Allison Piovesan, are located at DIMES at the University of Florence (fourth affiliation address), not at CSGI, the Research Center for Colloids and Nanoscience in Florence (third affliation address). Therefore, the author and affiliation details for this paper should have been presented as follows: ALESSANDRO SALVI1, MARIKA VEZZOLI2, SARA BUSATTO1, LUCIA PAOLINI1,3, TERESA FARANDA1, EDOARDO ABENI1, MARIA CARACAUSI4, FRANCESCA ANTONAROS4, ALLISON PIOVESAN4, CHIARA LOCATELLI5, GUIDO COCCHI5,6, GUALTIERO ALVISI7, GIUSEPPINA DE PETRO1, DORIS RICOTTA1, PAOLO BERGESE1,3 and ANNALISA RADEGHIERI1,3. 1Department of Molecular and Translational Medicine, University of Brescia; 2Unit of Biostatistics, Department of Molecular and Translational Medicine, University of Brescia, I­25123 Brescia; 3CSGI, Research Center for Colloids and Nanoscience, Sesto Fiorentino, I­50019 Florence; 4Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Unit of Histology, Embryology and Applied Biology, University of Bologna; 5Neonatology Unit, St. Orsola­Malpighi Polyclinic; 6Department of Medical and Surgical Sciences (DIMEC), University of Bologna, I­40138 Bologna; 7Department of Molecular Medicine, University of Padua, I­35121 Padua, Italy. The authors regret that this error with the author affiliations for Francesca Antonaros and Allison Piovesan was not noticed prior to the publication of their paper, and apologize for any inconvenience caused. [the original article was published in International Journal of Molecular Medicine 43: 2303­2318, 2018; DOI: 10.3892/ijmm.2019.4158].

5.
Int J Mol Med ; 43(6): 2303-2318, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31017260

RESUMEN

Down syndrome (DS) is caused by the presence of part or all of a third copy of chromosome 21. DS is associated with several phenotypes, including intellectual disability, congenital heart disease, childhood leukemia and immune defects. Specific microRNAs (miRNAs/miR) have been described to be associated with DS, although none of them so far have been unequivocally linked to the pathology. The present study focuses to the best of our knowledge for the first time on the miRNAs contained in nanosized RNA carriers circulating in the blood. Fractions enriched in nanosized RNA­carriers were separated from the plasma of young participants with DS and their non­trisomic siblings and miRNAs were extracted. A microarray­based analysis on a small cohort of samples led to the identification of the three most abundant miRNAs, namely miR­16­5p, miR­99b­5p and miR­144­3p. These miRNAs were then profiled for 15 pairs of DS and non­trisomic sibling couples by reverse transcription­quantitative polymerase chain reaction (RT­qPCR). Results identified a clear differential expression trend of these miRNAs in DS with respect to their non­trisomic siblings and gene ontology analysis pointed to their potential role in a number of typical DS features, including 'nervous system development', 'neuronal cell body' and certain forms of 'leukemia'. Finally, these expression levels were associated with certain typical quantitative and qualitative clinical features of DS. These results contribute to the efforts in defining the DS­associated pathogenic mechanisms and emphasize the importance of properly stratifying the miRNA fluid vehicles in order to probe biomolecules that are otherwise hidden and/or not accessible to (standard) analysis.


Asunto(s)
Síndrome de Down/genética , Perfilación de la Expresión Génica , MicroARNs/genética , Adolescente , Adulto , Niño , Preescolar , Estudios de Cohortes , Síndrome de Down/sangre , Femenino , Humanos , Masculino , MicroARNs/sangre , MicroARNs/aislamiento & purificación , Nanopartículas/química , Adulto Joven
6.
Int J Genomics ; 2017: 5913195, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29038785

