Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Eur Rev Med Pharmacol Sci ; 28(8): 3268-3274, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38708485

ABSTRACT

BACKGROUND: We describe the first case of a pediatric patient with acute intermittent porphyria and severe chronic porphyric neuropathy treated with givosiran, a small-interfering RNA that drastically decreases delta-aminolevulinic acid production and reduces porphyric attacks' recurrence. CASE REPORT: A 12-year-old male patient with refractory acute intermittent porphyria and severe porphyric neuropathy was followed prospectively for 12 months after givosiran initiation (subcutaneous, 2.5 mg/kg monthly). Serial neurological, structural, and resting-state functional magnetic resonance imaging (MRI) evaluations were performed, including clinical scales and neurophysiological tests. Delta-aminolevulinic acid urinary levels dropped drastically during treatment. In parallel, all the administered neurological rating scales and neurophysiological assessments showed improvement in all domains. Moreover, an improvement in central motor conduction parameters and resting-state functional connectivity in the sensory-motor network was noticed. At the end of the follow-up, the patient could walk unaided after using a wheelchair for 5 years. CONCLUSIONS: A clear beneficial effect of givosiran was demonstrated in our patient with both clinical and peripheral nerve neurophysiologic outcome measures. Moreover, we first reported a potential role of givosiran in recovering central motor network impairment in acute intermittent porphyria (AIP), which was previously unknown. This study provides Class IV evidence that givosiran improves chronic porphyric neuropathy.


Subject(s)
Acetylgalactosamine/analogs & derivatives , Porphyria, Acute Intermittent , Humans , Male , Porphyria, Acute Intermittent/drug therapy , Child , Acetylgalactosamine/therapeutic use , Aminolevulinic Acid/analogs & derivatives , Aminolevulinic Acid/urine , Magnetic Resonance Imaging , Pyrrolidines/therapeutic use , Uridine/analogs & derivatives , Uridine/therapeutic use , Uridine/administration & dosage , Recovery of Function , Chronic Disease , Treatment Outcome
2.
ESMO Open ; 7(5): 100585, 2022 10.
Article in English | MEDLINE | ID: mdl-36156447

ABSTRACT

BACKGROUND: The detection of homologous recombination deficiency (HRD) can identify patients who are more responsive to platinum and poly ADP ribose polymerase inhibitors (PARPi). MyChoice CDx (Myriad) is the most used HRD test in ovarian cancer (OC). However, some limitations of commercial tests exist, because of the high rate of inconclusive results, costs, and the impossibility of evaluating functional resistance mechanisms. PATIENTS AND METHODS: Two academic genomic tests and a functional assay, the RAD51 foci, were evaluated to detect HRD. One hundred patients with high-grade OC enrolled in the MITO16A/MaNGO-OV2 trial and treated with first-line therapy with carboplatin, paclitaxel, and bevacizumab were analyzed. RESULTS: The failure rate of the two genomic assays was 2%. The sensitivity in detecting HRD when compared with Myriad was 98.1% and 90.6%, respectively. The agreement rate with Myriad was 0.92 and 0.87, with a Cohen's κ coefficient corresponding to 0.84 and 0.74, respectively. For the RAD51 foci assay, the failure rate was 30%. When the test was successful, discordant results for deficient and proficient tumors were observed, and additional HRD patients were identified compared to Myriad; sensitivity was 82.9%, agreement rate was 0.65, and Cohen's κ coefficient was 0.18. The HRD detected by genomic assays and residual tumor at primary surgery and stage was correlated with progression-free survival at multivariate analysis. CONCLUSIONS: Results suggest the feasibility of academic tests for assessing HRD status that show robust concordance with Myriad and correlation with clinical outcome. The contribution of the functional information related to the RAD51 foci test to the genomic data needs further investigation.


Subject(s)
Mangifera , Ovarian Neoplasms , Female , Humans , Bevacizumab/therapeutic use , Carboplatin/therapeutic use , Homologous Recombination , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Paclitaxel/therapeutic use , Platinum/therapeutic use , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/therapeutic use
3.
Cell Mol Immunol ; 18(3): 711-722, 2021 03.
Article in English | MEDLINE | ID: mdl-32728200

