Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
3.
Ann Oncol ; 29(5): 1286-1291, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29509837

ABSTRACT

Background: Hepatocellular carcinomas (HCCs) are not routinely biopsied, resulting in a lack of tumor materials for molecular profiling. Here we sought to determine whether plasma-derived cell-free DNA (cfDNA) captures the genetic alterations of HCC in patients who have not undergone systemic therapy. Patients and methods: Frozen biopsies from the primary tumor and plasma were synchronously collected from 30 prospectively recruited, systemic treatment-naïve HCC patients. Deep sequencing of the DNA from the biopsies, plasma-derived cfDNA and matched germline was carried out using a panel targeting 46 coding and non-coding genes frequently altered in HCCs. Results: In 26/30 patients, at least one somatic mutation was detected in biopsy and/or cfDNA. Somatic mutations in HCC-associated genes were present in the cfDNA of 63% (19/30) of the patients and could be detected 'de novo' without prior knowledge of the mutations present in the biopsy in 27% (8/30) of the patients. Mutational load and the variant allele fraction of the mutations detected in the cfDNA positively correlated with tumor size and Edmondson grade. Crucially, among the seven patients in whom the largest tumor was ≥5 cm or was associated with metastasis, at least one mutation was detected 'de novo' in the cfDNA of 86% (6/7) of the cases. In these patients, cfDNA and tumor DNA captured 87% (80/92) and 95% (87/92) of the mutations, suggesting that cfDNA and tumor DNA captured similar proportions of somatic mutations. Conclusion: In patients with high disease burden, the use of cfDNA for genetic profiling when biopsy is unavailable may be feasible. Our results support further investigations into the clinical utility of cfDNA in a larger cohort of patients.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Circulating Tumor DNA/genetics , Liver Neoplasms/genetics , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Biopsy/methods , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/pathology , Circulating Tumor DNA/blood , DNA Mutational Analysis/methods , Feasibility Studies , Female , High-Throughput Nucleotide Sequencing , Humans , Liver/pathology , Liver Neoplasms/blood , Liver Neoplasms/pathology , Male , Middle Aged , Mutation , Pilot Projects , Tumor Burden/genetics
4.
Cell Death Differ ; 24(6): 1134, 2017 06.
Article in English | MEDLINE | ID: mdl-28106885

ABSTRACT

This corrects the article DOI: 10.1038/cdd.2010.65.

5.
Oncogene ; 32(34): 4001-8, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-22964638

ABSTRACT

Glioblastoma is the most frequent brain tumor in adults and is the most lethal form of human cancer. Despite the improvements in treatments, survival of patients remains poor. To define novel pathways that regulate susceptibility to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in glioma, we have performed genome-wide expression profiling of microRNAs (miRs). We show that in TRAIL-resistant glioma cells, levels of different miRs are increased, and in particular, miR-30b/c and -21. We demonstrate that these miRs impair TRAIL-dependent apoptosis by inhibiting the expression of key functional proteins. T98G-sensitive cells treated with miR-21 or -30b/c become resistant to TRAIL. Furthermore, we demonstrate that miR-30b/c and miR-21 target respectively the 3' untranslated region of caspase-3 and TAp63 mRNAs, and that those proteins mediate some of the effects of miR-30 and -21 on TRAIL resistance, even in human glioblastoma primary cells and in lung cancer cells. In conclusion, we show that high expression levels of miR-21 and -30b/c are needed to maintain the TRAIL-resistant phenotype, thus making these miRs as promising therapeutic targets for TRAIL resistance in glioma.