RESUMEN

MicroRNAs (miRNAs) are a class of small noncoding RNAs that act mainly as negative regulators of gene expression. Several studies demonstrated that miRNAs take part in numerous biological processes, such as proliferation, apoptosis, and migration. The dysregulation of miRNAs has been frequently observed in different types of disease, including cancer. Here, we provide a comprehensive review on the human miR-193a-3p by considering its role in both physiological and pathological contexts. Different mechanisms involved in regulating miR-193a-3p expression have been reported, including epigenetic modifications and transcription factors. In physiological contexts, miR-193a-3p seemed able to limit proliferation and cell cycle progression in normal cells. Remarkably, several publications demonstrated that miR-193a-3p acted as a tumor suppressor miRNA in cancer by targeting different genes involved in proliferation, apoptosis, migration, invasion, and metastasis. Furthermore, the downregulation of miR-193a-3p has been observed in many primary tumors and altered levels of circulating miR-193a-3p have been identified in serum or plasma of cancer patients and subjects affected by Parkinson's disease or by schizophrenia. In a clinical perspective, further studies are needed to explore the antitumor effects of the miR-193a-3p mimics delivery and the relevance of this miRNA detection as a possible diagnostic and prognostic biomarker.

7.
Int J Oncol ; 51(1): 128-144, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28560380

RESUMEN

Sorafenib is currently used to treat advanced and/or unresectable hepatocellular carcinoma (HCC), but the increase of the median survival was only 3 months. Moreover, sorafenib has severe side effects and patients develop resistance quickly. Epigenetic alterations such as DNA methylation play a decisive role in the development and progression of HCC. To our knowledge, there are no studies that analysed the global DNA methylation changes in HCC cells treated with sorafenib. Using MeDip-chip technologies, we found 1230 differentially methylated genes in HA22T/VGH cells treated with sorafenib compared to untreated cells. Gene ontology and pathway analysis allowed identifying several enriched signaling pathways involved in tumorigenesis and cancer progression. Among the genes differentially methylated we found genes related to apoptosis, angiogenesis and invasion, and genes belonging to pathways known to be deregulated in HCC such as RAF/MEK/ERK, JAK-STAT, PI3K/AKT/mTOR and NF-κB. Generally, we found that oncogenes tended to be hypermethylated and the tumor suppressor genes tended to be hypomethylated after sorafenib treatment. Finally, we validated MeDip-chip results for several genes found differentially methylated such as BIRC3, FOXO3, MAPK3, SMAD2 and TSC2, using both COBRA assay and direct bisulfite sequencing and we evaluated their mRNA expression. Our findings suggest that sorafenib could affect the methylation level of genes associated to cancer-related processes and pathways in HCC cells, some of which have been previously described to be directly targeted by sorafenib.


Asunto(s)
Antineoplásicos/farmacología , Biomarcadores de Tumor/genética , Carcinoma Hepatocelular/genética , Metilación de ADN/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Hepáticas/genética , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Niacinamida/farmacología , Regiones Promotoras Genéticas/genética , Transducción de Señal , Sorafenib , Células Tumorales Cultivadas
8.
Int J Oncol ; 46(5): 1901-12, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25760690

RESUMEN

Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related mortality worldwide. We have previously reported that LASP-1 is a downstream protein of the urokinase type plasminogen activator (uPA). Here we investigated the role of LASP-1 in HCC by a molecular and biological characterization of LASP-1 expression in human HCC specimens and in cultured HCC cells. We determined the LASP-1 mRNA expression levels in 55 HCC cases with different hepatic background disease. We identified 3 groups of patients with high, equal or low LASP-1 mRNA levels in HCC tissues compared to the peritumoral (PT) tissues. In particular we found that i) the HCCs displayed a higher LASP-1 mRNA level in HCC compared to PT tissues; ii) the expression levels of LASP-1 mRNA in female HCCs were significantly higher compared to male HCCs; iii) the cirrhotic HCCs displayed a higher LASP-1 mRNA. Further, the biological characterization of the ectopic LASP-1 overexpression in HCC cells, using MALDI-TOF mass spectrometer on the LASP-1 co-immunoprecipitated fractions, displayed vimentin as a novel putative partner of LASP-1. Our results suggest that LASP-1 mRNA overexpression may be mainly implicated in female HCCs and cirrhotic HCCs; and that LASP1 may play its role with vimentin in HCC cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Carcinoma Hepatocelular/genética , Proteínas del Citoesqueleto/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Proteínas con Dominio LIM/genética , Neoplasias Hepáticas/genética , Vimentina/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/genética , Factores Sexuales , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Células Tumorales Cultivadas
9.
Mol Cancer ; 12: 162, 2013 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-24330766