ABSTRACT

In cancer, myeloid cells have tumor-supporting roles. We reported that the protein GPNMB (glycoprotein nonmetastatic B) was profoundly upregulated in macrophages interacting with tumor cells. Here, using mouse tumor models, we show that macrophage-derived soluble GPNMB increases tumor growth and metastasis in Gpnmb-mutant mice (DBA/2J). GPNMB triggers in the cancer cells the formation of self-renewing spheroids, which are characterized by the expression of cancer stem cell markers, prolonged cell survival and increased tumor-forming ability. Through the CD44 receptor, GPNMB mechanistically activates tumor cells to express the cytokine IL-33 and its receptor IL-1R1L. We also determined that recombinant IL-33 binding to IL-1R1L is sufficient to induce tumor spheroid formation with features of cancer stem cells. Overall, our results reveal a new paracrine axis, GPNMB and IL-33, which is activated during the cross talk of macrophages with tumor cells and eventually promotes cancer cell survival, the expansion of cancer stem cells and the acquisition of a metastatic phenotype.


Subject(s)
Fibrosarcoma/pathology , Hyaluronan Receptors/metabolism , Interleukin-33/metabolism , Lung Neoplasms/pathology , Macrophages/immunology , Membrane Glycoproteins/metabolism , Neoplastic Stem Cells/pathology , Animals , Apoptosis , Cell Proliferation , Fibrosarcoma/etiology , Fibrosarcoma/metabolism , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/genetics , Interleukin-33/genetics , Lung Neoplasms/etiology , Lung Neoplasms/metabolism , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred DBA , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Sarcoma, Experimental/etiology , Sarcoma, Experimental/metabolism , Sarcoma, Experimental/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Brain Behav Immun ; 81: 598-607, 2019 10.
Article in English | MEDLINE | ID: mdl-31336144

ABSTRACT

Activation of Toll-like receptor 3 (TLR3) was previously shown to contribute to the generation of epileptic seizures in rodents by evoking a proinflammatory response in the forebrain. This suggests that TLR3 blockade may provide therapeutic effects in epilepsy. We report that brain activation of TLR3 using the synthetic receptor ligand Poly I:C may also result in remarkable dose- and time-dependent inhibitory effects on acute seizures in mice without inducing inflammation. These inhibitory effects are associated with reduced neuronal excitability in the hippocampus as shown by a decrease in the population spike amplitude of CA1 pyramidal neurons following Schaffer collaterals stimulation. TLR3 activation which results in seizure inhibition does not evoke NF-kB-dependent inflammatory molecules or morphological activation of glia, however, it induces the alternative interferon (IFN) regulatory factor (IRF)-3/IFN-ß signaling pathway. IFN-ß reproduced the inhibitory effects of Poly I:C on neuronal excitability in hippocampal slices. Seizure inhibition attained with activation the TLR3-IRF3/IFN-ß axis should be carefully considered when TLR3 are targeted for therapeutic purposes.


Subject(s)
Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Toll-Like Receptor 3/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Anticonvulsants/pharmacology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Neuroglia/metabolism , Poly I-C/pharmacology , Receptors, Cell Surface/metabolism , Seizures/metabolism , Signal Transduction/drug effects
5.
Pharmacogenomics J ; 18(1): 56-63, 2018 01.
Article in English | MEDLINE | ID: mdl-27958379

ABSTRACT

This study was designed to investigate the mode of action of trabectedin in myelomonocytic leukemia cells by applying systems biology approaches to mine gene expression profiling data and pharmacological assessment of the cellular effects. Significant enrichment was found in regulons of target genes inferred for specific transcription factors, among which MAFB was the most upregulated after treatment and was central in the transcriptional network likely to be relevant for the specific therapeutic effects of trabectedin against myelomonocytic cells. Using the Connectivity Map, similarity among transcriptional signatures elicited by treatment with different compounds was investigated, showing a high degree of similarity between transcriptional signatures of trabectedin and those of the topoisomerase I inhibitor, irinotecan, and an anti-dopaminergic antagonist, thioridazine. The study highlights the potential importance of systems biology approaches to generate new hypotheses that are experimentally testable to define the specificity of the mechanism of action of drugs.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Leukemia, Myeloid/drug therapy , Trabectedin/pharmacology , Cell Line, Tumor , Gene Expression Profiling/methods , Gene Regulatory Networks/drug effects , Humans , Systems Biology/methods , Transcription, Genetic/drug effects
6.
BMC Cancer ; 17(1): 107, 2017 Feb 06.
Article in English | MEDLINE | ID: mdl-28166781

