Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
J Clin Med ; 11(4)2022 Feb 17.
Article in English | MEDLINE | ID: mdl-35207326

ABSTRACT

BACKGROUND: Renal transplantation represents the therapeutic gold standard in patients with end stage renal disease (ESRD). Still the role of pre-transplant dialysis in affecting time to transplantation has yet to be determined. We wanted to verify whether the type of renal replacement therapy (hemodialysis vs. peritoneal dialysis) affects time to transplantation and to identify clinical features related to the longer time to transplantation. METHODS: We performed a retrospective single-center observational study on patients who had received a transplant in the Bologna Transplant Unit from 1991 to 2019, described through the analysis of digital transplant list documents for sex, age, body mass index (BMI), blood group, comorbidities, underlying disease, serology, type of dialysis, time to transplantation, Panel Reactive Antibodies (PRA) max, number of preformed anti Human Leukocyte Antigens (HLA) antibodies. A p-value < 0.05 was considered statistically significant. RESULTS: In the 1619 patients analyzed, we observed a significant difference in time to transplant, PRA max and Preformed Antibodies Number between patients who received Hemodialysis (HD) and Peritoneal dialysis (PD). Then we performed a multiple regression analysis with all the considered factors in order to identify features that support these differences. The clinical variables that independently and directly correlate with longer time to transplantation are PRA max (p < 0.0001), Antibodies number (p < 0.0001) and HD (p < 0.0001); though AB blood group (p < 0.0001), age (p < 0.003) and PD (p < 0.0001) inversely correlate with time to transplantation. CONCLUSIONS: In our work, PD population received renal transplants in a shorter period of time compared to HD and turned out to be less immunized. Considering immunization, the type of dialysis impacts both on PRA max and on anti HLA antibodies.

2.
In Vivo ; 35(3): 1617-1624, 2021.
Article in English | MEDLINE | ID: mdl-33910844

ABSTRACT

BACKGROUND/AIM: More than half of deaths among hemodialysis patients are due to cardiovascular disease. This study examined whether intravenous administration of ferric carboxymaltose (FCM) has an impact on cardiovascular events in iron-deficient hemodialysis patients. PATIENTS AND METHODS: We performed a retrospective study concerning patients undergoing hemodialysis in our center from September 2016 to December 2019. We identified those who began FCM therapy (FCM group) during this period and those who did not (control group). We analyzed clinical, echocardiographic and laboratory parameters at the beginning (t0) and after one year (t1), to detect differences between the two groups. RESULTS: We identified 53 patients for the FCM group and 19 for the control group. Median follow-up was 1 year±3 months for both groups. In the FCM group, we observed a reduction in the doses of erythropoiesis-stimulating agents (ESA) (p<0.001) and a significative difference in cardiovascular events (p<0.01), but no differences in echocardiographic parameters. CONCLUSION: Patients who received FCM reached satisfactory values of transferrin saturation and ferritin, presented fewer coronary artery events and cardiovascular events, and could reduce doses of ESA.


Subject(s)
Anemia, Iron-Deficiency , Hematinics , Administration, Intravenous , Anemia, Iron-Deficiency/drug therapy , Humans , Iron/therapeutic use , Renal Dialysis , Retrospective Studies
3.
PLoS One ; 14(6): e0217937, 2019.
Article in English | MEDLINE | ID: mdl-31158261

