831 results on '"Regina Elena Cancer Institute"'
Search Results
202. ID4: a new player in the cancer arena.
- Author
-
Dell'Orso S, Ganci F, Strano S, Blandino G, and Fontemaggi G
- Subjects
- Humans, Neoplasms pathology, Drug Resistance, Neoplasm, Inhibitor of Differentiation Proteins physiology, Neoplasms metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Id proteins (Id-1 to 4) are dominant negative regulators of basic helix-loop-helix transcription factors. They play a key role during development, preventing cell differentiation while inducing cell proliferation. They are poorly expressed in adult life but can be reactivated in tumorigenesis. Several evidences indicate that Id proteins are associated with loss of differentiation, unrestricted proliferation and neoangiogenesis in diverse human cancers. Recently, we identified Id4 as a transcriptional target of the protein complex mutant p53/E2F1/p300 in breast cancer. Id4 protein binds, stabilizes and enhances the translation of mRNAs encoding proangiogenic cytokines, such as IL8 and GRO-alpha, increasing the angiogenic potential of cancer cells. We present here an overview of the current experimental data that links Id4 to cancer. We provide evidence also of the induction of Id4 following anticancer treatments in mutant p53- carrying cells. Indeed, mutant p53 is recruited to a specific region of the Id4 promoter upon DNA damage. Our findings indicate that Id4, besides its proangiogenic role, might also participate in the chemoresistance associated to mutant p53 proteins exerting gain of function activities.
- Published
- 2010
- Full Text
- View/download PDF
203. Persistence of HPV after radio-chemotherapy in locally advanced cervical cancer.
- Author
-
Badaracco G, Savarese A, Micheli A, Rizzo C, Paolini F, Carosi M, Cutillo G, Vizza E, Arcangeli G, and Venuti A
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Combined Modality Therapy, DNA, Viral genetics, Female, Humans, Middle Aged, Neoplasm Recurrence, Local virology, Neoplasm Staging, Papillomaviridae isolation & purification, Papillomavirus Infections therapy, Polymerase Chain Reaction, Radiotherapy, Uterine Cervical Neoplasms pathology, Uterine Cervical Neoplasms therapy, Viral Load, DNA, Viral isolation & purification, Papillomaviridae drug effects, Papillomaviridae radiation effects, Papillomavirus Infections virology, Uterine Cervical Neoplasms virology
- Abstract
A causal association of high risk HPV persistent infections with cervical cancer is firmly established by epidemiological and experimental evidence. Since HPV is considered a necessary factor for cervix carcinoma development and disease severity, the HPV DNA persistence may represent an indicator of both therapy effectiveness and risk of recurrence. The presence of HPV in locally advanced cervical carcinoma was analysed at the beginning of therapy, shortly after treatment and during follow-up, in 18 patients with cervix carcinoma treated by radio/chemotherapy. Persistence of HPV DNA sequences was revealed in 62.5% (10/16) of HPV positive patients, in which the HPV type and its physical status were exactly the same as at the onset of therapy, even many years after surgery. Interestingly, in two patients the HPV18 sequence analysis detected the same point mutations in the samples before and after the chemotherapy, and during the follow-up. HPV DNA clearance was associated with a better patient outcome because the majority of the HPV cleared women showed a complete response (6/6), no disease recurrence (4/6), and are still alive. Nevertheless, statistically significant association was seen only with complete responses versus partial or no responses. In conclusion, we demonstrated that HPV DNA positive tumour cells might persist for years in the genital epithelia, even after the surgical removal of the cervix and that HPV DNA detection after therapy is a valid and significant (p=0.03) tool to assess the efficacy of the treatment.
- Published
- 2010
- Full Text
- View/download PDF
204. Synergistic effect of gefitinib and rofecoxib in mesothelioma cells.
- Author
-
Stoppoloni D, Canino C, Cardillo I, Verdina A, Baldi A, Sacchi A, and Galati R
- Subjects
- Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Cyclin-Dependent Kinase Inhibitor p27 metabolism, Cyclooxygenase 2 metabolism, Drug Synergism, ErbB Receptors metabolism, Gefitinib, Humans, Mesothelioma enzymology, Phosphorylation drug effects, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction drug effects, Lactones pharmacology, Mesothelioma pathology, Quinazolines pharmacology, Sulfones pharmacology
- Abstract
Background: Malignant mesothelioma (MM) is an aggressive tumor that is resistant to conventional modes of treatment with chemotherapy, surgery or radiation. Research into the molecular pathways involved in the development of MM should yield information that will guide therapeutic decisions. Epidermal growth factor receptor (EGFR) and cyclooxygenase-2 (COX-2) are involved in the carcinogenesis of MM. Combination of COX-2 and EGFR inhibitors, therefore, could be an effective strategy for reducing cell growth in those lines expressing the two molecular markers., Results: In order to verify the effect of COX-2 and EGFR inhibitors, five MM cell lines NCI-2452, MPP89, Ist-Mes-1, Ist-Mes-2 and MSTO-211 were characterized for COX-2 and EGFR and then treated with respective inhibitors (rofecoxib and gefitinib) alone and in combination. Only MPP89, Ist-Mes-1 and Ist-Mes-2 were sensitive to rofecoxib and showed growth-inhibition upon gefitinib treatment. The combination of two drugs demonstrated synergistic effects on cell killing only in Ist-Mes-2, the cell line that was more sensitive to gefitinib and rofecoxib alone. Down-regulation of COX-2, EGFR, p-EGFR and up-regulation of p21 and p27 were found in Ist-Mes-2, after treatment with single agents and in combination. In contrast, association of two drugs resulted in antagonistic effect in Ist-Mes-1 and MPP89. In these cell lines after rofecoxib exposition, only an evident reduction of p-AKT was observed. No change in p-AKT in Ist-Mes-1 and MPP89 was observed after treatment with gefitinib alone and in combination with rofecoxib., Conclusions: Gefitinib and rofecoxib exert cell type-specific effects that vary between different MM cells. Total EGFR expression and downstream signalling does not correlate with gefitinib sensitivity. These data suggest that the effect of gefitinib can be potentiated by rofecoxib in MM cell lines where AKT is not activated.
- Published
- 2010
- Full Text
- View/download PDF
205. In situ protein expression of RRM1, ERCC1, and BRCA1 in metastatic breast cancer patients treated with gemcitabine-based chemotherapy.
- Author
-
Metro G, Zheng Z, Fabi A, Schell M, Antoniani B, Mottolese M, Monteiro AN, Vici P, Lara Rivera S, Boulware D, Cognetti F, and Bepler G
- Subjects
- Adult, Aged, Biomarkers, Tumor metabolism, Deoxycytidine therapeutic use, Humans, Middle Aged, Neoplasm Metastasis, Ribonucleoside Diphosphate Reductase, Gemcitabine, BRCA1 Protein metabolism, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, DNA-Binding Proteins metabolism, Deoxycytidine analogs & derivatives, Endonucleases metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Ribonucleotide reductase 1 (RRM1) is a determinant of gemcitabine efficacy in non-small-cell lung cancer and pancreatic cancer. We investigated the protein levels of RRM1 and two other DNA repair enzymes, ERCC1 and BRCA1, in 55 metastatic breast cancer (MBC) patients undergoing gemcitabine-based chemotherapy. With automated in situ protein quantification (AQUA v1.6), the average scores for RRM1, ERCC1, and BRCA1 ranged from 245.6-2774.1, 74.0-410.3, and 54.4-1833.1, respectively. They were significantly associated with each other (Spearman's rho > or = .36; p < or = .007). Given their pattern of distribution, RRM1 and BRCA1 are potentially suitable markers for clinical decision making in MBC.
- Published
- 2010
- Full Text
- View/download PDF
206. Clinical significance of PTEN and p-Akt co-expression in HER2-positive metastatic breast cancer patients treated with trastuzumab-based therapies.
- Author
-
Fabi A, Metro G, Di Benedetto A, Nisticò C, Vici P, Melucci E, Antoniani B, Perracchio L, Sperduti I, Milella M, Cognetti F, and Mottolese M
- Subjects
- Adolescent, Aged, Antibodies, Monoclonal, Humanized, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Disease Progression, Female, Gene Expression Regulation, Neoplastic, Humans, Immunohistochemistry, Middle Aged, Neoplasm Metastasis pathology, PTEN Phosphohydrolase metabolism, Proto-Oncogene Proteins c-akt metabolism, Receptor, ErbB-2 metabolism, Trastuzumab, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Breast Neoplasms genetics, PTEN Phosphohydrolase genetics, Proto-Oncogene Proteins c-akt genetics, Receptor, ErbB-2 genetics
- Abstract
Objective: The phosphatase and tensine homologue gene (PTEN) plays a crucial role in proliferation and survival of cancer cells by antagonizing the function of phosphatidylinositol 3'-kinase (PI3K), which, in turn, results in decreased Akt activity. We investigated the clinical impact of the expression of PTEN, p-Akt and PI3K in HER2-positive metastatic breast cancer (MBC) patients treated with trastuzumab-based therapies., Methods: Seventy-three patients treated with trastuzumab-based therapies were included and followed prospectively. PTEN, p-Akt and PI3K expression was determined by immunohistochemistry., Results: PTEN, p-Akt and PI3K resulted positive in 48%, 71% and 46.5% of patients, respectively. A significant correlation between PTEN and p-Akt (kappa 0.22, p = 0.03) and p-Akt and PI3K (kappa 0.20, p = 0.05) was observed. PTEN-positive patients had a progression-free survival (PFS) longer than PTEN-negative ones (p = 0.06). When grouped together, patients co-expressing PTEN and p-Akt had a statistically significant longer PFS as compared to the rest of patients (p = 0.01). At the multivariate analysis, PTEN and p-Akt co-expression was an independent predictor of lower risk of progression (hazard ratio 0.53, p = 0.05)., Conclusion: In HER2-positive MBC, basal co-expression of PTEN and p-Akt might identify those patients who are more likely to benefit from trastuzumab-based therapies., (Copyright 2010 S. Karger AG, Basel.)
- Published
- 2010
- Full Text
- View/download PDF
207. Transgenic mice expressing an artificial zinc finger regulator targeting an endogenous gene.
- Author
-
Passananti C, Corbi N, Onori A, Di Certo MG, and Mattei E
- Subjects
- Animals, Blotting, Western, Chromatin Immunoprecipitation, Electrophoresis, Polyacrylamide Gel, Female, Genotype, Humans, Immunoblotting, Male, Mice, Mice, Transgenic, Transcription Factors genetics, Utrophin genetics, Genetic Therapy methods, Muscular Dystrophy, Duchenne genetics, Muscular Dystrophy, Duchenne therapy, Transcription Factors metabolism, Zinc Fingers genetics
- Abstract
Zinc finger (ZF) proteins belonging to the Cys2-His2 class provide a simple and versatile framework to design novel artificial transcription factors (ATFs) targeted to the desired genes. Our work is based on ZF ATFs engineered to up-regulate the expression level of the dystrophin-related gene utrophin in Duchenne muscular dystrophy (DMD). In particular, on the basis of the "recognition code" that defines specific rules between zinc finger primary structure and potential DNA-binding sites we engineered and selected a new family of artificial transcription factors, whose DNA-binding domain consists in a three zinc finger peptide called "Jazz." Jazz protein binds specifically the 9 bp DNA sequence (5(')-GCT-GCT-GCG-3(')) present in the promoter region of both the human and mouse utrophin gene. We generated a transgenic mouse expressing Jazz protein fused to the strong transcriptional activation domain VP16 and under the control of the muscle specific promoter of the myosin light chain gene. Vp16-Jazz mice display a strong up-regulation of the utrophin at both mRNA and protein levels. To our knowledge, this represents the first example of a transgenic mouse expressing an artificial gene coding for a zinc finger-based transcription factor.