Subject(s)
Apoptosis/drug effects , MicroRNAs/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , 3' Untranslated Regions/genetics , Blotting, Northern , Blotting, Western , Caspase 3/genetics , Caspase 3/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/metabolism , Glioma/pathology , HEK293 Cells , Humans , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
6.
J Signal Transduct ; 2012: 735135, 2012.
Article in English | MEDLINE | ID: mdl-22685651

ABSTRACT

Gliomas are the most common primary central nervous system tumors with a dismal prognosis. Despite recent advances in surgery, radiotherapy, and chemotherapy, current treatment regimens have a modest survival benefit. A crucial challenge is to deliver drugs effectively to invasive glioma cells residing in a sanctuary within the central nervous system. New therapies are essential, and oligonucleotide-based approaches, including antisense, microRNAs, small interfering RNAs, and nucleic acid aptamers, may provide a viable strategy. Thanks to their unique characteristics (low size, good affinity for the target, no immunogenicity, chemical structures that can be easily modified to improve their in vivo applications), these molecules may represent a valid alternative to antibodies particularly to overcome challenges presented by the blood-brain barrier. Here we will discuss recent results on the use of oligonucleotides that will hopefully provide new effective treatment for gliomas.

7.
Curr Mol Med ; 12(1): 27-33, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22082479

ABSTRACT

miRNAs are small non-coding RNAs of ~24 nt that can block mRNA translation and/or negatively regulate its stability. There is a large body of evidence that dysregulation of miRNAs is a hallmark of cancer. miRNAs are often aberrantly expressed and their function is linked to the regulation of oncogenes and/or tumor suppressor genes involved in cell signaling pathway. miR-221 and miR-222 are two highly homologous microRNAs, whose upregulation has been recently described in several types of human tumors. miR-221/222 have been considered to act as oncogenes or tumor suppressors, depending on tumor system. Silencing oncomiRs or gene therapy approaches, based on re-expression of miRNAs that are down-regulated in cancer cells, could represent a novel anti-tumor approach for integrated cancer therapy. Here we will review the role of miR-221/222 in cancer progression and their use as prognostic and therapeutic tools in cancer.


Subject(s)
Disease Progression , MicroRNAs/genetics , Neoplasms/genetics , Animals , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , MicroRNAs/metabolism , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology
8.
Oncogene ; 31(7): 858-68, 2012 Feb 16.
Article in English | MEDLINE | ID: mdl-21743492

ABSTRACT

Glioblastoma is the most frequent brain tumor in adults and is the most lethal form of human cancer. Despite the improvements in treatments, survival of patients remains poor. In order to identify microRNAs (miRs) involved in glioma tumorigenesis, we evaluated, by a miRarray, differential expression of miRs in the tumorigenic glioma LN-18, LN-229 and U87MG cells compared with the non-tumorigenic T98G cells. Among different miRs we focused our attention on miR-221 and -222. We demonstrated the presence of a binding site for these two miRs in the 3' untranslated region of the protein tyrosine phosphatase µ (PTPµ). Previous studies indicated that PTPµ suppresses cell migration and is downregulated in glioblastoma. Significantly, we found that miR-221 and -222 overexpression induced a downregulation of PTPµ as analyzed by both western blot and real-time PCR. Furthermore, miR-222 and -221 induced an increase in cell migration and growth in soft agar in glioma cells. Interestingly, the re-expression of PTPµ gene was able to revert the miR-222 and -221 effects on cell migration. Furthermore, we found an inverse correlation between miR-221 and -222 and PTPµ in human glioma cancer samples. In conclusion, our results suggest that miR-221 and -222 regulate glioma tumorigenesis at least in part through the control of PTPµ protein expression.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , MicroRNAs/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , 3' Untranslated Regions/genetics , Animals , Binding Sites/genetics , Blotting, Western , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Movement , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , In Situ Hybridization , Mice , Mice, Nude , MicroRNAs/metabolism , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Oligonucleotide Array Sequence Analysis , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Burden/genetics
9.
Cell Death Dis ; 2: e155, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21562587