ABSTRACT

BACKGROUND: Desmoplastic small round cell tumor (DSRCT) is a rare and highly aggressive disease, that can be described as a member of the family of small round blue cell tumors. The molecular diagnostic marker is the t(11;22)(p13;q12) translocation, which creates an aberrant transcription factor, EWS-WT1, that underlies the oncogenesis of DSRCT. Current treatments are not very effective so new active drugs are needed. Trabectedin, now used as a single agent for the treatment of soft tissue sarcoma, was reported to be active in some pre-treated DSRCT patients. Using JN-DSRCT-1, a cell line derived from DSRCT expressing the EWS-WT1 fusion protein, we investigated the ability of trabectedin to modify the function of the chimeric protein, as in other sarcomas expressing fusion proteins. After detailed characterization of the EWS-WT1 transcripts structure, we investigated the mode of action of trabectedin, looking at the expression and function of the oncogenic chimera. METHODS: We characterized JN-DSRCT-1 cells using cellular approaches (FISH, Clonogenicity assay) and molecular approaches (Sanger sequencing, ChIP, GEP). RESULTS: JN-DSRCT-1 cells were sensitive to trabectedin at nanomolar concentrations. The cell line expresses different variants of EWS-WT1, some already identified in patients. EWS-WT1 mRNA expression was affected by trabectedin and chimeric protein binding on its target gene promoters was reduced. Expression profiling indicated that trabectedin affects the expression of genes involved in cell proliferation and apoptosis. CONCLUSIONS: The JN-DSRCT-1 cell line, in vitro, is sensitive to trabectedin: after drug exposure, EWS-WT1 chimera expression decreases as well as binding on its target promoters. Probably the heterogeneity of chimera transcripts is an obstacle to precisely defining the molecular mode of action of drugs, calling for further cellular models of DSRCT, possibly growing in vivo too, to mimic the biological complexity of this disease.


Subject(s)
Desmoplastic Small Round Cell Tumor/drug therapy , Dioxoles/pharmacology , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/drug effects , Tetrahydroisoquinolines/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Desmoplastic Small Round Cell Tumor/metabolism , Desmoplastic Small Round Cell Tumor/physiopathology , Dioxoles/therapeutic use , Humans , Oncogene Proteins, Fusion/genetics , RNA-Binding Protein EWS , Tetrahydroisoquinolines/therapeutic use , Trabectedin , WT1 Proteins
7.
Ann Oncol ; 27(8): 1511-9, 2016 08.
Article in English | MEDLINE | ID: mdl-27194815

ABSTRACT

BACKGROUND: Clinical and pathological parameters of patients with epithelial ovarian cancer (EOC) do not thoroughly predict patients' outcome. Despite the good outcome of stage I EOC compared with that of stages III and IV, the risk assessment and treatments are almost the same. However, only 20% of stage I EOC cases relapse and die, meaning that only a proportion of patients need intensive treatment and closer follow-up. Thus, the identification of cell mechanisms that could improve outcome prediction and rationalize therapeutic options is an urgent need in the clinical practice. PATIENTS AND METHODS: We have gathered together 203 patients with stage I EOC diagnosis, from whom snap-frozen tumor biopsies were available at the time of primary surgery before any treatment. Patients, with a median follow-up of 7 years, were stratified into a training set and a validation set. RESULTS AND CONCLUSIONS: Integrated analysis of miRNA and gene expression profiles allowed to identify a prognostic cell pathway, composed of 16 miRNAs and 10 genes, wiring the cell cycle, 'Activins/Inhibins' and 'Hedgehog' signaling pathways. Once validated by an independent technique, all the elements of the circuit resulted associated with overall survival (OS) and progression-free survival (PFS), in both univariate and multivariate models. For each patient, the circuit expressions have been translated into an activation state index (integrated signature classifier, ISC), used to stratify patients into classes of risk. This prediction reaches the 89.7% of sensitivity and 96.6% of specificity for the detection of PFS events. The prognostic value was then confirmed in the external independent validation set in which the PFS events are predicted with 75% sensitivity and 94.7% specificity. Moreover, the ISC shows higher classification performance than conventional clinical classifiers. Thus, the identified circuit enhances the understanding of the molecular mechanisms lagging behind stage I EOC and the ISC improves our capabilities to assess, at the time of diagnosis, the patient risk of relapse.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic/genetics , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Prognosis , Adult , Aged , Carcinoma, Ovarian Epithelial , Disease-Free Survival , Female , Humans , MicroRNAs/genetics , Microarray Analysis , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Staging , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology
8.
Ann Oncol ; 27(4): 625-34, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26782955