- Published
- 2010
- Full Text
- View/download PDF
208. Phase II study of fixed dose rate gemcitabine as radiosensitizer for newly diagnosed glioblastoma multiforme.
- Author
-
Metro G, Fabi A, Mirri MA, Vidiri A, Pace A, Carosi M, Russillo M, Maschio M, Giannarelli D, Pellegrini D, Pompili A, Cognetti F, and Carapella CM
- Subjects
- Adult, Aged, Brain Neoplasms pathology, Combined Modality Therapy, DNA Modification Methylases genetics, DNA Modification Methylases metabolism, DNA Repair Enzymes genetics, DNA Repair Enzymes metabolism, Deoxycytidine therapeutic use, Female, Glioblastoma pathology, Humans, Magnetic Resonance Imaging, Male, Methylation, Middle Aged, Promoter Regions, Genetic, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Gemcitabine, Antineoplastic Agents therapeutic use, Brain Neoplasms drug therapy, Brain Neoplasms radiotherapy, Deoxycytidine analogs & derivatives, Glioblastoma drug therapy, Glioblastoma radiotherapy, Radiation-Sensitizing Agents therapeutic use
- Abstract
Purpose: In order to evaluate the activity of gemcitabine as radiosensitizer for newly diagnosed glioblastoma multiforme (GBM), a prospective single-center phase II study was conducted., Methods: Eligible patients were required to have histologically proven GBM with evaluable and/or measurable disease after surgery. They were treated by standard cranial irradiation plus concomitant fixed dose rate gemcitabine given intravenously at 175 mg/m(2) weekly for 6 weeks. After chemo-radiotherapy, irrespective of tumor response, patients went on to receive oral temozolomide at 150-200 mg/m(2) for 5 days every 28 days., Results: Twenty-three patients were enrolled. Median age was 57 years (range 43-72) and median Karnofsky performance status was 90 (range 70-100). Seventeen patients had received subtotal resection of the tumor, while six patients had biopsied-only tumors. Four patients responded to treatment (17.5%) with additional 14 (61%) experiencing stable disease for an overall disease control rate of 78.5%. Median progression-free and overall survival were 6.8 and 10.1 months, respectively. The concomitant radiotherapy-gemcitabine combination was well tolerated and severe adverse events were rare, consisting of grade 3 neutropenia and hypertransaminasemia in two cases each. Twenty patients were assessable for methylguanine methyltransferase (MGMT) promoter methylation, 11 of which were found methylated. In the methylated and unmethylated cohorts, disease control was obtained in 10/11 patients (91%) and 7/9 patients (77.5%), respectively., Conclusions: Concomitant radiotherapy-gemcitabine is active and well tolerated in newly diagnosed glioblastoma multiforme. Activity is observed both in tumors with methylated and unmethylated MGMT promoter.
- Published
- 2010
- Full Text
- View/download PDF
209. Time to first tumor progression as outcome predictor of a second trasuzumab-based therapy beyond progression in HER-2 positive metastatic breast cancer.
- Author
-
Metro G, Giannarelli D, Gemma D, Lanzetta G, Ciccarese M, Papaldo P, Gamucci T, Lorusso V, Mottolese M, Magnolfi E, Cognetti F, and Fabi A
- Subjects
- Adult, Aged, Antibodies, Monoclonal, Humanized, Biomarkers, Tumor analysis, Biomarkers, Tumor metabolism, Breast Neoplasms mortality, Breast Neoplasms secondary, Cohort Studies, Disease Progression, Dose-Response Relationship, Drug, Drug Administration Schedule, Female, Follow-Up Studies, Humans, Kaplan-Meier Estimate, Middle Aged, Neoplasm Invasiveness pathology, Neoplasm Metastasis, Neoplasm Staging, Predictive Value of Tests, Probability, Proportional Hazards Models, Receptor, ErbB-2 genetics, Remission Induction, Risk Assessment, Statistics, Nonparametric, Survival Analysis, Time Factors, Trastuzumab, Treatment Outcome, Antibodies, Monoclonal administration & dosage, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Receptor, ErbB-2 metabolism
- Abstract
In a previous analysis performed on a cohort of 37 HER-2 positive metastatic breast cancer (MBC) patients treated with trastuzumab beyond progression, we found that a second trastuzumab-based therapy is associated with a considerable response rate and preserved time to progression as compared with a first trastuzumab-based therapy. In the present study, we extended the analysis to a total of 69 patients treated in four different italian Institutions, also trying to identify clinical predictors of sensitivity to a second trastuzumab-based therapy beyond progression. Efficacy results on the overall population confirmed that a second trastuzumab-based therapy beyond progression is an active regimen (27.5% of responses and 6.5 months of time to progression, respectively). Median time to progression to the first trastuzumab therapy (TTP1) identified two groups of patients with different sensitivity to trastuzumab beyond progression (group A, TTP1 >or= 8 months and group B, TTP1 < 8 months) in terms of time to second progression and post-progression survival (group A versus group B showed respectively a time to second progression of 7.6 versus 4.7 months, p = 0.05, and a post-progression survival of 31.7 months versus 21.8 months, p = 0.04). In the multivariate analysis, only TTP1 was a predictor of time to second progression and post-progression survival. Despite the recent approval of lapatinib plus capecitabine for trastuzumab-progressing patients, it is still reasonable to offer trastuzumab beyond progression to HER-2 positive MBC patients, because these data confirm the potential utility of such a conduct. In the clinic, time to first tumor progression may represent a useful tool to identify patients who are more likely to benefit from trastuzumab beyond progression.
- Published
- 2010
- Full Text
- View/download PDF
210. Intracellular presence of insulin and its phosphorylated receptor in non-small cell lung cancer.
- Author
-
Mattarocci S, Abbruzzese C, Mileo AM, Visca P, Antoniani B, Alessandrini G, Facciolo F, Felsani A, Radulescu RT, and Paggi MG
- Subjects
- Adenocarcinoma metabolism, Aged, Aging metabolism, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Squamous Cell metabolism, Cell Membrane metabolism, Cytoplasm metabolism, Cytoplasmic Granules metabolism, Disease-Free Survival, Female, Gene Expression genetics, Humans, Insulin genetics, Kaplan-Meier Estimate, Male, Middle Aged, Phosphorylation, Antigens, CD metabolism, Carcinoma, Non-Small-Cell Lung metabolism, Insulin metabolism, Intracellular Space metabolism, Receptor, Insulin metabolism
- Abstract
Insulin has been known for a long time to influence the growth and differentiation of normal and transformed cells. In order to delineate the role of insulin specifically in non-small cell lung cancer (NSCLC), we have now searched by immunohistochemistry (IHC) for the presence of insulin in NSCLC samples. Among the 112 samples we studied, 30 were found to contain insulin, which was detected in the form of intracytoplasmic granula. Moreover, its expression significantly correlated with (a) the morphological/histopathological subtype of NSCLC, being more frequent in adenocarcinomas; (b) the grade of tumor differentiation, displaying an increase in low-grade carcinomas; (c) tumor size, occurring predominantly in smaller tumors; (d) the presence of phosphorylated, activated insulin receptor; (e) the median patient age, being present in relatively younger individuals. Furthermore and interestingly, surrounding atypical adenomatous hyperplastic areas and normal alveolar pneumocytes scored insulin-positive in some of the insulin-negative tumors. In addition, PCR exploration for insulin transcripts in some samples positive for immunoreactive insulin was negative, indicating a possibly exogenous origin for the intracellular insulin in our NSCLC cohort. Taken together, our data suggest that an intracellular insulin activity is important for the progression of low-grade human lung adenocarcinomas.
- Published
- 2009
- Full Text
- View/download PDF
211. Methyl-CpG-binding protein 2 is phosphorylated by homeodomain-interacting protein kinase 2 and contributes to apoptosis.
- Author
-
Bracaglia G, Conca B, Bergo A, Rusconi L, Zhou Z, Greenberg ME, Landsberger N, Soddu S, and Kilstrup-Nielsen C
- Subjects
- Amino Acid Substitution physiology, Animals, Apoptosis drug effects, Carrier Proteins antagonists & inhibitors, Cells, Cultured, DNA metabolism, Embryo, Mammalian, HeLa Cells, Humans, Methyl-CpG-Binding Protein 2 genetics, Mice, Mutant Proteins genetics, Mutant Proteins metabolism, NIH 3T3 Cells, Phosphorylation drug effects, Phosphorylation genetics, Protein Binding drug effects, Protein Binding genetics, Protein Serine-Threonine Kinases antagonists & inhibitors, RNA, Small Interfering pharmacology, Serine genetics, Serine metabolism, Apoptosis genetics, Carrier Proteins metabolism, Methyl-CpG-Binding Protein 2 metabolism, Methyl-CpG-Binding Protein 2 physiology, Protein Serine-Threonine Kinases metabolism
- Abstract
Mutations in the methyl-CpG-binding protein 2 (MeCP2) are associated with Rett syndrome and other neurological disorders. MeCP2 represses transcription mainly by recruiting various co-repressor complexes. Recently, MeCP2 phosphorylation at Ser 80, Ser 229 and Ser 421 was shown to occur in the brain and modulate MeCP2 silencing activities. However, the kinases directly responsible for this are largely unknown. Here, we identify the homeodomain-interacting protein kinase 2 (HIPK2) as a kinase that binds MeCP2 and phosphorylates it at Ser 80 in vitro and in vivo. HIPK2 modulates cell proliferation and apoptosis, and the neurological defects of Hipk2-null mice indicate its role in proper brain functions. We show that MeCP2 cooperates with HIPK2 in induction of apoptosis and that Ser 80 phosphorylation is required together with the DNA binding of MeCP2. These data are, to our knowledge, the first that describe a kinase associating with MeCP2, causing its specific phosphorylation in vivo and, furthermore, they reinforce the role of MeCP2 in regulating cell growth.
- Published
- 2009
- Full Text
- View/download PDF
212. A restricted signature of miRNAs distinguishes APL blasts from normal promyelocytes.
- Author
-
Careccia S, Mainardi S, Pelosi A, Gurtner A, Diverio D, Riccioni R, Testa U, Pelosi E, Piaggio G, Sacchi A, Lavorgna S, Lo-Coco F, Blandino G, Levrero M, and Rizzo MG
- Subjects
- Granulocyte Precursor Cells pathology, Humans, Leukemia, Promyelocytic, Acute metabolism, Leukemia, Promyelocytic, Acute pathology, MicroRNAs genetics, Leukemia, Promyelocytic, Acute genetics, MicroRNAs analysis
- Abstract
MicroRNAs (miRNAs) are small non-coding RNAs involved in the regulation of critical cell processes such as apoptosis, cell proliferation and differentiation. A small set of miRNAs is differentially expressed in hematopoietic cells and seemingly has an important role in granulopoiesis and lineage differentiation. In this study, we analysed, using a quantitative real-time PCR approach, the expression of 12 granulocytic differentiation signature miRNAs in a cohort of acute promyelocytic leukemia (APL) patients. We found nine miRNAs overexpressed and three miRNAs (miR-107, -342 and let-7c) downregulated in APL blasts as compared with normal promyelocytes differentiated in vitro from CD34+ progenitors. Patients successfully treated with all-trans-retinoic acid (ATRA) and chemotherapy showed downregulation of miR-181b and upregulation of miR-15b, -16, -107, -223, -342 and let-7c. We further investigated whether the APL-associated oncogene, promyelocytic leukemia gene (PML)/retinoic acid receptor alpha (RARalpha), might be involved in the transcriptional repression of miR-107, -342 and let-7c. We found that PML/RARalpha binds the regulatory sequences of the intragenic miR-342 and let-7c. In addition, we observed, in response to ATRA, the release of PML/RARalpha paralleled by their transcriptional activation, together with their host genes, EVL and C21orf34alpha. In conclusion, we show that a small subset of miRNAs is differentially expressed in APL and modulated by ATRA-based treatment.
- Published
- 2009
- Full Text
- View/download PDF
213. The Hippo tumor suppressor pathway: a brainstorming workshop.
- Author
-
Blandino G, Shaul Y, Strano S, Sudol M, and Yaffe M
- Subjects
- Education, Italy, Neoplasms, Organ Size, Signal Transduction, Congresses as Topic, Tumor Suppressor Proteins metabolism
- Abstract
Researchers from around the world met for two days in April this year in Rome, Italy, to discuss progress in the rapidly developing field of Hippo signaling, which is relevant to cancer and the control of organ size. Most of the participants presented data related to previously uncharacterized proteins that physically and functionally interact with known components of the Hippo pathway and regulate its biological output.
- Published
- 2009
- Full Text
- View/download PDF
214. Liposarcoma of the colon presenting as an endoluminal mass.
- Author
-
D'Annibale M, Cosimelli M, Covello R, and Stasi E
- Subjects
- Abdominal Pain etiology, Colonic Neoplasms complications, Colonic Neoplasms diagnosis, Colonic Neoplasms surgery, Colonoscopy, Diagnosis, Differential, Fatal Outcome, Female, Humans, Liposarcoma complications, Liposarcoma diagnosis, Liposarcoma surgery, Prognosis, Radiotherapy, Adjuvant, Tomography, X-Ray Computed, Colonic Neoplasms pathology, Liposarcoma pathology
- Abstract
Background: Liposarcoma is one of the most common soft tissue sarcoma of adult life, usually occurring in the retroperitoneum and the extremities. Primary liposarcoma of the colon is very rare. The optimal treatment has not been established due to the small number of cases reported. We report a case of primary liposarcoma of the colon presenting as a massive intraluminal lesion., Case Presentation: A 79-year-old woman presented with abdominal pain, progressive constipation and weight loss. A CT scan and a colonoscopy revealed an intraluminal mass in the transverse colon and multiple intraperitoneal lesions. The patient underwent surgical resection of the lesions. Pathologic examination was consistent with pleomorphic liposarcoma of the colon., Conclusion: Although no guidelines are available for the management of liposarcoma of the colon, surgical resection should be performed when feasible. Our patient's overall survival was satisfactory in spite of the multiple negative prognostic factors.