ABSTRACT

AIM: Our goal has been to evaluate the importance that the incorporation of Bcl2 in the ER/PGR/Her2/Ki67 bio-profile can have as predictor of the Oncotype Dx categories. MATERIAL AND METHODS: 156 consecutive cases of HR+/Her2- pN0/1 primary breast carcinoma were sent to the Oncotype Dx test. Immunohistochemical determination of Bcl2/ER/PGR/Ki67/Her2 expression was evaluated for each case. After the selection of the appropriate cut-off values for PGR and Ki67, explorative as well as confirmative statistical analyses were performed to build and validate predictive risk-of-recurrence immunohistochemical only bio-profiles. RESULTS: The predictive capacity of these immunohistochemical profiles was compared with both traditional and TAILORx Oncotype Dx risk class classification. This comparison showed that immunohistochemical bio-profiles select those cases not associated with high risk-of-recurrence of disease (luminal-A/B and luminal A/B Bcl2) and those that are instead at high risk and therefore worthy of chemotherapy (luminal-B ki67 and luminal-B Bcl2/Ki67), strongly suggesting to only submit PGR-positive/Bcl2-Ki67 altered cases to Oncotype Dx, thus reducing the number of cases to be tested. CONCLUSIONS: Our results indicate that the addition of Bcl2 to an immunohistochemical bio-profile definitely improves its predictive capacity to correctly select which cases to send to the Oncotype Dx test. We have also suggested that institutions with a significant number of breast carcinomas sent to the Oncotype Dx test can use these latter to derive their own PGR and Ki67 cut-off values, overcoming the drawbacks of sharing common inter-laboratory values. Validation of these bio-profiles as predictors of the Oncotype Dx categories is ongoing in a prospective series of new cases.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Female , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Logistic Models , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , ROC Curve , Reproducibility of Results
4.
Sci Rep ; 7(1): 7458, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28785079

ABSTRACT

Patients with long-standing ulcerative colitis (UC) have an increased colorectal cancer (CRC) risk. In this pilot study we evaluated the effect of Eicosapentaenoic acid as free fatty acid (EPA-FFA) supplementation on mucosal disease activity, colonic differentiation markers and microbiota composition in UC patients. Twenty long-standing UC patients in stable clinical remission and with fecal calprotectin (FC) > 150 µg/g were enrolled (T0) and supplemented with EPA-FFA 2 g/daily for 90 days (T3). Endoscopic and histologic disease activities were measured by Mayo and Geboes scores, respectively. HES1, KLF4, STAT3, IL-10 and SOCS3 levels were determined using western blotting and qRT-PCR, while phospho-STAT3 levels were assessed by western blotting. Goblet cells were stained by Alcian blue. Microbiota analyses were performed on both fecal and colonic samples. Nineteen patients completed the study; seventeen (89.5%) were compliant. EPA-FFA treatment reduced FC levels at T3. Patients with FC > 150 µg/g at T3 (n = 2) were assumed as non-responders. EPA-FFA improved endoscopic and histological inflammation and induced IL-10, SOCS3, HES1 and KLF4 in compliant and responder patients. Importantly, long-term UC-driven microbiota composition was partially redressed by EPA-FFA. In conclusion, EPA-FFA supplementation reduced mucosal inflammation, promoted goblet cells differentiation and modulated intestinal microbiota composition in long-standing UC patients.


Subject(s)
Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/immunology , Eicosapentaenoic Acid/administration & dosage , Fatty Acids, Nonesterified/administration & dosage , Microbiota/drug effects , Adult , Aged , Aged, 80 and over , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/microbiology , Eicosapentaenoic Acid/pharmacology , Fatty Acids, Nonesterified/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Interleukin-10/genetics , Interleukin-10/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Leukocyte L1 Antigen Complex/metabolism , Male , Middle Aged , Phosphorylation/drug effects , Pilot Projects , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Treatment Outcome , Young Adult
5.
Sci Rep ; 6: 20670, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26864323

ABSTRACT

Aberrant NOTCH1 signalling is critically involved in multiple models of colorectal cancer (CRC) and a prominent role of NOTCH1 activity during inflammation has emerged. Epithelial to Mesenchymal Transition (EMT), a crucial event promoting malignant transformation, is regulated by inflammation and Metalloproteinase-9 (MMP9) plays an important role in this process. Eicosapentaenoic Acid (EPA), an omega-3 polyunsaturated fatty acid, was shown to prevent colonic tumors in different settings. We recently found that an extra-pure formulation of EPA as Free Fatty Acid (EPA-FFA) protects from colon cancer development in a mouse model of Colitis-Associated Cancer (CAC) through modulation of NOTCH1 signalling. In this study, we exposed colon cancer cells to an inflammatory stimulus represented by a cytokine-enriched Conditioned Medium (CM), obtained from THP1-differentiated macrophages. We found, for the first time, that CM strongly up-regulated NOTCH1 signalling and EMT markers, leading to increased invasiveness. Importantly, NOTCH1 signalling was dependent on MMP9 activity, upon CM exposure. We show that a non-cytotoxic pre-treatment with EPA-FFA antagonizes the effect of inflammation on NOTCH1 signalling, with reduction of MMP9 activity and invasiveness. In conclusion, our data suggest that, in CRC cells, inflammation induces NOTCH1 activity through MMP9 up-regulation and that this mechanism can be counteracted by EPA-FFA.