ABSTRACT

BACKGROUND: Neoadjuvant chemotherapy (NACT) has been recognized as a reliable therapeutic strategy in patients with unresectable advanced epithelial ovarian cancer (EOC). The molecular events leading to platinum (Pt) response in NACT settings have hitherto not been explored. In the present work, longitudinal changes of miRNA expression profile were investigated to identify miRNA families with prognostic role in high-grade serous EOC patients who received the NACT regimen. PATIENTS AND METHODS: One hundred sixty-four matched tumor biopsies taken at initial laparoscopic evaluation and at interval-debulking surgery (IDS) after four courses of Pt-based therapy were selected from 82 stage IIIC-IV high-grade serous-EOC patients that were judged unsuitable for complete primary debulking and subjected the NACT protocol. miRNA profiling by microarray, real-time PCR and immuno-histochemical staining for Smad2 phosphorylation (P-Smad2) were used for data analysis. RESULTS: Analysis revealed that 369 miRNAs were differentially expressed in matched biopsies (referred to as DEMs). DEMs were not scattered across the genome, but clustered into families: miR-199, let-7, miR-30, miR-181 and miR-29. Multivariate analysis showed that miR-199a-3p, miR-199a-5p, miR-181a-5p and let-7g-5p associated with overall and progression-free survival (P < 0.05); miR-199a-3p, miR-199a-5p and miR-181a-5p associated with residual tumor volume and Pt-free interval (P < 0.05). Immuno-histochemical staining confirmed an enrichment of P-Smad2, a marker of transforming growth factor-ß activation, in tumors from patients with shorter PFS and OS, and with high levels of expression of miR-181a-5p (P < 0.05). Kaplan-Meier curves plotting concomitant expression of P-Smad2 and miR-181a-5p show significant differences in PFS and OS compared with those depicting the expression of each biomarker alone (P < 0.001). CONCLUSIONS: This study describes several miRNA families with a prognostic role in the NACT setting. It also confirms that concomitant analysis of P-Smad2 and miR-181a-5p in surgical samples may be capable of identifying those ovarian cancer patients with poor outcome and little chance of response to Pt-based NACT.


Subject(s)
Cystadenocarcinoma, Serous/drug therapy , MicroRNAs/biosynthesis , Neoadjuvant Therapy , Ovarian Neoplasms/drug therapy , Smad2 Protein/biosynthesis , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Biopsy , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MicroRNAs/genetics , Middle Aged , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Prognosis , Smad2 Protein/genetics
9.
Ann Oncol ; 26(7): 1363-71, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25846551

ABSTRACT

BACKGROUND: The majority of patients with stage III-IV epithelial ovarian cancer (EOC) relapse after initially responding to platinum-based chemotherapy, and develop resistance. The genomic features involved in drug resistance are unknown. To unravel some of these features, we investigated the mutational profile of genes involved in pathways related to drug sensitivity in a cohort of matched tumors obtained at first surgery (Ft-S) and second surgery (Sd-S). PATIENTS AND METHODS: Matched biopsies (33) taken at Ft-S and Sd-S were selected from the 'Pandora' tumor tissue collection. DNA libraries for 65 genes were generated using the TruSeq Custom Amplicon kit and sequenced on MiSeq (Illumina). Data were analyzed using a high-performance cluster computing platform (Cloud4CARE project) and independently validated. RESULTS: A total of 2270 somatic mutations were identified (89.85% base substitutions 8.19% indels, and 1.92% unknown). Homologous recombination (HR) genes and TP53 were mutated in the majority of Ft-S, while ATM, ATR, TOP2A and TOP2B were mutated in the entire dataset. Only 2% of mutations were conserved between matched Ft-S and Sd-S. Mutations detected at second surgery clustered patients in two groups characterized by different mutational profiles in genes associated with HR, PI3K, miRNA biogenesis and signal transduction. CONCLUSIONS: There was a low level of concordance between Ft-S and Sd-S in terms of mutations in genes involved in key processes of tumor growth and drug resistance. This result suggests the importance of future longitudinal analyses to improve the clinical management of relapsed EOC.