- Published
- 2009
- Full Text
- View/download PDF
215. Human papillomavirus-16 E7 interacts with glutathione S-transferase P1 and enhances its role in cell survival.
- Author
-
Mileo AM, Abbruzzese C, Mattarocci S, Bellacchio E, Pisano P, Federico A, Maresca V, Picardo M, Giorgi A, Maras B, Schininà ME, and Paggi MG
- Subjects
- Cell Line, Tumor, Cell Survival, Cell Transformation, Neoplastic, Gene Expression Regulation, Viral, Humans, Keratinocytes virology, Mass Spectrometry methods, Molecular Conformation, Mutation, Papillomavirus E7 Proteins, Protein Binding, Protein Interaction Mapping, Repressor Proteins metabolism, Glutathione S-Transferase pi metabolism, Human papillomavirus 16 metabolism, Oncogene Proteins, Viral metabolism
- Abstract
Background: Human Papillomavirus (HPV)-16 is a paradigm for "high-risk" HPVs, the causative agents of virtually all cervical carcinomas. HPV E6 and E7 viral genes are usually expressed in these tumors, suggesting key roles for their gene products, the E6 and E7 oncoproteins, in inducing malignant transformation., Methodology/principal Findings: By protein-protein interaction analysis, using mass spectrometry, we identified glutathione S-transferase P1-1 (GSTP1) as a novel cellular partner of the HPV-16 E7 oncoprotein. Following mapping of the region in the HPV-16 E7 sequence that is involved in the interaction, we generated a three-dimensional molecular model of the complex between HPV-16 E7 and GSTP1, and used this to engineer a mutant molecule of HPV-16 E7 with strongly reduced affinity for GSTP1.When expressed in HaCaT human keratinocytes, HPV-16 E7 modified the equilibrium between the oxidized and reduced forms of GSTP1, thereby inhibiting JNK phosphorylation and its ability to induce apoptosis. Using GSTP1-deficient MCF-7 cancer cells and siRNA interference targeting GSTP1 in HaCaT keratinocytes expressing either wild-type or mutant HPV-16 E7, we uncovered a pivotal role for GSTP1 in the pro-survival program elicited by its binding with HPV-16 E7., Conclusions/significance: This study provides further evidence of the transforming abilities of this oncoprotein, setting the groundwork for devising unique molecular tools that can both interfere with the interaction between HPV-16 E7 and GSTP1 and minimize the survival of HPV-16 E7-expressing cancer cells.
- Published
- 2009
- Full Text
- View/download PDF
216. The execution of the transcriptional axis mutant p53, E2F1 and ID4 promotes tumor neo-angiogenesis.
- Author
-
Fontemaggi G, Dell'Orso S, Trisciuoglio D, Shay T, Melucci E, Fazi F, Terrenato I, Mottolese M, Muti P, Domany E, Del Bufalo D, Strano S, and Blandino G
- Subjects
- Breast Neoplasms metabolism, Cell Line, Tumor, Cell Proliferation, Cytokines metabolism, E2F1 Transcription Factor metabolism, Humans, Inhibitor of Differentiation Proteins metabolism, Interleukin-8 metabolism, Microcirculation, Tumor Suppressor Protein p53 metabolism, Vascular Endothelial Growth Factor A metabolism, E2F1 Transcription Factor genetics, Gene Expression Regulation, Neoplastic, Inhibitor of Differentiation Proteins genetics, Mutation, Neovascularization, Pathologic, Transcription, Genetic, Tumor Suppressor Protein p53 genetics
- Abstract
ID4 (inhibitor of DNA binding 4) is a member of a family of proteins that function as dominant-negative regulators of basic helix-loop-helix transcription factors. Growing evidence links ID proteins to cell proliferation, differentiation and tumorigenesis. Here we identify ID4 as a transcriptional target of gain-of-function p53 mutants R175H, R273H and R280K. Depletion of mutant p53 protein severely impairs ID4 expression in proliferating tumor cells. The protein complex mutant p53-E2F1 assembles on specific regions of the ID4 promoter and positively controls ID4 expression. The ID4 protein binds to and stabilizes mRNAs encoding pro-angiogenic factors IL8 and GRO-alpha. This results in the increase of the angiogenic potential of cancer cells expressing mutant p53. These findings highlight the transcriptional axis mutant p53, E2F1 and ID4 as a still undefined molecular mechanism contributing to tumor neo-angiogenesis.
- Published
- 2009
- Full Text
- View/download PDF
217. Sexual dysfunction following surgery for rectal cancer - a clinical and neurophysiological study.
- Author
-
Pietrangeli A, Pugliese P, Perrone M, Sperduti I, Cosimelli M, and Jandolo B
- Subjects
- Adult, Aged, Antineoplastic Agents adverse effects, Combined Modality Therapy, Evoked Potentials, Motor, Evoked Potentials, Somatosensory, Female, Genitalia innervation, Humans, Male, Middle Aged, Postoperative Complications epidemiology, Postoperative Complications physiopathology, Radiotherapy adverse effects, Reflex, Sexual Dysfunction, Physiological epidemiology, Sexual Dysfunction, Physiological physiopathology, Adenocarcinoma surgery, Digestive System Surgical Procedures adverse effects, Electrodiagnosis methods, Postoperative Complications diagnosis, Rectal Neoplasms surgery, Sexual Dysfunction, Physiological diagnosis
- Abstract
Background: Sexual dysfunction following surgery for rectal cancer may be frequent and often severe. The aim of the present study is to evaluate the occurrence of this complication from both a clinical point of view and by means of neurophysiological tests., Methods: We studied a group of 57 patients submitted to rectal resection for adenocarcinoma. All the patients underwent neurological, psychological and the following neurophysiological tests: sacral reflex (SR), pudendal somatosensory evoked potentials (PEPs), motor evoked potential (MEPs) and sympathetic skin responses (SSRs). The results were compared with a control group of 67 rectal cancer patients studied before surgery. Only 10 of these patients could be studied both pre- and postoperatively. 10 patients submitted to high dose preoperative chemoradiation were studied to evaluate the effect of this treatment on sexual function. Statistical analysis was performed by means of the two-tailed Student's t test for paired observations and k concordance test., Results: 59.6% of patients operated reported sexual dysfunction, while this symptom occurred in 16.4% in the control group. Moreover, a significantly higher rate of alterations of the neurophysiological tests and longer mean latencies of the SR, PEPs, MEPs and SSRs were observed in the patients who had undergone resection. In the 10 patients studied both pre and post-surgery impotence occurred in 6 of them and the mean latencies of SSRs were longer after operation. In the 10 patients studied pre and post chemoradiation impotence occurred in 1 patient only, showing the mild effect of these treatments on sexual function., Conclusion: Patients operated showed severe sexual dysfunctions. The neurophysiological test may be a useful tool to investigate this complication. The neurological damage could be monitored to decide the rehabilitation strategy.
- Published
- 2009
- Full Text
- View/download PDF
218. Genetic profile identification in clinically localized prostate carcinoma.
- Author
-
Gallucci M, Merola R, Leonardo C, De Carli P, Farsetti A, Sentinelli S, Sperduti I, Mottolese M, Carlini P, Vico E, Simone G, and Cianciulli A
- Subjects
- Chromosome Aberrations, Chromosomes, Human, Pair 8 genetics, Chromosomes, Human, X genetics, Disease Progression, Gene Dosage, Humans, In Situ Hybridization, Fluorescence, Lipoprotein Lipase genetics, Male, Neoplasm Staging, Prognosis, Prostate-Specific Antigen blood, Proto-Oncogene Proteins c-myc genetics, Retrospective Studies, Biomarkers, Tumor genetics, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology
- Abstract
Purpose: To confirm our previously obtained results, we genetically characterized prostate cancer from patients undergo radical prostatectomy in a retrospective study., Materials and Methods: Histological sections were evaluated for 106 patients treated with surgery from 1991 to 2004. With fluorescence in situ hybridization (FISH) method, the status of LPL (8p22), c-MYC (8q24) genes and 7, 8, X chromosomes was evaluated., Results: Chromosomes 7, 8, X aneusomy was demonstrated in 91.5%, 78.3%, and 51.9% of the samples, respectively, whereas LPL deletion and MYC amplification were found in 76.0% and 1.6%. A genetic profile was considered as unfavorable when at least two aneusomic chromosomes and one altered gene were present. Tumors with an adverse genetic profile were more frequently present in patients with higher stages (P = 0.02), biochemical/clinical progression (P = 0.03), and Gleason grade 4 + 3 (P = 0.02). Multiple correspondence analysis identified one tumor group characterized by chromosome 8 aneusomy, X polysomy, LPL gene deletion, Gleason > 7 and 4 + 3 associated with progression., Conclusions: In this study, we recognized the predictive power of previously identified cytogenetic profiles. Assessment of genetic set may characterize each patient and have influence on postoperative therapeutic strategies.
- Published
- 2009
- Full Text
- View/download PDF
219. Stabilization of quadruplex DNA perturbs telomere replication leading to the activation of an ATR-dependent ATM signaling pathway.
- Author
-
Rizzo A, Salvati E, Porru M, D'Angelo C, Stevens MF, D'Incalci M, Leonetti C, Gilson E, Zupi G, and Biroccio A
- Subjects
- Ataxia Telangiectasia Mutated Proteins, Cell Line, DNA Damage, DNA Replication drug effects, Humans, Ligands, Signal Transduction, Telomere chemistry, Telomere metabolism, Acridines pharmacology, Cell Cycle Proteins metabolism, DNA-Binding Proteins metabolism, G-Quadruplexes drug effects, Protein Serine-Threonine Kinases metabolism, Telomere drug effects, Tumor Suppressor Proteins metabolism
- Abstract
Functional telomeres are required to maintain the replicative ability of cancer cells and represent putative targets for G-quadruplex (G4) ligands. Here, we show that the pentacyclic acridinium salt RHPS4, one of the most effective and selective G4 ligands, triggers damages in cells traversing S phase by interfering with telomere replication. Indeed, we found that RHPS4 markedly reduced BrdU incorporation at telomeres and altered the dynamic association of the telomeric proteins TRF1, TRF2 and POT1, leading to chromosome aberrations such as telomere fusions and telomere doublets. Analysis of the molecular damage pathway revealed that RHPS4 induced an ATR-dependent ATM signaling that plays a functional role in the cellular response to RHPS4 treatment. We propose that RHPS4, by stabilizing G4 DNA at telomeres, impairs fork progression and/or telomere processing resulting in telomere dysfunction and activation of a replication stress response pathway. The detailed understanding of the molecular mode of action of this class of compounds makes them attractive tools to understand telomere biology and provides the basis for a rational use of G4 ligands for the therapy of cancer.
- Published
- 2009
- Full Text
- View/download PDF
220. Functional activity of CXCL8 receptors, CXCR1 and CXCR2, on human malignant melanoma progression.
- Author
-
Gabellini C, Trisciuoglio D, Desideri M, Candiloro A, Ragazzoni Y, Orlandi A, Zupi G, and Del Bufalo D
- Subjects
- Animals, Cell Line, Tumor, Cell Movement, Cell Proliferation, Humans, Melanoma blood supply, Mice, Mice, Nude, Neoplasm Invasiveness, Neovascularization, Pathologic etiology, Interleukin-8 metabolism, Melanoma metabolism, Neoplasm Proteins metabolism, Receptors, Interleukin-8A metabolism, Receptors, Interleukin-8B metabolism
- Abstract
We examined the autocrine/paracrine role of interleukin-8 (CXCL8) and the functional significance of CXCL8 receptors, CXCR1 and CXCR2, in human malignant melanoma proliferation, migration, invasion and angiogenesis. We found that a panel of seven cell lines, even though at different extent, secreted CXCL8 protein, and expressed CXCR1 and CXCR2 independently from the CXCL8 expression, but depending on the oxygen level. In fact, hypoxic exposure increases the expression of CXCR1 and CXCR2. The cell proliferation of both M20 and A375SM lines, expressing similar levels of both CXCR1 and CXCR2 but secreting low and high amounts of CXCL8, respectively, was significantly enhanced by CXCL8 exposure and reduced by CXCL8, CXCR1 and CXCR2 neutralising antibodies, indicating the autocrine/paracrine role of CXCL8 in melanoma cell proliferation. Moreover, an increased invasion and migration in response to CXCL8 was observed in several cell lines, and a further enhancement evidenced under hypoxic conditions. A CXCL8-dependent in vivo vessel formation, evaluated through a matrigel assay, was also demonstrated. Furthermore, when neutralising antibodies against CXCR1 or CXCR2 were used, only the involvement of CXCR2, but not CXCR1 was observed on cell migration and invasion, while both receptors played a role in angiogenesis. In summary, our data demonstrate that CXCL8 induces cell proliferation and angiogenesis through both receptors and that CXCR2 plays an important role in regulating the CXCL8-mediated invasive and migratory behaviour of human melanoma cells. Thus, blocking the CXCL8 signalling axis promises an improvement for the therapy of cancer and, in particular, of metastatic melanoma.