Subject(s)
Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Eicosapentaenoic Acid/pharmacology , Matrix Metalloproteinase 9/genetics , Monocytes/metabolism , Receptor, Notch1/genetics , Cell Differentiation , Cell Line , Cell Movement/drug effects , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation , HCT116 Cells , HT29 Cells , Humans , Inflammation , Lipopolysaccharides/pharmacology , Matrix Metalloproteinase 9/metabolism , Monocytes/cytology , Monocytes/drug effects , Receptor, Notch1/agonists , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/metabolism , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology
6.
New Microbiol ; 38(2): 211-23, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25938746

ABSTRACT

A high-throughput matrix-assisted laser desorption ionisation-time of flight (MALDI-TOF) mass spectrometry (MS)-based method was here developed to genotype 16 high-risk human papillomavirus (HPV) types in cervical cytology specimens. This method was compared to a commercial kit, the Inno-LiPA HPV genotyping assay, which detects a broad spectrum of HPV types. HPV DNA was assessed by the two methods in a total of 325 cervical cytology specimens collected in PreservCyt® solution. The overall agreement was almost perfect (Cohen's k=0.86) in term of positive and negative cases. Indeed, HPV types 16, 35, 56 and 66 showed the highest agreement values (>0.80). The highest agreement values (K >0.80) were found for all 16 HPV types in single infections, but only for HPV 16, 35, 45 and 56 in multiple infections. In conclusion, the high-throughput MS-based method developed here is well-suited for broad spectrum HPV genotyping in large-scale epidemiological studies.


Subject(s)
High-Throughput Screening Assays/methods , Papillomaviridae/isolation & purification , Papillomavirus Infections/virology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Female , Genotype , Humans , Papillomaviridae/chemistry , Papillomaviridae/classification , Papillomaviridae/genetics
7.
Oncotarget ; 5(21): 10607-20, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25431954

ABSTRACT

To successfully target Notch receptors as part of a multidrug anticancer strategy, it will be essential to fully characterize the factors that are modulated by Notch signaling. We recently reported that Notch3 silencing in HCC results in p53 up-regulation in vitro and, therefore, we focused on the mechanisms that associate Notch3 to p53 protein expression. We explored the regulation of p53 by Notch3 signalling in three HCC cell lines HepG2, SNU398 and Hep3B.We found that Notch3 regulates p53 at post-transcriptional level controlling both Cyclin G1 expression and the feed-forward circuit involving p53, miR-221 and MDM2. Moreover, our results were validated in human HCCs and in a rat model of HCC treated with Notch3 siRNAs. Our findings are becoming an exciting area for further in-depth research toward targeted inactivation of Notch3 receptor as a novel therapeutic approach for increasing the drug-sensitivity, and thereby improving the treatment outcome of patients affected by HCC. Indeed, we proved that Notch3 silencing strongly increases the effects of Nutilin-3.With regard to therapeutic implications, Notch3-specific drugs could represent a valuable strategy to limit Notch signaling in the context of hepatocellular carcinoma over-expressing this receptor.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cyclin G1/metabolism , Liver Neoplasms/metabolism , MicroRNAs/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Receptors, Notch/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Apoptosis , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Cycle , Cell Movement , Cell Proliferation , Cyclin G1/antagonists & inhibitors , Cyclin G1/genetics , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Liver Neoplasms/genetics , Male , Mutation/genetics , Proto-Oncogene Proteins c-mdm2/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Receptor, Notch3 , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
8.
Int J Cancer ; 135(9): 2004-13, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-24676631