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) is a challenging malignancy of global importance, it is the third most common cause of cancer-related mortality worldwide. In the last years the multikinase inhibitor sorafenib has been used for advanced HCC, but some patients do not benefit from this therapy; thus, novel therapeutic options based on molecular approaches are urgently needed. microRNAs are short non coding RNAs involved in several physiological and pathological conditions including HCC and increasing evidence describes miRs as good tools for the molecular targeted therapies in HCC. The purpose of this study was to identify novel approaches to sensitize the HCC cells to sorafenib by microRNAs targeting urokinase-type plasminogen activator (uPA). METHODS: The miR-193a was validated as negative regulator of urokinase-type plasminogen activator (uPA) in 2 HCC undifferentiated cell lines by transient transfection of miR and anti-miR molecules. The molecular interaction between miR-193a and uPA mRNA target was verified by luciferase reporter assay. The miR-193a expression level was evaluated by stem-loop real time PCR in tumoral tissues from 39 HCC patients. The HCC cells were co-treated with sorafenib and miR-193a and the effects on cellular proliferation, apoptosis were tested. The effect of sorafenib on c-met expression levels was assessed by western blotting. RESULTS: The miR-193a has resulted a negative regulator of uPA in both the HCC cell lines tested. The miR-193a expression has resulted dysregulated in tumoral tissues from 39 HCC patients. We found miR-193a down-regulation in HCC respect to peritumoral (PT) tissues and more in the cirrhotic HCCs than in non-cirrhotic ones. Transfection of HA22T/VGH HCC cells with miR-193a decreased proliferation and increased apoptosis, and combined treatment with miR-193a and sorafenib led to further proliferation inhibition. CONCLUSIONS: Our results present new advances in the post-transcriptional miR-mediated mechanisms of uPA and they suggest a new strategy to impair the aggressive behavior of HCC cells. Our findings could be helpful to explore novel approaches for multi-target and multi-agent therapies of the HCC.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , MicroARNs/metabolismo , MicroARNs/farmacología , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , MicroARNs/genética , Niacinamida/farmacología , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Sorafenib
10.
Int J Oncol ; 42(2): 391-402, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23229173

RESUMEN

microRNAs (miRs) are 18-25 nucleotide non-coding RNAs that regulate gene expression in several physiological and pathological conditions. To gather more knowledge on microRNAs in human hepatocellular carcinoma (HCC) we generated a small RNA library in the human HCC cell line HA22T/VGH by cloning and sequencing the cDNA obtained following the size selection of 18-24 nucleotide RNAs. We determined the expression levels of the most frequently cloned microRNAs by qPCR in HCC tissues and in their peritumoral counterparts from biopsy specimens of 41 HCC patients. The most frequently cloned miRs were miR-24, miR-27a and miR-21, and their expression levels in human HCC tissues indicate that these miRs were dysregulated in HCC. We showed that miR-24 and miR-27a were significantly downregulated in HCCs from cirrhotic liver tissues in comparison to those from non-cirrhotic liver tissues. In cirrhotic HCCs the downregulation of miR-24 was correlated with poorer prognosis in patients with HBV and HCV virus infections. miR-21 was generally upregulated in HCC tissues versus the corresponding peritumoral tissues, particularly in non-cirrhotic HCC. Furthermore, by sequence alignment we identified the human miR orthologue of Mus musculus miR-1199 not yet annotated. Our results outline the differential expression of miRs in cirrhotic and non-cirrhotic HCCs, thereby contributing to advances in the discovery and validation of novel molecular biomarkers of HCC progression.


Asunto(s)
Carcinoma Hepatocelular/genética , Cirrosis Hepática/genética , Neoplasias Hepáticas/genética , MicroARNs/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Carcinoma Hepatocelular/complicaciones , Carcinoma Hepatocelular/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Cirrosis Hepática/complicaciones , Cirrosis Hepática/patología , Neoplasias Hepáticas/complicaciones , Neoplasias Hepáticas/patología , Masculino , Ratones , Persona de Mediana Edad , Estadificación de Neoplasias , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...