- Published
- 2009
- Full Text
- View/download PDF
221. Adjuvant electrochemotherapy in veterinary patients: a model for the planning of future therapies in humans.
- Author
-
Spugnini EP, Citro G, and Baldi A
- Subjects
- Animals, Chemotherapy, Adjuvant methods, Disease Models, Animal, Electrochemotherapy methods, Humans, Neoplasms drug therapy, Neoplasms radiotherapy, Radiotherapy, Adjuvant methods, Electrochemotherapy veterinary, Neoplasms veterinary
- Abstract
The treatment of soft tissue tumors needs the coordinated adoption of surgery with radiation therapy and eventually, chemotherapy. The radiation therapy (delivered with a linear accelerator) can be preoperative, intraoperative, or postoperative. In selected patients adjuvant brachytherapy can be adopted. The goal of these associations is to achieve tumor control while maximally preserving the normal tissues from side effects. Unfortunately, the occurrence of local and distant complications is still elevated. Electrochemotherapy is a novel technique that combines the administration of anticancer agents to the application of permeabilizing pulses in order to increase the uptake of antitumor molecules. While its use in humans is still confined to the treatment of cutaneous neoplasms or the palliation of skin tumor metastases, in veterinary oncology this approach is rapidly becoming a primary treatment. This review summarizes the recent progresses in preclinical oncology and their possible transfer to humans.
- Published
- 2009
- Full Text
- View/download PDF
222. HIPK2 regulation by MDM2 determines tumor cell response to the p53-reactivating drugs nutlin-3 and RITA.
- Author
-
Rinaldo C, Prodosmo A, Siepi F, Moncada A, Sacchi A, Selivanova G, and Soddu S
- Subjects
- Apoptosis drug effects, Apoptosis physiology, Carrier Proteins genetics, Carrier Proteins metabolism, Cell Line, Tumor, HCT116 Cells, Humans, Mitosis drug effects, Protein Serine-Threonine Kinases deficiency, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-mdm2 antagonists & inhibitors, RNA, Messenger biosynthesis, RNA, Messenger genetics, Up-Regulation drug effects, Carrier Proteins biosynthesis, Furans pharmacology, Imidazoles pharmacology, Piperazines pharmacology, Protein Serine-Threonine Kinases biosynthesis, Proto-Oncogene Proteins c-mdm2 metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
In the past few years, much effort has been devoted to show the single-target specificity of nongenotoxic, p53 reactivating compounds. However, the divergent biological responses induced by the different compounds, even in the same tumor cells, demand additional mechanistic insights, whose knowledge may lead to improved drug design or selection of the most potent drug combinations. To address the molecular mechanism underlying induction of mitotic arrest versus clinically more desirable apoptosis, we took advantage of two MDM2 antagonists, Nutlin-3 and RITA, which respectively produce these two outcomes. We show that, along with p53 reactivation, the proapoptotic p53-activator HIPK2 is degraded by MDM2 in Nutlin-3-treated cells, but activated by transiently reduced MDM2 levels in RITA-treated ones. Gain- and loss-of-function experiments revealed the functional significance of MDM2-mediated HIPK2 regulation in cell decision between mitotic arrest and apoptosis in both types of p53 reactivation. These data indicate that strategies of p53 reactivation by MDM2 inhibition should also take into consideration MDM2 targets other than p53, such as the apoptosis activator HIPK2.
- Published
- 2009
- Full Text
- View/download PDF
223. Negative regulation of beta4 integrin transcription by homeodomain-interacting protein kinase 2 and p53 impairs tumor progression.
- Author
-
Bon G, Di Carlo SE, Folgiero V, Avetrani P, Lazzari C, D'Orazi G, Brizzi MF, Sacchi A, Soddu S, Blandino G, Mottolese M, and Falcioni R
- Subjects
- Blotting, Western, Breast Neoplasms genetics, Breast Neoplasms metabolism, Breast Neoplasms pathology, Carrier Proteins genetics, Cell Line, Tumor, Cell Proliferation, Cytoplasm metabolism, Disease Progression, Female, Gene Expression Regulation, Neoplastic, HCT116 Cells, HT29 Cells, Histone Acetyltransferases metabolism, Humans, Immunohistochemistry, Integrin beta4 genetics, Mitogen-Activated Protein Kinases metabolism, Mutation, Neoplasms genetics, Neoplasms metabolism, Phosphorylation, Protein Serine-Threonine Kinases genetics, Proto-Oncogene Proteins c-akt metabolism, Reverse Transcriptase Polymerase Chain Reaction, Transcription, Genetic, Tumor Suppressor Protein p53 genetics, p300-CBP Transcription Factors metabolism, Carrier Proteins metabolism, Integrin beta4 metabolism, Neoplasms pathology, Protein Serine-Threonine Kinases metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
Increased expression of alpha(6)beta(4) integrin in several epithelial cancers promotes tumor progression; however, the mechanism underlying its transcriptional regulation remains unclear. Here, we show that depletion of homeodomain-interacting protein kinase 2 (HIPK2) activates beta(4) transcription that results in a strong increase of beta(4)-dependent mitogen-activated protein kinase and Akt phosphorylation, anchorage-independent growth, and invasion. In contrast, stabilization of HIPK2 represses beta(4) expression in wild-type p53 (wtp53)-expressing cells but not in p53-null cells or cells expressing mutant p53, indicating that HIPK2 requires a wtp53 to inhibit beta(4) transcription. Consistent with our in vitro findings, a strong correlation between beta(4) overexpression and HIPK2 inactivation by cytoplasmic relocalization was observed in wtp53-expressing human breast carcinomas. Under loss of function of HIPK2 or p53, the p53 family members TAp63 and TAp73 strongly activate beta(4) transcription. These data, by revealing that beta(4) expression is transcriptionally repressed in tumors by HIPK2 and p53 to impair beta(4)-dependent tumor progression, suggest that loss of p53 function favors the formation of coactivator complex with the TA members of the p53 family to allow beta(4) transcription.
- Published
- 2009
- Full Text
- View/download PDF
224. Solitary true cyst of the pancreas in adults. A report of two cases.
- Author
-
Carboni F, Mancini P, Lorusso R, and Santoro E
- Subjects
- Female, Humans, Middle Aged, Pancreas surgery, Pancreatectomy methods, Pancreatic Cyst surgery, Treatment Outcome, Young Adult, Pancreas pathology, Pancreatic Cyst diagnosis
- Abstract
Context: Solitary true cysts of the pancreas in adults are extremely rare and only few cases have been reported in the literature. The etiology and natural history of these lesions remain unknown and treatment is not standardized. We describe two additional resected cases., Case Reports: The first patient was a young woman with an incidental 3 cm cyst located in the pancreatic head who underwent enucleation. The second patient was a young woman with a large 8 cm symptomatic cyst located in the pancreatic tail who underwent a laparoscopic spleen-preserving distal pancreatectomy. Histological examination revealed fibrous walls lined by a monolayer of cuboidal epithelium in both cases., Conclusions: A preoperative work-up alone does not always allow an accurate diagnosis, but it is useful in determining lesion characteristics and guiding therapeutic decision making. When surgery is indicated, a limited resection is warranted in most cases.
- Published
- 2009
225. Pseudoprogression and MGMT status in glioblastoma patients: implications in clinical practice.
- Author
-
Fabi A, Russillo M, Metro G, Vidiri A, Di Giovanni S, and Cognetti F
- Subjects
- Aged, Antineoplastic Agents therapeutic use, Brain Neoplasms drug therapy, Brain Neoplasms enzymology, Brain Neoplasms radiotherapy, Combined Modality Therapy, Dacarbazine analogs & derivatives, Dacarbazine therapeutic use, Disease Progression, Female, Glioblastoma drug therapy, Glioblastoma enzymology, Glioblastoma radiotherapy, Humans, Magnetic Resonance Imaging, Male, Middle Aged, Temozolomide, Brain Neoplasms pathology, DNA Modification Methylases metabolism, DNA Repair Enzymes metabolism, Glioblastoma pathology, Tumor Suppressor Proteins metabolism
- Abstract
Pseudoprogression (PsPD) is a pathological feature recently reported by some authors in malignant glioma patients treated with radiotherapy in combination with temozolomide. In radiological imaging, it is shown as an increase in the size of the tumor lesion and contrast enhancement occurring within a few months from the completion of radio-chemotherapy without worsening of the neurological signs and symptoms. In 21%-50% of the patients, the same lesion disappears a few months after its appearance. In 12 glioblastoma patients treated with radio-chemotherapy, 4 cases of early radiological progression without discontinuation of temozolomide treatment are reported. At the sunsequent tumor assessment, 2 cases (13%) were revealed to be PsPD. The two patients who experienced PsPD had the longest progression and survival times of all patients. In both patients with PsPD, the O(6)-methylguanine-DNA methyltransferase (MGMT) promoter was found to be methylated. The PsPD phenomenon opens the prospect of a new era for the management of glioblastoma patients undergoing radio-chemotherapy.
- Published
- 2009
226. HIPK2 is involved in cell proliferation and its suppression promotes growth arrest independently of DNA damage.
- Author
-
Iacovelli S, Ciuffini L, Lazzari C, Bracaglia G, Rinaldo C, Prodosmo A, Bartolazzi A, Sacchi A, and Soddu S
- Subjects
- Apoptosis physiology, Base Sequence, Blotting, Western, Bone Marrow Cells cytology, Carrier Proteins genetics, Cell Differentiation, Cells, Cultured, DNA Primers, Humans, Muscle, Skeletal cytology, Protein Serine-Threonine Kinases genetics, Reverse Transcriptase Polymerase Chain Reaction, Carrier Proteins physiology, Cell Proliferation, DNA Damage, Protein Serine-Threonine Kinases physiology
- Abstract
Introduction/objectives: The serine/threonine kinase homeodomain-interacting protein kinase 2 (HIPK2) is a co-regulator of an increasing number of transcription factors and cofactors involved in DNA damage response and development. We and others have cloned HIPK2 as an interactor of the p53 oncosuppressor, and have studied the role of this interaction in cell response to stress. Nevertheless, our original cloning of HIPK2 as a p53-binding protein, was aimed at discovering partners of p53 involved in cell differentiation and development, still controversial p53 functions. To this aim, we used p53 as bait in yeast two-hybrid screening of a cDNA library from mouse embryo (day 11 postcoitus) when p53 is highly expressed., Methods and Results: In this study, we directly explored whether HIPK2 and p53 cooperate in cell differentiation. By measuring HIPK2 expression and activity in skeletal muscle and haemopoietic differentiation, we observed inverse behaviour of HIPK2 and p53--excluding cooperation activity of these two factors in this event. However, by HIPK2 depletion experiments, we showed that drastic HIPK2 suppression promotes cell-cycle arrest by induction of the cyclin-dependent kinase inhibitor p21(Waf-1/Cip-1). HIPK2 activity is independent of DNA damage and takes place in cell-cycle-arresting conditions, such as terminal differentiation, growth factor deprivation, and G(0) resting., Conclusions: HIPK2 was found to be involved in cell-cycle regulation dependent on p21(Waf-1/Cip-1) and independent of DNA damage.
- Published
- 2009
- Full Text
- View/download PDF
227. Progress and challenges in the vaccine-based treatment of head and neck cancers.
- Author
-
Venuti A
- Subjects
- Humans, Cancer Vaccines therapeutic use, Carcinoma, Squamous Cell drug therapy, Head and Neck Neoplasms drug therapy
- Abstract
Head and neck (HN) cancer represents one of the most challenging diseases because the mortality remains high despite advances in early diagnosis and treatment. Although vaccine-based approaches for the treatment of advanced squamous cell carcinoma of the head and neck have achieved limited clinical success, advances in cancer immunology provide a strong foundation and powerful new tools to guide current attempts to develop effective cancer vaccines. This article reviews what has to be rather what has been done in the field for the development of future vaccines in HN tumours.