ABSTRACT

Inflammatory bowel diseases are associated with increased risk of developing colitis-associated colorectal cancer (CAC). Epidemiological data show that the consumption of ω-3 polyunsaturated fatty acids (ω-3 PUFAs) decreases the risk of sporadic colorectal cancer (CRC). Importantly, recent data have shown that eicosapentaenoic acid-free fatty acid (EPA-FFA) reduces polyp formation and growth in models of familial adenomatous polyposis. However, the effects of dietary EPA-FFA are unknown in CAC. We tested the effectiveness of substituting EPA-FFA, for other dietary fats, in preventing inflammation and cancer in the AOM-DSS model of CAC. The AOM-DSS protocols were designed to evaluate the effect of EPA-FFA on both initiation and promotion of carcinogenesis. We found that EPA-FFA diet strongly decreased tumor multiplicity, incidence and maximum tumor size in the promotion and initiation arms. Moreover EPA-FFA, in particular in the initiation arm, led to reduced cell proliferation and nuclear ß-catenin expression, whilst it increased apoptosis. In both arms, EPA-FFA treatment led to increased membrane switch from ω-6 to ω-3 PUFAs and a concomitant reduction in PGE2 production. We observed no significant changes in intestinal inflammation between EPA-FFA treated arms and AOM-DSS controls. Importantly, we found that EPA-FFA treatment restored the loss of Notch signaling found in the AOM-DSS control and resulted in the enrichment of Lactobacillus species in the gut microbiota. Taken together, our data suggest that EPA-FFA is an excellent candidate for CRC chemoprevention in CAC.


Subject(s)
Colitis/complications , Colon/pathology , Colorectal Neoplasms/prevention & control , Eicosapentaenoic Acid/administration & dosage , Fatty Acids, Nonesterified/administration & dosage , Gastrointestinal Tract/drug effects , Microbiota/physiology , Receptors, Notch/metabolism , Animals , Apoptosis , Cell Proliferation , Colitis/chemically induced , Colitis/pathology , Colon/microbiology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Immunoenzyme Techniques , Inflammation/etiology , Inflammation/pathology , Inflammation/prevention & control , Male , Mice , Mice, Inbred C57BL , Microbiota/drug effects , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
9.
Mol Cancer Res ; 12(2): 203-16, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24324033

ABSTRACT

UNLABELLED: The overexpression of microRNA-221 (miR-221) is reported in several human cancers including hepatocellular carcinoma, and its targeting by tailored treatments has been proposed. The evidence supporting the role of miR-221 in cancer is growing and has been mainly focused on the discovery of miR-221 targets as well as on its possible therapeutic exploitations. However, the mechanism sustaining miR-221 aberrant expression remains to be elucidated. In this study, MDM2 (E3 ubiquitin-protein ligase homolog), a known p53 (TP53) modulator, is identified as a direct target of miR-221, and a feed-forward loop is described that sustains miR-221 aberrant expression. Interestingly, miR-221 can activate the p53/mdm2 axis by inhibiting MDM2 and, in turn, p53 activation contributes to miR-221 enhanced expression. Moreover, by modulating the p53 axis, miR-221 impacts cell-cycle progression and apoptotic response to doxorubicin in hepatocellular carcinoma-derived cell lines. Finally, CpG island methylation status was assessed as a causative event associated with miR-221 upregulation in hepatocellular carcinoma cells and primary tumor specimens. In hepatocellular carcinoma-derived cell lines, pharmacologically induced DNA hypomethylation potentiated a significant increase in miR-221 expression. These data were confirmed in clinical specimens of hepatocellular carcinoma in which elevated miR-221 expression was associated with the simultaneous presence of wild-type p53 and DNA hypomethylation. IMPLICATIONS: These findings reveal a novel miR-221-sustained regulatory loop that determines a p53-context-specific response to doxorubicin treatment in hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , MicroRNAs/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/genetics , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Doxorubicin/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Male , MicroRNAs/metabolism , Middle Aged , Phosphorylation , Proto-Oncogene Proteins c-mdm2/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Suppressor Protein p53/metabolism
11.
Oncotarget ; 4(10): 1618-31, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24113128