Subject(s)
Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Mucinous/genetics , Cystadenocarcinoma, Serous/genetics , Endometrial Neoplasms/genetics , Genes, Neoplasm/genetics , High-Throughput Nucleotide Sequencing/methods , Mutation/genetics , Ovarian Neoplasms/genetics , Adenocarcinoma, Clear Cell/mortality , Adenocarcinoma, Clear Cell/secondary , Adenocarcinoma, Clear Cell/therapy , Adenocarcinoma, Mucinous/mortality , Adenocarcinoma, Mucinous/secondary , Adenocarcinoma, Mucinous/therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biopsy , Combined Modality Therapy , Cystadenocarcinoma, Serous/mortality , Cystadenocarcinoma, Serous/secondary , Cystadenocarcinoma, Serous/therapy , Drug Resistance, Neoplasm/genetics , Endometrial Neoplasms/mortality , Endometrial Neoplasms/secondary , Endometrial Neoplasms/therapy , Female , Follow-Up Studies , Homologous Recombination , Humans , Longitudinal Studies , Lymphatic Metastasis , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/therapy , Neoplasm Staging , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Prognosis , Retrospective Studies , Survival Rate
10.
Oncogene ; 33(44): 5201-10, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-24213580

ABSTRACT

To elucidate the mechanisms behind the high sensitivity of myxoid/round cell liposarcoma (MRCL) to trabectedin and the suggested selectivity for specific subtypes, we have developed and characterized three MRCL xenografts, namely ML017, ML015 and ML004 differing for the break point of the fusion gene FUS-CHOP, respectively of type I, II and III. FUS-CHOP binding to the promoters of some target genes such as Pentraxin 3 or Fibronectin 1, assessed by chromatin immunoprecipitation, was strongly reduced in the tumor 24 h after the first or the third weekly dose of trabectedin, indicating that the drug at therapeutic doses causes a detachment of the FUS-CHOP chimera from its target promoters as previously shown in vitro. Moreover, the higher sensitivity of MRCL types I and II appears to be related to a more prolonged block of the transactivating activity of the fusion protein. Doxorubicin did not affect the binding of FUS-CHOP to target promoters. Histologically, the response to trabectedin in ML017 and ML015 was associated with a marked depletion of non-lipogenic tumoral cells and vascular component, as well as lipidic maturation as confirmed by PPARγ2 expression in western Blot. By contrast, in ML004 no major changes either in the cellularity or in the amount of mature were found, and consistently PPARγ2 was null. In conclusion, the data support the view that the selective mechanism of action of trabectedin in MRCL is specific and related to its ability to cause a functional inactivation of the oncogenic chimera with consequent derepression of the adypocytic differentiation.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Dioxoles/pharmacology , Liposarcoma, Myxoid/drug therapy , Oncogene Proteins, Fusion/genetics , RNA-Binding Protein FUS/genetics , Tetrahydroisoquinolines/pharmacology , Transcription Factor CHOP/genetics , Adult , Animals , Biopsy , Cell Differentiation/drug effects , Cell Differentiation/genetics , Doxorubicin/pharmacology , Female , Humans , Liposarcoma, Myxoid/genetics , Mice, Nude , Oncogene Proteins, Fusion/metabolism , RNA-Binding Protein FUS/metabolism , Trabectedin , Transcription Factor CHOP/metabolism , Xenograft Model Antitumor Assays
11.
Neurobiol Dis ; 59: 183-93, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23938763

ABSTRACT

We studied whether pharmacological blockade of the IL-1ß-mediated signaling, rapidly activated in forebrain by epileptogenic injuries, affords neuroprotection in two different rat models of status epilepticus (SE). As secondary outcome, we measured treatment's effect on SE-induced epileptogenesis. IL-1ß signaling was blocked by systemic administration of two antiinflammatory drugs, namely human recombinant IL-1 receptor antagonist (anakinra), the naturally occurring and clinically used competitive IL-1 receptor type 1 antagonist, and VX-765 a specific non-peptide inhibitor of IL-1ß cleavage and release. Antiinflammatory drugs were given 60min after antiepileptic (AED) drug-controlled SE induced by pilocarpine, or 180min after unrestrained electrical SE, for 7days using a protocol yielding therapeutic drug levels in brain. This drug combination significantly decreased both IL-1ß expression in astrocytes and cell loss in rat forebrain. Neuroprotection and the antiinflammatory effect were more pronounced in the electrical SE model. Onset of epilepsy, and frequency and duration of seizures 3months after electrical SE were not significantly modified. Transcriptomic analysis in the hippocampus showed that the combined treatment did not affect the broad inflammatory response induced by SE during epileptogenesis. In particular, the treatment did not prevent the induction of the complement system and Toll-like receptors, both contributing to cell loss and seizure generation. We conclude that the IL-1ß signaling represents an important target for reducing cell loss after SE. The data highlight a new class of clinically tested agents affording neuroprotection after a delayed post-injury intervention. Earlier blockade of this rapid onset inflammatory pathway during SE, or concomitant treatment with antiinflammatory drugs targeting additional components of the broad inflammatory response to SE, or co-treatment with AEDs, is likely to be required for optimizing beneficial outcomes.