- Published
- 2009
- Full Text
- View/download PDF
228. An E7-based therapeutic vaccine protects mice against HPV16 associated cancer.
- Author
-
Venuti A, Massa S, Mett V, Vedova LD, Paolini F, Franconi R, and Yusibov V
- Subjects
- Animals, Female, Mice, Mice, Inbred C57BL, Neoplasm Transplantation, Papillomavirus E7 Proteins, Nicotiana genetics, Vaccination, Neoplasms, Experimental prevention & control, Oncogene Proteins, Viral immunology, Papillomavirus Infections prevention & control, Papillomavirus Vaccines therapeutic use, Vaccines, Synthetic therapeutic use
- Abstract
Plant-derived vaccines represent an attractive strategy for cancer immunotherapy due to their relative safety and cost-effectiveness. We evaluated the anti-tumour activity of a Nicotiana benthamiana produced vaccine candidate based on the non-transforming E7 protein of HPV-16 fused to beta-1,3-1,4-glucanase of Clostridium thermocellum. Two doses of vaccine at two week intervals were administered to groups of C57BL/6 mice starting 3 or 6 days after challenge with tumourigenic E7-expressing TC-1* cells. Inhibition of tumour growth and increased survival was observed in both groups treated with vaccine. These data suggest the potential of plants as a platform for producing therapeutic vaccines.
- Published
- 2009
- Full Text
- View/download PDF
229. p63 regulation by microRNAs.
- Author
-
Blandino G and Moll UM
- Subjects
- Animals, Gene Expression Regulation, Humans, Organ Specificity, Tumor Suppressor Proteins genetics, MicroRNAs genetics, Tumor Suppressor Proteins metabolism
- Published
- 2009
- Full Text
- View/download PDF
230. Endothelial NOS, estrogen receptor beta, and HIFs cooperate in the activation of a prognostic transcriptional pattern in aggressive human prostate cancer.
- Author
-
Nanni S, Benvenuti V, Grasselli A, Priolo C, Aiello A, Mattiussi S, Colussi C, Lirangi V, Illi B, D'Eletto M, Cianciulli AM, Gallucci M, De Carli P, Sentinelli S, Mottolese M, Carlini P, Strigari L, Finn S, Mueller E, Arcangeli G, Gaetano C, Capogrossi MC, Donnorso RP, Bacchetti S, Sacchi A, Pontecorvi A, Loda M, and Farsetti A
- Subjects
- Basic Helix-Loop-Helix Transcription Factors genetics, Biomarkers, Cell Hypoxia physiology, Cell Line, Tumor, Cell Nucleus metabolism, Chromatin Assembly and Disassembly physiology, Cytoplasm metabolism, Enzyme Inhibitors pharmacology, Estradiol pharmacology, Gene Expression drug effects, Gene Expression genetics, Glucose Transporter Type 1 genetics, Humans, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Male, Nitric Oxide Synthase Type III antagonists & inhibitors, Nitric Oxide Synthase Type III genetics, Prognosis, Promoter Regions, Genetic genetics, Prostatic Neoplasms metabolism, Protein Binding drug effects, Protein Binding genetics, Response Elements genetics, Telomerase genetics, Telomerase metabolism, Tissue Array Analysis, Vascular Endothelial Growth Factor A genetics, Vascular Endothelial Growth Factor Receptor-2 genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Estrogen Receptor beta metabolism, Gene Expression Regulation, Neoplastic physiology, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Nitric Oxide Synthase Type III metabolism, Prostatic Neoplasms diagnosis
- Abstract
The identification of biomarkers that distinguish between aggressive and indolent forms of prostate cancer (PCa) is crucial for diagnosis and treatment. In this study, we used cultured cells derived from prostate tissue from patients with PCa to define a molecular mechanism underlying the most aggressive form of PCa that involves the functional activation of eNOS and HIFs in association with estrogen receptor beta (ERbeta). Cells from patients with poor prognosis exhibited a constitutively hypoxic phenotype and increased NO production. Upon estrogen treatment, formation of ERbeta/eNOS, ERbeta/HIF-1alpha, or ERbeta/HIF-2alpha combinatorial complexes led to chromatin remodeling and transcriptional induction of prognostic genes. Tissue microarray analysis, using an independent cohort of patients, established a hierarchical predictive power for these proteins, with expression of eNOS plus ERbeta and nuclear eNOS plus HIF-2alpha being the most relevant indicators of adverse clinical outcome. Genetic or pharmacologic modulation of eNOS expression and activity resulted in reciprocal conversion of the transcriptional signature in cells from patients with bad or good outcome, respectively, highlighting the relevance of eNOS in PCa progression. Our work has considerable clinical relevance, since it may enable the earlier diagnosis of aggressive PCa through routine biopsy assessment of eNOS, ERbeta, and HIF-2alpha expression. Furthermore, proposing eNOS as a therapeutic target fosters innovative therapies for PCa with NO inhibitors, which are employed in preclinical trials in non-oncological diseases.
- Published
- 2009
- Full Text
- View/download PDF
231. Treatment of recurrent malignant gliomas with fotemustine monotherapy: impact of dose and correlation with MGMT promoter methylation.
- Author
-
Fabi A, Metro G, Russillo M, Vidiri A, Carapella CM, Maschio M, Cognetti F, Jandolo B, Mirri MA, Sperduti I, Telera S, Carosi M, and Pace A
- Subjects
- Adult, Aged, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Antineoplastic Agents therapeutic use, DNA Methylation, Dose-Response Relationship, Drug, Follow-Up Studies, Glioma genetics, Glioma pathology, Humans, Karnofsky Performance Status, Middle Aged, Multivariate Analysis, Nausea chemically induced, Neoplasm Recurrence, Local, Neutropenia chemically induced, Nitrosourea Compounds administration & dosage, Nitrosourea Compounds adverse effects, O(6)-Methylguanine-DNA Methyltransferase genetics, Organophosphorus Compounds administration & dosage, Organophosphorus Compounds adverse effects, Promoter Regions, Genetic genetics, Retrospective Studies, Survival Analysis, Thrombocytopenia chemically induced, Treatment Outcome, Vomiting chemically induced, Glioma drug therapy, Nitrosourea Compounds therapeutic use, Organophosphorus Compounds therapeutic use
- Abstract
Background: In recurrent malignant gliomas (MGs), a high rate of haematological toxicity is observed with the use of fotemustine at the conventional schedule (100 mg/m(2) weekly for 3 consecutive weeks followed by triweekly administration after a 5-week rest period). Also, the impact of O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status on fotemustine activity has never been explored in the clinical setting., Methods: 40 patients with recurrent pretreated MG were identified as being treated with fotemustine at doses ranging from 65 mg/m(2) to 100 mg/m(2). Patients were classified into 3 groups according to the dose of fotemustine received, from the lowest dosage received in group A, to the highest in group C. Analysis of MGMT promoter methylation in tumor tissue was successfully performed in 19 patients., Results: Overall, 20% of patients responded to treatment, for a disease control rate (DCR, responses plus stabilizations) of 47.5%. Groups A and B experienced a response rate of 40% and 26.5% respectively, while the corresponding value for group C was 10%. Out of 19 patients, MGMT promoter was found methylated in 12 cases among which a DCR of 66.5% was observed. All 7 patients with unmethylated MGMT promoter were progressive to fotemustine., Conclusion: Low-dose fotemustine at 65-75 mg/m(2) (induction phase) followed by 75-85 mg/m(2) (maintenance phase) has an activity comparable to that of the conventional schedule. By determination of the MGMT promoter methylation status patients might be identified who are more likely to benefit from fotemustine chemotherapy.
- Published
- 2009
- Full Text
- View/download PDF
232. Endothelin-1 stimulates lymphatic endothelial cells and lymphatic vessels to grow and invade.
- Author
-
Spinella F, Garrafa E, Di Castro V, Rosanò L, Nicotra MR, Caruso A, Natali PG, and Bagnato A
- Subjects
- Cell Growth Processes physiology, Endothelial Cells pathology, Endothelin-1 biosynthesis, Humans, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Lymphangiogenesis, Receptor, Endothelin B biosynthesis, Signal Transduction, Vascular Endothelial Growth Factor A biosynthesis, Vascular Endothelial Growth Factor C biosynthesis, Vascular Endothelial Growth Factor Receptor-3 biosynthesis, Endothelial Cells metabolism, Endothelin-1 metabolism
- Abstract
The lymphatic vasculature is essential for tissue fluid homeostasis and cancer metastasis, although the molecular mechanisms involved remain poorly characterized. Endothelin-1 (ET-1) axis plays a crucial role in angiogenesis and tumorigenesis. Here, we first report that ET-1 acts as a lymphangiogenic mediator. We performed in vitro and in vivo studies and show that lymphatic endothelial cells produce ET-1, ET-3, and express the endothelin B receptor (ET(B)R). In these cells, ET-1 promotes proliferation, invasiveness, vascular-like structures formation, and phosphorylation of AKT and p42/44 mitogen-activated protein kinase through ET(B)R. In normoxic conditions, ET-1 is also able to up-regulate the expression of vascular endothelial growth factor (VEGF)-C, VEGF receptor-3, and VEGF-A, and to stimulate hypoxia-inducible factor (HIF)-1alpha expression similarly to hypoxia. Moreover, HIF-1alpha silencing by siRNA desensitizes VEGF-C and VEGF-A production in response to ET-1 or hypoxia, implicating HIF-1alpha/VEGF as downstream signaling molecules of ET-1 axis. Double immunofluorescence analysis of human lymph nodes reveals that lymphatic vessels express ET(B)R together with the lymphatic marker podoplanin. Furthermore, a Matrigel plug assay shows that ET-1 promotes the outgrowth of lymphatic vessels in vivo. ET(B)R blockade with the specific antagonist, BQ788, inhibits in vitro and in vivo ET-1-induced effects, demonstrating that ET-1 through ET(B)R directly regulates lymphatic vessel formation and by interacting with the HIF-1alpha-dependent machinery can amplify the VEGF-mediated lymphatic vascularization. Our results suggest that ET-1 axis is indeed a new player in lymphangiogenesis and that targeting pharmacologically ET(B)R and related signaling cascade may be therapeutically exploited in a variety of diseases including cancer.
- Published
- 2009
- Full Text
- View/download PDF
233. Class I HLA folding and antigen presentation in beta 2-microglobulin-defective Daudi cells.
- Author
-
Martayan A, Sibilio L, Tremante E, Lo Monaco E, Mulder A, Fruci D, Cova A, Rivoltini L, and Giacomini P
- Subjects
- Antibodies, Monoclonal metabolism, Antigen Presentation immunology, Cell Line, Tumor, Gene Expression Regulation immunology, HLA Antigens chemistry, HLA Antigens genetics, HLA Antigens immunology, HLA-A1 Antigen biosynthesis, HLA-A1 Antigen genetics, HLA-A1 Antigen immunology, HLA-A2 Antigen biosynthesis, HLA-A2 Antigen genetics, HLA-A2 Antigen immunology, HLA-B Antigens biosynthesis, HLA-B Antigens genetics, HLA-B Antigens immunology, Histocompatibility Antigens Class I chemistry, Histocompatibility Antigens Class I genetics, Histocompatibility Antigens Class I immunology, Humans, Membrane Proteins biosynthesis, Membrane Proteins genetics, Membrane Proteins immunology, Protein Precursors biosynthesis, Protein Precursors genetics, Protein Precursors immunology, Antigen Presentation genetics, HLA Antigens metabolism, Histocompatibility Antigens Class I metabolism, Protein Folding, beta 2-Microglobulin deficiency, beta 2-Microglobulin genetics
- Abstract
To present virus and tumor Ags, HLA class I molecules undergo a complex multistep assembly involving discrete but transient folding intermediates. The most extensive folding abnormalities occur in cells lacking the class I L chain subunit, called beta(2)-microglobulin (beta(2)m). Herein, this issue was investigated taking advantage of eight conformational murine mAbs (including the prototypic W6/32 mAb) to mapped H chain epitopes of class I molecules, four human mAbs to class I alloantigens, as well as radioimmunoprecipitation, in vitro assembly, pulse-chase, flow cytometry, and peptide-pulse/ELISPOT experiments. We show that endogenous (HLA-A1, -A66, and -B58) as well as transfected (HLA-A2) heavy chains in beta(2)m-defective Burkitt lymphoma Daudi cells are capable of being expressed on the cell surface, although at low levels, and exclusively as immature glycoforms. In addition, HLA-A2 is: 1) partially folded at crucial interfaces with beta(2)m, peptide Ag, and CD8; 2) receptive to exogenous peptide; and 3) capable of presenting exogenous peptide epitopes (from virus and tumor Ags) to cytotoxic T lymphocytes (bulk populations as well as clones) educated in a beta(2)m-positive environment. These experiments demonstrate a precursor-product relationship between novel HLA class I folding intermediates, and define a stepwise mechanism whereby distinct interfaces of the class I H chain undergo successive, ligand-induced folding adjustments in vitro as well as in vivo. Due to this unprecedented class I plasticity, Daudi is the first human cell line in which folding and function of class I HLA molecules are observed in the absence of beta(2)m. These findings bear potential implications for tumor immunotherapy.