ABSTRACT

Sorafenib (Nexavar), a multiple kinase inhibitor, is the only clinically approved drug for patients with advanced HCC. However, its therapeutic success is limited by the emergence of drug resistance. Here we found that p21 and pGSK3ßSer9 are major players in the resistance to sorafenib. We recently reported that aberrant Notch3 expression in HCC contributes to doxorubicin resistance in vitro and, therefore, we focused on the mechanisms that associate Notch3 to acquired drug resistance. In this study we first found that Notch3 inhibition significantly increased the apoptosis inducing effect of sorafenib in HCC cells via specific down-regulation of p21 and up-regulation of pGSK3ßSer9. Using a mouse xenograft model we further found that Notch3 depletion combined with 21 days of sorafenib treatment exerts a substantial antitumor effect in vivo. Interestingly, we showed that, upon exposure to sorafenib treatment, Notch3 depleted xenografts maintain lower levels of p21 and higher levels of pGSK3ßSer9 than control xenografts. Thus, this study demonstrated that inhibition of Notch3 signaling prevents HCC-mediate drug resistance and sensitizes HCC cells to sorafenib. Finally, we validated our in vitro and in vivo results in primary human HCCs showing that Notch3 protein expression positively correlated with p21 protein expression and negatively correlated with pGSK3ßSer9 expression. In conclusion, the results presented in this study demonstrated that Notch3 silencing enhances the effect of sorafenib by overcoming drug resistance. Notch3 inhibition in combination with sorafenib can be a promising strategy for treatment of HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Glycogen Synthase Kinase 3/metabolism , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Receptors, Notch/antagonists & inhibitors , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Down-Regulation , Drug Synergism , Female , Gene Knockdown Techniques , Glycogen Synthase Kinase 3 beta , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Niacinamide/pharmacology , Phosphorylation , Receptor, Notch3 , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Sorafenib , Xenograft Model Antitumor Assays
12.
Prog Histochem Cytochem ; 47(4): 211-333, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23146330

ABSTRACT

Microscopy-based imaging is booming and the need for tools to retrieve quantitative data from images is urgent. This book provides simple but reliable tools to generate valid quantitative gene expression data, at the mRNA, protein and activity level, from microscopic images in relation to structures in cells, tissues and organs in 2D and 3D. Volumes, areas, lengths and numbers of cells and tissues can be calculated and related to these gene expression data while preserving the 2D and 3D morphology. Image cytometry thus provides a comprehensive toolkit to study molecular processes and structural changes at the level of cells and tissues.


Subject(s)
Image Cytometry/methods , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Image Processing, Computer-Assisted/instrumentation , Image Processing, Computer-Assisted/trends , Imaging, Three-Dimensional/instrumentation , Imaging, Three-Dimensional/trends
13.
Am J Pathol ; 181(2): 413-22, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22705236

ABSTRACT

CDKN1C/P57 is a cyclin-dependent kinase inhibitor implicated in different human cancers, including hepatocellular carcinoma (HCC); however, little is known regarding the role of CDKN1C/P57 and its regulation in HCC. In this study, we show that the down-regulation of Notch1 and Notch3 in two HCC cell lines resulted in Hes1 down-regulation, CDKN1C/P57 up-regulation, and reduced cell growth. In line with these data, we report that CDKN1C/P57 is a target of transcriptional repression by the Notch effector, Hes1. We found that the up-regulation of CDKN1C/P57 by cDNA transfection decreased tumor growth, as determined by growth curve, flow cytometry analysis, and cyclin D1 down-regulation, without affecting the apoptosis machinery. Indeed, the expression of Bax, Noxa, PUMA, BNIP(3), and cleaved caspase-3 was not affected by CDKN1C/P57 induction. Morphologically CDKN1C/p57-induced HCC cells became flat and lengthened in shape, accumulated the senescence-associated ß-galactosidase marker, and increased P16 protein expression. Evaluation of senescence in cells depleted both for Hes1 and CDKN1C/P57 revealed that the senescent state really depends on the accumulation of CDKN1C/p57. Finally, we validated our in vitro results in primary HCCs, showing that Hes1 protein expression inversely correlates with CDKN1C/P57 mRNA levels. In addition, reduced Hes1 protein expression is accompanied by a shorter time to recurrence after curative resection, suggesting that Hes1 may represent a biomarker for prediction of patients with poor prognosis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p57/genetics , Homeodomain Proteins/genetics , Receptor, Notch1/metabolism , Receptors, Notch/metabolism , Aged , Apoptosis/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Middle Aged , Receptor, Notch3 , Recurrence , Signal Transduction , Time Factors , Transcription Factor HES-1
14.
Cancer Lett ; 321(2): 187-94, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22343320