Subject(s)
Epilepsy, Temporal Lobe/metabolism , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin-1beta/metabolism , Receptors, Interleukin-1 Type I/metabolism , Animals , Cell Death/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Dipeptides/therapeutic use , Disease Models, Animal , Electric Stimulation/adverse effects , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/prevention & control , Female , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Interleukin 1 Receptor Antagonist Protein/blood , Interleukin 1 Receptor Antagonist Protein/cerebrospinal fluid , Lithium/toxicity , Male , Pilocarpine/toxicity , Rats , Rats, Sprague-Dawley , para-Aminobenzoates/therapeutic use
12.
Int J Cancer ; 131(1): 59-69, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-21805478

ABSTRACT

Myxoid Liposarcomas (MLS), characterized by the expression of FUS-CHOP fusion gene are clinically very sensitive to the DNA binding antitumor agent, trabectedin. However, resistance eventually occurs, preventing disease eradication. To investigate the mechanisms of resistance, a trabectedin resistant cell line, 402-91/ET, was developed. The resistance to trabectedin was not related to the expression of MDR related proteins, uptake/efflux of trabectedin or GSH levels that were similar in parental and resistant cells. The 402-91/ET cells were hypersensitive to UV light because of a nucleotide excision repair defect: XPG complementation decreased sensitivity to UV rays, but only partially to trabectedin. 402-91/ET cells showed collateral sensitivity to temozolomide due to the lack of O(6) -methylguanine-DNA-methyltransferase (MGMT) activity, related to the hypermethylation of MGMT promoter. In 402-91 cells chromatin immunoprecipitation (ChIP) assays showed that FUS-CHOP was bound to the PTX3 and FN1 gene promoters, as previously described, and trabectedin caused FUS-CHOP detachment from DNA. Here we report that, in contrast, in 402-91/ET cells, FUS-CHOP was not bound to these promoters. Differences in the modulation of transcription of genes involved in different pathways including signal transduction, apoptosis and stress response between the two cell lines were found. Trabectedin activates the transcription of genes involved in the adipogenic-program such as c/EBPα and ß, in 402-91 but not in 402-91/ET cell lines. The collateral sensitivity of 402-91/ET to temozolomide provides the rationale to investigate the potential use of methylating agents in MLS patients resistant to trabectedin.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Cell Line, Tumor , Dioxoles/pharmacology , Liposarcoma, Myxoid/genetics , Liposarcoma, Myxoid/metabolism , Tetrahydroisoquinolines/pharmacology , Apoptosis , C-Reactive Protein/genetics , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-beta/genetics , DNA Methylation , DNA Modification Methylases/deficiency , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair , DNA Repair Enzymes/deficiency , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Drug Resistance, Neoplasm , Fibronectins/genetics , Humans , Liposarcoma, Myxoid/drug therapy , Liposarcoma, Myxoid/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Promoter Regions, Genetic , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Serum Amyloid P-Component/genetics , Signal Transduction , Temozolomide , Trabectedin , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ultraviolet Rays
13.
Cell Mol Biol (Noisy-le-grand) ; 55(2): 70-8, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19656454

ABSTRACT

Coproporphyrin (CP) is one of the main by-products of heme biosynthesis and its abnormal accumulation is associated with different forms of porphyria. Indirect data obtained from animal and human models have suggested a possible role for Multidrug Resistance-associated Protein 2 (MRP2) and other MRPs in hepatocyte excretion of CP. Using normal, MRP2-deficient and a cholestatic rat model, we have assessed the role of MRPs in CP disposition. MRP levels were assayed using immunofluorescence. Biliary and urinary excretion patterns of CP and conjugate bilirubin were measured during equimolar infusions of CP isomers with and without phenoldibromopthalein sulfonate (BSP), a well-known MRP2 substrate. Our results suggest a role for the MRP system as a possible regulator of CP traffic and accumulation in normal and pathological conditions. Alteration in this systems (as observed in cholestatic disease) may play an important role in triggering clinical expression of porphyria in individuals with underlying mutations leading to porphyrin accumulation and may help explain the phenotypic heterogeneity in patients affected by different forms of porphyrias.