- Published
- 2009
- Full Text
- View/download PDF
234. Beta-arrestin links endothelin A receptor to beta-catenin signaling to induce ovarian cancer cell invasion and metastasis.
- Author
-
Rosanò L, Cianfrocca R, Masi S, Spinella F, Di Castro V, Biroccio A, Salvati E, Nicotra MR, Natali PG, and Bagnato A
- Subjects
- Blotting, Western, Cell Line, Tumor, ErbB Receptors genetics, Female, Humans, Microscopy, Fluorescence, Ovarian Neoplasms metabolism, Phosphorylation, Transcriptional Activation, Transplantation, Heterologous, Tyrosine metabolism, beta Catenin chemistry, beta-Arrestin 1, beta-Arrestin 2, beta-Arrestins, Arrestins metabolism, Neoplasm Invasiveness, Neoplasm Metastasis, Ovarian Neoplasms pathology, Receptor, Endothelin A metabolism, Signal Transduction, beta Catenin metabolism
- Abstract
The activation of endothelin-A receptor (ET(A)R) by endothelin-1 (ET-1) has a critical role in ovarian tumorigenesis and progression. To define the molecular mechanism in ET-1-induced tumor invasion and metastasis, we focused on beta-arrestins as scaffold and signaling proteins of G protein-coupled receptors. Here, we demonstrate that, in ovarian cancer cells, beta-arrestin is recruited to ET(A)R to form two trimeric complexes: one through the interaction with Src leading to epithelial growth factor receptor (EGFR) transactivation and beta-catenin Tyr phosphorylation, and the second through the physical association with axin, contributing to release and inactivation of glycogen synthase kinase (GSK)-3beta and beta-catenin stabilization. The engagement of beta-arrestin in these two signaling complexes concurs to activate beta-catenin signaling pathways. We then demonstrate that silencing of both beta-arrestin-1 and beta-arrestin-2 inhibits ET(A)R-driven signaling, causing suppression of Src, mitogen-activated protein kinase (MAPK), AKT activation, as well as EGFR transactivation and a complete inhibition of ET-1-induced beta-catenin/TCF transcriptional activity and cell invasion. ET(A)R blockade with the specific ET(A)R antagonist ZD4054 abrogates the engagement of beta-arrestin in the interplay between ET(A)R and the beta-catenin pathway in the invasive program. Finally, ET(A)R is expressed in 85% of human ovarian cancers and is preferentially co-expressed with beta-arrestin-1 in the advanced tumors. In a xenograft model of ovarian metastasis, HEY cancer cells expressing beta-arrestin-1 mutant metastasize at a reduced rate, highlighting the importance of this molecule in promoting metastases. ZD4054 treatment significantly inhibits metastases, suggesting that specific ET(A)R antagonists, by disabling multiple signaling activated by ET(A)R/beta-arrestin, may represent new therapeutic opportunities for ovarian cancer.
- Published
- 2009
- Full Text
- View/download PDF
235. Novel activation domain derived from Che-1 cofactor coupled with the artificial protein Jazz drives utrophin upregulation.
- Author
-
Desantis A, Onori A, Di Certo MG, Mattei E, Fanciulli M, Passananti C, and Corbi N
- Subjects
- Acetylation drug effects, Binding Sites genetics, DNA-Binding Proteins chemistry, Genetic Therapy methods, HeLa Cells, Histones genetics, Histones metabolism, Humans, Peptides chemical synthesis, Peptides genetics, Peptides metabolism, Promoter Regions, Genetic genetics, Protein Engineering, Protein Structure, Tertiary genetics, Transcription Factors chemistry, Transcriptional Activation genetics, Utrophin metabolism, Apoptosis Regulatory Proteins genetics, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Repressor Proteins genetics, Transcription Factors genetics, Transcription Factors metabolism, Up-Regulation genetics, Utrophin genetics
- Abstract
Our aim is to upregulate the expression level of the dystrophin related gene utrophin in Duchenne muscular dystrophy, thus complementing the lack of dystrophin functions. To this end, we have engineered synthetic zinc finger based transcription factors. We have previously shown that the artificial three-zinc finger protein named Jazz fused with the Vp16 activation domain, is able to bind utrophin promoter A and to increase the endogenous level of utrophin in transgenic mice. Here, we report on an innovative artificial protein, named CJ7, that consists of Jazz DNA binding domain fused to a novel activation domain derived from the regulatory multivalent adaptor protein Che-1/AATF. This transcriptional activation domain is 100 amino acids in size and it is very powerful as compared to the Vp16 activation domain. We show that CJ7 protein efficiently promotes transcription and accumulation of the acetylated form of histone H3 on the genomic utrophin promoter locus.
- Published
- 2009
- Full Text
- View/download PDF
236. Adenocarcinoma of the esophagogastric junction: the role of abdominal-transhiatal resection.
- Author
-
Carboni F, Lorusso R, Santoro R, Lepiane P, Mancini P, Sperduti I, and Santoro E
- Subjects
- Adenocarcinoma pathology, Adult, Aged, Aged, 80 and over, Cardia pathology, Cardia surgery, Esophagogastric Junction pathology, Female, Humans, Lymph Nodes pathology, Lymph Nodes surgery, Lymphatic Metastasis, Male, Middle Aged, Neoplasm Invasiveness, Neoplasm Staging, Prognosis, Prospective Studies, Stomach Neoplasms pathology, Survival Rate, Treatment Outcome, Adenocarcinoma surgery, Digestive System Surgical Procedures, Esophagogastric Junction surgery, Stomach Neoplasms surgery
- Abstract
The surgical strategy for adenocarcinoma of the esophagogastric junction is still controversial. The aim of this study was to evaluate surgical results of the abdominal-transhiatal approach for 100 consecutively operated type II and III cardia adenocarcinoma, to clarify clinicopathological differences between these tumors, and to define prognostic factors. A prospectively maintained database identified 100 consecutively operated patients with Siewert type II and III cardia adenocarcinoma. Survival was analyzed by the Kaplan-Meier method. Differences between subgroups and prognostic factors were evaluated by the log rank test and Cox regression. Concerning clinicopathological characteristics, only the incidence of T1-2 stage was significantly higher in Siewert II type (P = .006). A complete (R0) resection was obtained in 74 patients (74%). Overall postoperative mortality and morbidity rates were 6% and 28%, respectively. Overall actuarial 5-year survival rate in resected patients was 27.4% (median 27 months), with 20.6% for type II and 34 for type III cancers (P = .07). Considering R0 resections, overall actuarial 5-year survival rate was 33.9% (median 33 months), with 26.7% for type II and 40.5 for type III cancer (P = .06). Pathologic T and N stage and R status were independent prognostic factors by multivariate analysis, and Siewert type showed a trend toward significance. The abdominal-transhiatal approach is a safe surgical approach, allowing complete tumor resection and adequate lymphadenectomy in these patients. True carcinoma of the cardia may be a distinct clinical entity with a more aggressive natural history than subcardial gastric carcinoma.
- Published
- 2009
- Full Text
- View/download PDF
237. Transcriptional regulation of hypoxia-inducible factor 1alpha by HIPK2 suggests a novel mechanism to restrain tumor growth.
- Author
-
Nardinocchi L, Puca R, Guidolin D, Belloni AS, Bossi G, Michiels C, Sacchi A, Onisto M, and D'Orazi G
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, Chromatin Assembly and Disassembly genetics, Humans, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Mice, Neoplasms blood supply, Neoplasms enzymology, Neovascularization, Pathologic genetics, Promoter Regions, Genetic genetics, Protein Binding, Protein Serine-Threonine Kinases deficiency, Repressor Proteins metabolism, Vascular Endothelial Growth Factor A genetics, Carrier Proteins metabolism, Gene Expression Regulation, Neoplastic, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Neoplasms genetics, Neoplasms pathology, Protein Serine-Threonine Kinases metabolism, Transcription, Genetic
- Abstract
HIPK2 has been implicated in restraining tumor progression by more than one mechanism, involving both its catalytic and transcriptional co-repressor functions. Starting from the finding that HIPK2 knockdown by RNA-interference (HIPK2i) induced significant up-regulation of HIF-1alpha mRNA and of its target VEGF in tumor cells, we evaluated the role of HIPK2 in transcriptional regulation of HIF-1alpha. We found that HIPK2 overexpression downmodulated both HIF-1alpha reporter activity and mRNA levels and showed that HIPK2 was bound in vivo to the HIF-1alpha promoter likely in a multiprotein co-repressor complex with histone deacetylase 1 (HDAC1). Thus, the HIF-1alpha promoter was strongly acetylated following HIPK2 knockdown. The HIF-1alpha-dependent VEGF transcription was evaluated by co-transfection of a dominant negative (DN) construct of HIF-1alpha that inhibited VEGF reporter activity induced by HIPK2 knockdown. HIF-1alpha and VEGF up-regulation in HIPK2i cells correlated with increased vascularity of tumor xenografts in vivo and tube formation in HUVEC in vitro. These findings provide the first evidence of HIPK2-mediated transcriptional regulation of HIF-1alpha that might play a critical role in VEGF expression.
- Published
- 2009
- Full Text
- View/download PDF
238. Inhibition of HIF-1alpha activity by homeodomain-interacting protein kinase-2 correlates with sensitization of chemoresistant cells to undergo apoptosis.
- Author
-
Nardinocchi L, Puca R, Sacchi A, and D'Orazi G
- Subjects
- Antibiotics, Antineoplastic pharmacology, Blotting, Western, Cell Hypoxia drug effects, Cell Hypoxia physiology, Cell Line, Tumor, Doxorubicin pharmacology, Humans, Reverse Transcriptase Polymerase Chain Reaction, Transfection, Tumor Suppressor Protein p53 metabolism, Apoptosis physiology, Carrier Proteins metabolism, Drug Resistance, Neoplasm physiology, Gene Expression Regulation, Neoplastic physiology, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
Background: Homeodomain-interacting protein kinase-2 (HIPK2), a transcriptional co-repressor with apoptotic function, can affect hypoxia-inducible factor 1 (HIF-1) transcriptional activity, through downmodulation of its HIF-1alpha subunit, in normoxic condition. Under hypoxia, a condition often found in solid tumors, HIF-1alpha is activated to induce target genes involved in chemoresistance, inhibition of apoptosis and tumor progression. Here, we investigated whether the HIPK2 overexpression could downregulate HIF-1alpha expression and activity in tumor cells treated with hypoxia-mimicking condition, and evaluated whether HIPK2-dependent downregulation of HIF-1alpha could sensitize chemoresistant tumor cells to adriamycin (ADR)-induced apoptosis., Methods: Tumor cell lines carrying wild-type p53, siRNA p53, or mutant p53 were overexpressed with HIPK2 (full length or catalytic inactive mutant) and treated with cobalt chloride (CoCl2) to mimic hypoxia, in the presence or absence of ADR treatment. HIF-1alpha expression was measured by semiquantitative reverse-transcriptase (RT)-PCR and Western immunoblotting and HIF-1 activity was evaluated by luciferase assay using reporter plasmid containing hypoxia response elements (HREs) upstream of luciferase gene. HIF-1 target genes, including multidrug resistance 1 (MDR1) and the antiapoptotic Bcl2 were determined by RT-PCR. Cell survival and apoptosis were measured by colony assay and cleavage of the caspase-3 substrate PARP, respectively., Results: Overexpression of HIPK2 resulted in downmodulation of cobalt-stabilized HIF-1alpha protein and HIF-1alpha mRNA levels, with subsequent inhibition of HIF-1 transcriptional activity. MDR1 and Bcl-2 gene expression was downmodulated by HIPK2 overexpression in cobalt-treated cells. Inhibition of HIF-1 transcriptional activity was dependent on HIPK2 catalytic activity. HIPK2 overexpression did not induce per se apoptosis of cobalt-treated cells, on the contrary it sensitized cobalt-treated cells to ADR-induced apoptosis, regardless of their p53 status., Conclusion: The ability of HIPK2 to restore the apoptosis-inducing potential of chemotherapeutic drug in hypoxia-mimicking condition and therefore to sensitize chemoresistant tumor cells suggests that HIPK2 may induce fundamental alterations in cell signaling pathways, involving or not p53 function. Thus potential use of HIPK2 is promising for cancer treatment by potentiating cytotoxic therapies, regardless of p53 cell status.