ABSTRACT

Contrast-induced nephropathy accounts for >10% of all causes of hospital-acquired renal failure, causes a prolonged in-hospital stay and represents a powerful predictor of poor early and late outcome. Mechanisms of contrast-induced nephropathy are not completely understood. In vitro data suggests that contrast media (CM) induces a direct toxic effect on renal tubular cells through the activation of the intrinsic apoptotic pathway. It is unclear whether this effect has a role in the clinical setting. In this work, we evaluated the effects of CM both in vivo and in vitro. By analyzing urine samples obtained from patients who experienced contrast-induced acute kidney injury (CI-AKI), we verified, by western blot and immunohistochemistry, that CM induces tubular renal cells apoptosis. Furthermore, in cultured cells, CM caused a dose-response increase in reactive oxygen species (ROS) production, which triggered Jun N-terminal kinases (JNK1/2) and p38 stress kinases marked activation and thus apoptosis. Inhibition of JNK1/2 and p38 by different approaches (i.e. pharmacological antagonists and transfection of kinase-death mutants of the upstream p38 and JNK kinases) prevented CM-induced apoptosis. Interestingly, N-acetylcysteine inhibited ROS production, and thus stress kinases and apoptosis activation. Therefore, we conclude that CM-induced tubular renal cells apoptosis represents a key mechanism of CI-AKI.


Subject(s)
Acute Kidney Injury/pathology , Apoptosis/drug effects , Contrast Media/adverse effects , Kidney Tubules/pathology , Signal Transduction/drug effects , Acute Kidney Injury/chemically induced , Adult , Aged , Aged, 80 and over , Caspase 3/metabolism , Cells, Cultured , Enzyme Assays , Female , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Kidney Tubules/drug effects , Male , Middle Aged , Reactive Oxygen Species/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism
10.
Cell Death Differ ; 17(12): 1908-16, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20508645

ABSTRACT

Akt is a serine-threonine kinase that has an important role in transducing survival signals. Akt also regulates a number of proteins involved in the apoptotic process. To find new Akt interactors, we performed a two-hybrid screening in yeast using full-length Akt cDNA as bait and a human cDNA heart library as prey. Among 200 clones obtained, two of them were identified as coding for the c-FLIP(L) protein. c-FLIP(L) is an endogenous inhibitor of death receptor-induced apoptosis through the caspase-8 pathway. Using co-immunoprecipitation experiments of either transfected or endogenous proteins, we confirmed the interaction between Akt and c-FLIP(L). Furthermore, we observed that c-FLIP(L) overexpression interferes with Gsk3-ß phosphorylation levels. Moreover, through its effects on Gsk3ß, c-FLIP(L) overexpression in cancer cells induced resistance to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). This effect was mediated by the regulation of p27(Kip1) and caspase-3 expression. These results indicate the existence of a new mechanism of resistance to TRAIL in cancer cells, and unexpected functions of c-FLIP(L).


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Glycogen Synthase Kinase 3/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 3/metabolism , Caspase 8/metabolism , Cell Line , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinase Inhibitor p27/physiology , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Lithium Chloride/pharmacology , Phosphorylation , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/pharmacology
11.
Oncogene ; 27(27): 3845-55, 2008 Jun 19.
Article in English | MEDLINE | ID: mdl-18246122

ABSTRACT

To define novel pathways that regulate susceptibility to tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) in non-small cell lung cancer (NSCLC), we have performed genome-wide expression profiling of microRNAs (miRs). We show that in TRAIL-resistant NSCLC cells, levels of different miRs are increased, and in particular, miR-221 and -222. We demonstrate that these miRs impair TRAIL-dependent apoptosis by inhibiting the expression of key functional proteins. Indeed, transfection with anti-miR-221 and -222 rendered CALU-1-resistant cells sensitive to TRAIL. Conversely, H460-sensitive cells treated with -221 and -222 pre-miRs become resistant to TRAIL. miR-221 and -222 target the 3'-UTR of Kit and p27(kip1) mRNAs, but interfere with TRAIL signaling mainly through p27(kip1). In conclusion, we show that high expression levels of miR-221 and -222 are needed to maintain the TRAIL-resistant phenotype, thus making these miRs as promising therapeutic targets or diagnostic tool for TRAIL resistance in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , MicroRNAs/genetics , TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Apoptosis/drug effects , Calcium-Binding Proteins/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Humans , Lung Neoplasms/pathology , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, Tumor Necrosis Factor/genetics , TNF-Related Apoptosis-Inducing Ligand/toxicity , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...