Subject(s)
Bile/metabolism , Coproporphyrins/metabolism , Multidrug Resistance-Associated Proteins/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Bilirubin/urine , Biological Transport , Cholestasis/metabolism , Cholestasis/pathology , Coproporphyrins/urine , Disease Models, Animal , Isomerism , Jaundice, Chronic Idiopathic/metabolism , Jaundice, Chronic Idiopathic/pathology , Male , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/deficiency , Multidrug Resistance-Associated Proteins/genetics , Rats , Rats, Wistar
14.
Cell Mol Biol (Noisy-le-grand) ; 55(2): 79-88, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19656455

ABSTRACT

Variegate Porphyria (VP) is an autosomal dominant disorder found worldwide but is rare in Italy. In this study we provide an overview of clinical, biochemical and genetic background of 33 Italian VP patients diagnosed in the last fifteen years. About 70% of patients had experienced clinical symptoms: 43.4% had photosensivity, 8.7% acute attacks and 47.8% both. Among the 33 patients, 14 different mutations were identified. Of these only 6 defects have been previously described in other countries and 8 are unique having been identified for the first time in Italy. Two of these, the c.851G>T and the c.1013C>G, were found in two and four unrelated families respectively. No mutation has been found in homozygosis and no significant correlation has been observed between specific clinical and biochemical manifestations and the type of mutation. In contrast, normal faecal protoporphyrin excretion was high predictive of silent phenotype. Normal urinary excretion of PBG and ALA, predicted absence of neurovisceral symptoms. This paper represents the first compilation of data on genotype-phenotype relation in Italian patients with VP.


Subject(s)
Flavoproteins/genetics , Mitochondrial Proteins/genetics , Porphyria, Variegate/genetics , Protoporphyrinogen Oxidase/genetics , White People/genetics , Adult , Aged , Aminolevulinic Acid/urine , Female , Flavoproteins/metabolism , Genetic Association Studies , Genotype , Humans , Italy , Male , Middle Aged , Mitochondrial Proteins/metabolism , Polymorphism, Genetic , Porphobilinogen/urine , Porphyria, Variegate/diagnosis , Porphyrins/urine , Protoporphyrinogen Oxidase/metabolism
16.
HIV Med ; 10(1): 28-34, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19125962

ABSTRACT

OBJECTIVE: We evaluated hyperhomocysteinaemia (HHcy) in a cohort of HIV-infected patients in order to assess its relation to cardiovascular risk (CVR) and identify determinants of HHcy variability. METHODS: Cross-sectional observational study. HIV-infected patients on stable highly active antiretroviral therapy (ART) were evaluated for the presence of the metabolic syndrome, lipodystrophy and traditional CVR factors. Plasma homocysteine levels were measured using high-performance liquid chromatography. RESULTS: Five hundred and sixty-seven patients (38% female) with a median age of 44 years were included in the study. Homocysteine (Hcy) was significantly higher in patients with the metabolic syndrome and lipodystrophy. No significant association was found between Hcy levels and the use of ART. However, Hcy was associated with higher blood pressure, waist circumference and waist-to-hip ratio, total lean body mass, visceral adipose tissue (VAT), VAT/total adipose tissue, homeostasis model assessment of insulin resistance (HOMA-IR), triglycerides, high-density lipoprotein cholesterol, apolipoprotein A1, B, and creatinine. All 10-year CVR assessment scores were significantly associated with Hcy. In a multivariate regression model, systolic blood pressure, vitamin supplementation and HOMA-IR were significantly and independently related to Hcy. CONCLUSIONS: Hcy is elevated in HIV-infected patients and is significantly associated with increased CVR. Measurement of Hcy might be useful in identifying particularly high-risk populations at whom therapeutic interventions could be targeted.


Subject(s)
Cardiovascular Diseases/etiology , HIV Infections/complications , HIV-1 , HIV-Associated Lipodystrophy Syndrome/complications , Hyperhomocysteinemia/complications , Metabolic Syndrome/complications , Adult , Anthropometry , Antiretroviral Therapy, Highly Active , Biomarkers , Cardiovascular Diseases/metabolism , Cross-Sectional Studies , Female , HIV Infections/metabolism , HIV-Associated Lipodystrophy Syndrome/metabolism , Homocysteine/blood , Humans , Hyperhomocysteinemia/metabolism , Male , Metabolic Syndrome/metabolism , Predictive Value of Tests , Prospective Studies , Risk Assessment
17.
Eur J Cancer ; 45(3): 443-53, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19008096