- Published
- 2009
- Full Text
- View/download PDF
239. Zoledronic acid for the treatment of appendicular osteosarcoma in a dog.
- Author
-
Spugnini EP, Vincenzi B, Caruso G, Baldi A, Citro G, Santini D, and Tonini G
- Subjects
- Animals, Bone Neoplasms diagnostic imaging, Bone Neoplasms drug therapy, Dog Diseases diagnostic imaging, Dogs, Euthanasia, Animal, Lameness, Animal complications, Lymph Node Excision, Lymphatic Metastasis, Male, Osteosarcoma diagnostic imaging, Osteosarcoma drug therapy, Radiography, Zoledronic Acid, Bone Density Conservation Agents administration & dosage, Bone Neoplasms veterinary, Diphosphonates administration & dosage, Dog Diseases drug therapy, Forelimb diagnostic imaging, Imidazoles administration & dosage, Osteosarcoma veterinary
- Abstract
A 10-year-old male intact Corso dog was referred for lameness and for a large neoplasm affecting the right foreleg. Physical examination of the patient revealed a 5 x 5 x 3 cm mass in the distal right foreleg. Histopathology was consistent with a diagnosis of appendicular osteosarcoma. The staging process found no evidence of metastasis. Because of the large size of the patient, the owners elected to treat their dog with antiresorptive therapy. The patient was treated with an infusion of zoledronic acid every 28 days. The tumour remained stable for 16 months and the lameness of the dog greatly improved. At that time, the patient returned for evaluation of a large rapidly growing prescapular mass. Biopsy confirmed lymph node metastasis and the dog was euthanased. Zoledronic acid showed remarkable palliation in our patient and possibly anti-tumour action and warrants further investigation.
- Published
- 2009
- Full Text
- View/download PDF
240. Pharmacokinetics of gemcitabine at fixed-dose rate infusion in patients with normal and impaired hepatic function.
- Author
-
Felici A, Di Segni S, Milella M, Colantonio S, Sperduti I, Nuvoli B, Contestabile M, Sacconi A, Zaratti M, Citro G, and Cognetti F
- Subjects
- Adenocarcinoma complications, Adenocarcinoma drug therapy, Adult, Aged, Antimetabolites, Antineoplastic therapeutic use, Area Under Curve, Biliary Tract Neoplasms complications, Biliary Tract Neoplasms drug therapy, Chromatography, High Pressure Liquid, Deoxycytidine pharmacokinetics, Deoxycytidine therapeutic use, Dose-Response Relationship, Drug, Drug Administration Schedule, Female, Floxuridine analogs & derivatives, Floxuridine pharmacokinetics, Humans, Infusions, Intravenous, Liver Diseases complications, Liver Diseases physiopathology, Liver Function Tests, Male, Middle Aged, Pancreatic Neoplasms complications, Pancreatic Neoplasms drug therapy, Prospective Studies, Tandem Mass Spectrometry, Gemcitabine, Adenocarcinoma blood, Antimetabolites, Antineoplastic pharmacokinetics, Biliary Tract Neoplasms blood, Deoxycytidine analogs & derivatives, Liver Diseases blood, Pancreatic Neoplasms blood
- Abstract
Background and Objectives: Gemcitabine (2,2-difluorodeoxycytidine [dFdC]) can be administered in a standard 30-minute infusion or in a fixed-dose-rate (FDR) infusion to maximize the rate of accumulation of triphosphate, its major intracellular metabolite. The standard 30-minute infusion requires dose adjustment in patients with organ dysfunction, especially in patients with elevated baseline serum bilirubin levels. On the other hand, the FDR infusion is burdened by increased haematological toxicity. The primary aim of this study was to evaluate the pharmacokinetics of dFdC and its metabolite difluorodeoxyuridine (dFdU) in patients with normal and impaired hepatic function., Patients and Methods: In this prospective study, patients with pancreatic or biliary tract carcinoma and normal or impaired hepatic function tests were considered eligible for recruitment. Patients were recruited according to the following criteria: (i) serum bilirubin <1.6 mg/dL and AST and ALT <2 times the upper the limit of normal (ULN) [cohort I]; and (ii) serum bilirubin >1.6 mg/dL and/or AST/ALT >2 times the ULN (cohort II). An FDR infusion of gemcitabine 1000 mg/m2 was administered on days 1, 8 and 15 every 4 weeks. The pharmacokinetic analysis of gemcitabine and dFdU was performed with high-performance liquid chromatography-tandem mass spectrometry assay in cycles 1 and 2., Results: Thirteen patients were enrolled, four in cohort I and nine in cohort II. All patients were assessable for toxicity and pharmacokinetic analysis. The grade and rate of toxicities were similar in both groups, and patients with elevation of bilirubin and/or transaminases did not require dose reduction of gemcitabine. Pharmacokinetic analysis revealed a reduction of the experimental area under the plasma concentration-time curve for gemcitabine and dFdU in patients with hepatic dysfunction when compared with patients with normal hepatic function. All other pharmacokinetic parameters were similar in the two cohorts. No statistical difference was demonstrated for all parameters evaluated between cycle 1 and cycle 2 in the two groups., Conclusion: Gemcitabine 1000 mg/m2 can be administered as an FDR infusion in patients with altered hepatic function without causing additional toxicity compared with patients with normal hepatic function.
- Published
- 2009
- Full Text
- View/download PDF
241. Restoring wtp53 activity in HIPK2 depleted MCF7 cells by modulating metallothionein and zinc.
- Author
-
Puca R, Nardinocchi L, Bossi G, Sacchi A, Rechavi G, Givol D, and D'Orazi G
- Subjects
- Apoptosis drug effects, Carrier Proteins metabolism, Cell Line, Tumor, DNA, Neoplasm metabolism, Doxorubicin pharmacology, Gene Expression Regulation, Neoplastic drug effects, Humans, Lentivirus, Phosphorylation drug effects, Phosphoserine metabolism, Protein Binding drug effects, Protein Conformation drug effects, Protein Folding drug effects, Protein Serine-Threonine Kinases metabolism, Transcription, Genetic drug effects, Transcriptional Activation drug effects, Tumor Suppressor Protein p53 chemistry, Tumor Suppressor Protein p53 genetics, Up-Regulation drug effects, Zinc pharmacology, Metallothionein metabolism, Protein Serine-Threonine Kinases deficiency, Tumor Suppressor Protein p53 metabolism, Zinc metabolism
- Abstract
The maintenance of p53 transactivation activity is important for p53 apoptotic function. We have shown that stable knockdown of HIPK2 induces p53 misfolding with inhibition of p53 target gene transcription. In this study we established a lentiviral-based system for doxycyclin (Dox)-induced conditional interference of HIPK2 expression to evaluate the molecular mechanisms involved in p53 deregulation. We found that HIPK2 knockdown induced metallothionein 2A (MT2A) upregulation as assessed by RT-PCR analysis, increased promoter acetylation, and increased promoter luciferase activity. The MT2A upregulation correlated with resistance to Adriamycin (ADR)-driven apoptosis and with p53 inhibition. Thus, acute knockdown of HIPK2 (HIPK2i) induced misfolded p53 protein in MCF7 breast cancer cells and inhibited p53 DNA-binding and transcription activities in response to ADR treatment. Previous works show that MT may modulate p53 activity through zinc exchange. Here, we found that inhibition of MT2A expression by siRNA in the HIPK2i cells restored p53 transcription activity. Similarly zinc supplementation to HIPK2i cells restored p53 transcription activity and drug-induced apoptosis. These data support the notion that MT2A is involved in p53 deregulation and strengthen the possibility that combination of chemotherapy and zinc might be useful to treat tumors with inactive wtp53.
- Published
- 2009
- Full Text
- View/download PDF
242. Electrochemotherapy for the treatment of squamous cell carcinoma in cats: a preliminary report.
- Author
-
Spugnini EP, Vincenzi B, Citro G, Tonini G, Dotsinsky I, Mudrov N, and Baldi A
- Subjects
- Animals, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell pathology, Cat Diseases pathology, Cats, Electrochemotherapy methods, Female, Male, Safety, Skin Neoplasms drug therapy, Skin Neoplasms pathology, Time Factors, Treatment Outcome, Carcinoma, Squamous Cell veterinary, Cat Diseases drug therapy, Electrochemotherapy veterinary, Skin Neoplasms veterinary
- Abstract
Squamous cell carcinomas (SCC) of the skin are commonly described in cats. Reported treatments include surgery, radiation therapy and photodynamic therapy. This preliminary study reports on the management of these lesions combining the local administration of bleomycin (plus hyaluronidase for a more uniform distribution) with permeabilizing biphasic electric pulses. Nine cats with SCC graded T(2)-T(4) were treated over a 5 year period, and each cat received two sessions of electrochemotherapy (ECT) 1 week apart. The side effects of this treatment were minimal and limited to mild erythema of the nose. Seven of the cats (77.7%) had a complete response lasting up to 3 years. ECT seems to be a safe and effective option for the treatment of feline sun-induced squamous cell carcinomas and warrants further investigation.
- Published
- 2009
- Full Text
- View/download PDF
243. Chemotherapy enhances vaccine-induced antitumor immunity in melanoma patients.
- Author
-
Nisticò P, Capone I, Palermo B, Del Bello D, Ferraresi V, Moschella F, Aricò E, Valentini M, Bracci L, Cognetti F, Ciccarese M, Vercillo G, Roselli M, Fossile E, Tosti ME, Wang E, Marincola F, Imberti L, Catricalà C, Natali PG, Belardelli F, and Proietti E
- Subjects
- Adult, CD8-Positive T-Lymphocytes metabolism, Female, Humans, Immunotherapy methods, Interferon-alpha metabolism, Leukocytes, Mononuclear metabolism, Male, Middle Aged, Neoplasm Metastasis, Pilot Projects, Treatment Outcome, Antineoplastic Agents pharmacology, Cancer Vaccines therapeutic use, Melanoma drug therapy, Melanoma immunology
- Abstract
Combination of chemotherapy with cancer vaccines is currently regarded as a potentially valuable therapeutic approach for the treatment of some metastatic tumors, but optimal modalities remain unknown. We designed a phase I/II pilot study for evaluating the effects of dacarbazine (DTIC) on the immune response in HLA-A2(+) disease-free melanoma patients who received anticancer vaccination 1 day following chemotherapy (800 mg/mq i.v.). The vaccine, consisting of a combination of HLA-A2 restricted melanoma antigen A (Melan-A/MART-1) and gp100 analog peptides (250 microg each, i.d.), was administered in combination or not with DTIC to 2 patient groups. The combined treatment is nontoxic. The comparative immune monitoring demonstrates that patients receiving DTIC 1 day before the vaccination have a significantly improved long-lasting memory CD8(+) T cell response. Of relevance, these CD8(+) T cells recognize and lyse HLA-A2(+)/Melan-A(+) tumor cell lines. Global transcriptional analysis of peripheral blood mononuclear cells (PBMC) revealed a DTIC-induced activation of genes involved in cytokine production, leukocyte activation, immune response and cell motility that can favorably condition tumor antigen-specific CD8(+) T cell responses. This study represents a proof in humans of a chemotherapy-induced enhancement of CD8(+) memory T cell response to cancer vaccines, which opens new opportunities to design novel effective combined therapies improving cancer vaccination effectiveness.
- Published
- 2009
- Full Text
- View/download PDF
244. YAP: at the crossroad between transformation and tumor suppression.
- Author
-
Bertini E, Oka T, Sudol M, Strano S, and Blandino G
- Subjects
- Animals, Apoptosis, Cell Differentiation, DNA-Binding Proteins metabolism, Humans, Nuclear Proteins metabolism, Phosphoproteins chemistry, Tumor Protein p73, Cell Transformation, Neoplastic metabolism, Phosphoproteins metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Yap is a small protein that binds to many transcription factors and modulates their activity. Yap was described to increase the ability of p73 in inducing apoptosis as a consequence of damage to the DNA, and therefore its activity was thought to favor tumor-suppression. However, other studies have recently shown a role for Yap in cell differentiation, cell transformation and in the regulation of organ size. It has been demonstrated that the Drosophila Hippo pathway has a mammalian equivalent, and that Yap is part of this pathway, where it might stimulate proliferation. In light of these new findings we ought to re-consider the role of Yap, which seems to be in service of several masters, and whose regulation--likely at the level of PTM--and cellular context might have a pivotal role in the choice of its partners and consequently on the final outcome.