ABSTRACT

The p73 locus gene has a complex structure encoding a plethora of isoforms. The different DeltaN truncated isoforms of p73 may exert different activities depending on the cellular context. The beta isoform of DeltaNp73 seems to have a particular pattern of action even if its role in cell cycle and mitosis is still under investigation. To gain further knowledge of DeltaNp73beta's function, we investigated the effects of its over-expression in tumour cellular models, using the tetracycline-inducible expression system. In the human lung carcinoma cell line H1299, DeltaNp73beta over-expression resulted in suppression of cell growth and in cell death. Surprisingly stable over-expression of DeltaNp73beta impaired the genomic stability of tumour cells, leading to the formation of tetraploid cells. The cells become enlarged and multinucleate, with incorrect mitotic figures, and died by apoptotic-independent pathways. Our data suggest that DeltaNp73beta-induced aberrant mitosis evades the control of the mitotic spindle assay checkpoint, leading to tetraploidy and cell death through mitotic catastrophe rather than apoptosis. The various C-terminal regions of DeltaNp73 may influence the final cellular phenotype and we assume that the beta one in particular could be important in both cell growth control and regulation of mitosis.


Subject(s)
Apoptosis/genetics , DNA-Binding Proteins/genetics , Mitosis/genetics , Nuclear Proteins/genetics , Polyploidy , Small Cell Lung Carcinoma/genetics , Tumor Suppressor Proteins/genetics , Apoptosis/physiology , Cell Cycle/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation/genetics , Humans , Mitosis/physiology , Nuclear Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Small Cell Lung Carcinoma/metabolism , Tumor Suppressor Proteins/metabolism
18.
Phys Rev Lett ; 101(12): 126102, 2008 Sep 19.
Article in English | MEDLINE | ID: mdl-18851393

ABSTRACT

The structure of a single layer of graphene on Ru(0001) has been studied using surface x-ray diffraction. A surprising superstructure containing 1250 carbon atoms has been determined, whereby 25 x 25 graphene unit cells lie on 23 x 23 unit cells of Ru. Each supercell contains 2 x 2 crystallographically inequivalent subcells caused by corrugation. Strong intensity oscillations in the superstructure rods demonstrate that the Ru substrate is also significantly corrugated down to several monolayers and that the bonding between graphene and Ru is strong and cannot be caused by van der Waals bonds. Charge transfer from the Ru substrate to the graphene expands and weakens the C-C bonds, which helps accommodate the in-plane tensile stress. The elucidation of this superstructure provides important information in the potential application of graphene as a template for nanocluster arrays.

19.
Phys Chem Chem Phys ; 10(24): 3530-4, 2008 Jun 28.
Article in English | MEDLINE | ID: mdl-18548159

ABSTRACT

Epitaxial graphene on Ru(0001) was studied by means of large-scale density functional theory (DFT) calculations. The results agree well with scanning tunneling microscopy experiments. In contrast to the current understanding, we show that the measured corrugation originates mainly from a geometric buckling of the graphene sheet, induced by alternating weak and strong chemical interactions with the Ru support. In the strong contact regions, charge transfer is evidenced and the opening of a considerable band gap in the graphene is found.

20.
Ann Oncol ; 19(3): 501-7, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17998283

ABSTRACT

BACKGROUND: P63 belongs to the 'p53 family' whose role in cancer progression has been recently revisited in light of the plethora of splicing variants that are generated. We analyzed the expression of the full-length TAp63 gene and its dominant-negative form deltaNp63 in ovarian cancer biopsies to correlate their expression with clinical outcome. MATERIALS AND METHODS: Real-time RT-PCR analysis was used to determine the levels of TAp63 and deltaNp63 in 83 stage I and in 86 stage III ovarian cancer biopsies and in seven human ovarian cancer cell. RESULTS: TAp63 levels were comparable in stage I and stage III, but deltaNp63 levels increased 77-fold in stage III, independently of the p53 status. Patients with high deltaNp63 expression had the worst overall survival (OS); patients with a deltaNp63/TAp63 ratio >2 had a poor OS. Patients with a high deltaNp63/TAp63 ratio were those with a poor response to platinum-based therapy. CONCLUSIONS: Data indicate a role for deltaNp63 as a potential biomarker to predict patient's outcome and tumor progression in ovarian cancer. This would have particularly clinical relevance in ovarian cancer where the high rate of mortality reflects our lack of knowledge of molecular mechanisms underlying cell progression toward malignancy.


Subject(s)
Biomarkers, Tumor/analysis , DNA-Binding Proteins/analysis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/mortality , Trans-Activators/analysis , Tumor Suppressor Proteins/analysis , Biopsy , Disease Progression , Female , Humans , Middle Aged , Mutation , Neoplasm Staging , Ovarian Neoplasms/pathology , Survival Analysis , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...