- Published
- 2009
- Full Text
- View/download PDF
245. Posttranslational regulation of NF-YA modulates NF-Y transcriptional activity.
- Author
-
Manni I, Caretti G, Artuso S, Gurtner A, Emiliozzi V, Sacchi A, Mantovani R, and Piaggio G
- Subjects
- Amino Acid Sequence, Animals, CCAAT-Binding Factor genetics, Cell Line, Humans, Lysine genetics, Lysine metabolism, Mice, Molecular Sequence Data, Promoter Regions, Genetic, Proteasome Endopeptidase Complex metabolism, Proteasome Inhibitors, Protein Subunits genetics, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Sequence Alignment, Ubiquitination, p300-CBP Transcription Factors metabolism, CCAAT-Binding Factor metabolism, Gene Expression Regulation, Protein Processing, Post-Translational, Protein Subunits metabolism, Transcription, Genetic
- Abstract
NF-Y binds to CCAAT motifs in the promoter region of a variety of genes involved in cell cycle progression. The NF-Y complex comprises three subunits, NF-YA, -YB, and -YC, all required for DNA binding. Expression of NF-YA fluctuates during the cell cycle and is down-regulated in postmitotic cells, indicating its role as the regulatory subunit of the complex. Control of NF-YA accumulation is posttranscriptional, NF-YA mRNA being relatively constant. Here we show that the levels of NF-YA protein are regulated posttranslationally by ubiquitylation and acetylation. A NF-YA protein carrying four mutated lysines in the C-terminal domain is more stable than the wild-type form, indicating that these lysines are ubiquitylated Two of the lysines are acetylated in vitro by p300, suggesting a competition between ubiquitylation and acetylation of overlapping residues. Interestingly, overexpression of a degradation-resistant NF-YA protein leads to sustained expression of mitotic cyclin complexes and increased cell proliferation, indicating that a tight regulation of NF-YA levels contributes to regulate NF-Y activity.
- Published
- 2008
- Full Text
- View/download PDF
246. Oral ondansetron is highly active as rescue antiemetic treatment for moderately emetogenic chemotherapy: results of a randomized phase II study.
- Author
-
Fabi A, Ciccarese M, Metro G, Savarese A, Giannarelli D, Nuzzo CM, Russillo M, Sperduti I, Carbone I, Bria E, and Cognetti F
- Subjects
- Administration, Oral, Adult, Aged, Aged, 80 and over, Dose-Response Relationship, Drug, Female, Humans, Injections, Intramuscular, Male, Middle Aged, Antiemetics administration & dosage, Antineoplastic Agents adverse effects, Ondansetron administration & dosage, Vomiting drug therapy
- Abstract
Aims: In the present phase II randomized study, two different schedules of ondansetron were investigated as rescue antiemetic treatment for delayed emesis related to moderately emetogenic chemotherapy (MEC)., Materials and Methods: Patients scheduled to receive a first course of MEC were randomized to ondansetron 8 mg intramuscularly (arm A) or ondansetron 16 mg orally (arm B) as rescue antiemetic treatment for delayed emesis. Efficacy and safety evaluation was performed from days 2 to 6 through the administration of a diary plus a questionnaire in which the emetic episodes and the use of the assigned rescue treatment were recorded. All patients received standard prophylaxis for delayed emesis with oral dexamethasone 8 mg daily for 4 days starting on day 2., Results: Eighty-nine patients were enrolled into the study, of whom 44 were randomized to arm A and 45 to arm B. Twenty-two patients in each arm developed grade 1-2 delayed nausea/vomiting, all of which recurred to the rescue study treatment. Oral ondansetron resulted superior to intramuscular ondansetron in terms of complete response for nausea (77.3% vs 40.9%, respectively, p = 0.01) and vomiting (81.8% vs 31.8%, respectively, p = 0.001). Both schedules resulted to be very well tolerated, and no differences in toxicity were observed between the two arms of treatment. Furthermore, personal satisfaction about the use of the assigned rescue study medication was significantly higher in arm B., Conclusions: Due to its high efficacy and excellent tolerability, oral ondansetron is an important option in the management of MEC-related delayed emesis refractory to standard antiemetic prophylaxis.
- Published
- 2008
- Full Text
- View/download PDF
247. Detection of bovine papillomavirus type 2 in the peripheral blood of cattle with urinary bladder tumours: possible biological role.
- Author
-
Roperto S, Brun R, Paolini F, Urraro C, Russo V, Borzacchiello G, Pagnini U, Raso C, Rizzo C, Roperto F, and Venuti A
- Subjects
- Animals, Bovine papillomavirus 1 genetics, Carcinoma pathology, Carcinoma veterinary, Carcinoma virology, Cattle, Cattle Diseases blood, Cattle Diseases pathology, DNA, Viral analysis, DNA, Viral isolation & purification, Hematuria veterinary, Hematuria virology, Leukocytes, Mononuclear virology, Papilloma pathology, Papilloma veterinary, Papilloma virology, Papillomavirus Infections blood, Papillomavirus Infections virology, Sequence Analysis, DNA, Urinary Bladder pathology, Urinary Bladder Neoplasms blood, Urinary Bladder Neoplasms pathology, Urinary Bladder Neoplasms virology, Bovine papillomavirus 1 isolation & purification, Cattle Diseases virology, Papillomavirus Infections veterinary, Urinary Bladder virology, Urinary Bladder Neoplasms veterinary
- Abstract
Bovine papillomavirus type 2 (BPV-2) infection has been associated with urinary bladder tumours in adult cattle grazing on bracken fern-infested land. In this study, we investigated the simultaneous presence of BPV-2 in whole blood and urinary bladder tumours of adult cattle in an attempt to better understand the biological role of circulating BPV-2. Peripheral blood samples were collected from 78 cattle clinically suffering from a severe chronic enzootic haematuria. Circulating BPV-2 DNA was detected in 61 of them and in two blood samples from healthy cows. Fifty of the affected animals were slaughtered at public slaughterhouses and neoplastic proliferations in the urinary bladder were detected in all of them. BPV-2 DNA was amplified and sequenced in 78 % of urinary bladder tumour samples and in 38.9 % of normal samples as a control. Circulating episomal BPV-2 DNA was detected in 78.2 % of the blood samples. Simultaneous presence of BPV-2 DNA in neoplastic bladder and blood samples was detected in 37 animals. Specific viral E5 mRNA and E5 oncoprotein were also detected in blood by RT-PCR and Western blot/immunocytochemistry, respectively. It is likely that BPV-2 can persist and be maintained in an active status in the bloodstream, in particular in the lymphocytes, as a reservoir of viral infection that, in the presence of co-carcinogens, may cause the development of urinary bladder tumours.
- Published
- 2008
- Full Text
- View/download PDF
248. Gemcitabine at fixed dose-rate in patients with advanced soft-tissue sarcomas: a mono-institutional phase II study.
- Author
-
Ferraresi V, Ciccarese M, Cercato MC, Nuzzo C, Zeuli M, Di Filippo F, Giannarelli D, and Cognetti F
- Subjects
- Adult, Aged, Antimetabolites, Antineoplastic administration & dosage, Antimetabolites, Antineoplastic adverse effects, Combined Modality Therapy, Deoxycytidine administration & dosage, Deoxycytidine adverse effects, Deoxycytidine therapeutic use, Disease Progression, Dose-Response Relationship, Drug, Female, Gastrointestinal Diseases chemically induced, Hematologic Diseases chemically induced, Humans, Male, Middle Aged, Sarcoma pathology, Sarcoma therapy, Soft Tissue Neoplasms pathology, Soft Tissue Neoplasms therapy, Gemcitabine, Antimetabolites, Antineoplastic therapeutic use, Deoxycytidine analogs & derivatives, Sarcoma drug therapy, Soft Tissue Neoplasms drug therapy
- Abstract
Purpose: To explore the clinical activity and toxicity of gemcitabine infused at the fixed dose of 10 mg/m(2)/min over 100 min in patients with soft tissue sarcomas (STSs)., Patients and Methods: Fourteen patients with advanced locally unresectable and/or metastatic, pretreated STSs (seven leiomyosarcoma, three malignant schwannoma, one synovialsarcoma, one malignant fibrous histiocytoma, one endometrial stromal cell sarcoma, one undifferentiated) were treated with gemcitabine 10 mg/m(2)/min/week over 100 min given for 3 weeks out of 4. The median age was 52 years (range 27-77), male/female ratio was 3/11, and the median WHO performance status was 0 (range 0-1). The median number of previous medical treatments for advanced disease was 1 (range 1-2)., Results: A median number of three cycles (range 1-10 cycles) and a total of 151 weekly administrations (median 9, range 3-27) of gemcitabine were administered. Treatment was well tolerated and the main causes of dose-reduction or omission/delay were hematological and liver toxicities. One patient (7%; 95% confidence interval: 0.2-33.9%) with a metastatic uterine leiomyosarcoma obtained a partial response that lasted for 6.5 months. Three patients (two leiomyosarcoma and one schwannoma) (21%) obtained a stabilization of disease. The median time to progression was 3.1 months (range 1.0-9.5). The median overall survival was 11.8 months (range 1.0-54.5+)., Conclusions: Gemcitabine infused at the fixed dose of 10 mg/m(2)/min over 100 min shows a good tolerability but an overall modest activity in unselected STSs histotypes. Nevertheless, an interesting tumor growth control rate was observed in specific histological variants (i.e., leiomyosarcoma), thus confirming data from recent controlled clinical trials.
- Published
- 2008
- Full Text
- View/download PDF
249. Identification of the minimal melanocyte-specific promoter in the melanocortin receptor 1 gene.
- Author
-
Miccadei S, Pascucci B, Picardo M, Natali PG, and Civitareale D
- Subjects
- Animals, Codon, Initiator, Electrophoretic Mobility Shift Assay, HeLa Cells, Humans, Mice, NIH 3T3 Cells, Receptor, Melanocortin, Type 1 metabolism, Transcription, Genetic, Transfection, Melanocytes metabolism, Promoter Regions, Genetic genetics, Receptor, Melanocortin, Type 1 genetics
- Abstract
Background: The understanding of cutaneous pigmentation biology is relevant from the biologic and clinical point of view. The binding of alpha-melanocortin and its specific receptor, on the plasma membrane of melanin synthesising cells, plays a crucial role in melanins biosynthesis. Furthermore, loss of MC1R function is associated with an increased incidence of melanoma and non-melanoma skin cancer. The expression of the alpha-melanocortin receptor gene is highly controlled but, at the present, region responsible for tissue-specific activity of the gene promoter has not been identified., Methods: We have cloned the genomic sequences upstream the human MC1R coding gene. A DNA fragment of 5 kilobases upstream the human MC1R encoding sequence was placed in front of a reporter gene and several deletion mutants of such fragment have been prepared. These constructs have been tested for the ability to drive the melanocyte-specific gene expression of the reporter gene using transfection experiments in melanocyte and non-melanocyte cell lines. From these experiments we identified a DNA fragment with the ability to drive the gene transcription in a tissue-specific way and we used this small DNA fragment in DNA-protein interaction assays., Results: We show that the 150 base pairs upstream the MC1R gene initiation codon are able to drive the melanocyte-specific gene transcription. Furthermore, we provide experimental evidences suggesting that on such minimal melanocyte-specific gene promoter can assemble tissue-specific complexes., Conclusion: The present results strongly imply that the transcriptional regulation of the melanocyte-specific MC1R gene requires an internal promoter located in the 150 base pairs upstream the initiation codon.
- Published
- 2008
- Full Text
- View/download PDF
250. G-quadruplex ligand RHPS4 potentiates the antitumor activity of camptothecins in preclinical models of solid tumors.
- Author
-
Leonetti C, Scarsella M, Riggio G, Rizzo A, Salvati E, D'Incalci M, Staszewsky L, Frapolli R, Stevens MF, Stoppacciaro A, Mottolese M, Antoniani B, Gilson E, Zupi G, and Biroccio A
- Subjects
- Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Camptothecin administration & dosage, Drug Synergism, Female, G-Quadruplexes drug effects, Humans, Immunohistochemistry, Ligands, Male, Mice, Mice, Nude, Xenograft Model Antitumor Assays, Acridines therapeutic use, Antineoplastic Agents therapeutic use, Neoplasms, Experimental drug therapy
- Abstract
Purpose: The formation of G-quadruplex structures at telomeric DNA sequences blocks telomerase activity, offering an original strategy to design and develop new antitumor agents. The pentacyclic acridinium salt RHPS4 is one of the most effective and selective G4 ligands able to rapidly disrupt telomere architecture, resulting in apoptosis of cancer cells. Here, we studied the therapeutic index of RHPS4 and its integration with chemotherapeutics in preclinical model of solid tumors., Experimental Design: The antitumoral activity of RHPS4 was evaluated on human xenografts of different histotypes and compared with that of standard antineoplastic agents. Moreover, the effect of RHPS4/chemotherapeutics combinations on cell survival was studied and the most favorable combination was evaluated on tumor-bearing mice., Results: RHPS4 was active in vivo as single agent and showed a high therapeutic efficacy when compared with conventional drugs. Moreover, RHPS4 had antitumoral activity in human melanoma xenografts inherently resistant to chemotherapy and exhibited antimetastatic activity. RHPS4 also showed a strong synergistic interaction with camptothecins and this effect was strictly dependent on the drug sequence employed. Treatment of mice with irinotecan followed by RHPS4 was able to inhibit and delay tumor growth and to increase mice survival., Conclusions: Our data show that RHPS4 has a good pharmacodynamic profile and in combination therapy produces a strong antitumoral activity, identifying this drug as promising agent for clinical development.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.