ABSTRACT

The ability to induce changes in cell membrane properties is nowadays considered an additional mechanism to explain the pharmacological effects of non-steroidal anti-inflammatory drugs (NSAIDs). We previously demonstrated that the NSAID Licofelone, a dual cyclooxygenase/5-lipoxygenase inhibitor, triggers apoptosis in HCA-7 colon cancer cells independently from the inhibition of these enzymes. Here, we provide evidence that, in HCA-7 cells, the pro-apoptotic effect of this drug relies, at least in part, on its ability to inhibit epidermal growth factor receptor (EGFR) signalling by a decrease of cell membrane fluidity. Indeed, Licofelone induced a relevant change in the relative proportions of some saturated, monounsaturated and polyunsaturated fatty acids constituting HCA-7 phospholipid fraction and significantly increased the levels of cholesterol in HCA-7 cell membrane. All of these changes resulted in a remarkable decrease of membrane fluidity. Such phenomenon was associated with the block of EGFR kinase activity and of its downstream targets, the p44-42 mitogen-activated protein kinase (MAPK) and AKT cascades, whose inhibitions were found to induce apoptosis in HCA-7 cells. Overall, these findings provide a new additional mechanism by which NSAIDs are effective toward colon cancer cells.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Cell Membrane/metabolism , Cell Survival/drug effects , Colonic Neoplasms/metabolism , ErbB Receptors/drug effects , Membrane Fluidity/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Pyrroles/pharmacology , Signal Transduction/drug effects , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Blotting, Western , Cell Line, Tumor , Colonic Neoplasms/drug therapy , ErbB Receptors/metabolism , Fluorescent Antibody Technique , Humans , Mitogen-Activated Protein Kinases/metabolism , Pyrroles/therapeutic use
15.
J Pathol ; 227(3): 275-85, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22262409

ABSTRACT

MiR-519d belongs to the chromosome 19 miRNA cluster (C19MC), the largest human miRNA cluster. One of its members, miR-519d, is over-expressed in hepatocellular carcinoma (HCC) and we characterized its contribution to hepatocarcinogenesis. In HCC cells, the over-expression of miR-519d promotes cell proliferation, invasion and impairs apoptosis following anticancer treatments. These functions are, at least in part, exerted through the direct targeting of CDKN1A/p21, PTEN, AKT3 and TIMP2. The mechanisms underlying miR-519d aberrant expression in HCC were assayed by genomic DNA amplification, methylation analysis and ChIP assay. The aberrant hypomethylation of C19MC and TP53 were respectively identified as an epigenetic change allowing the aberrant expression of miR-519d and one of the factors able to activate its transcription. In conclusion, we assessed the oncogenic role of miR-519d in HCC by characterizing its biological functions, including the modulation of response to anticancer treatments and by identifying CDKN1A/p21, PTEN, AKT3 and TIMP2 among its targets.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Methylation , Liver Neoplasms/enzymology , MicroRNAs/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tumor Suppressor Protein p53/metabolism , Aged , Aged, 80 and over , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Doxorubicin/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Tissue Inhibitor of Metalloproteinase-2/genetics , Transcription, Genetic/drug effects , Transfection , Tumor Suppressor Protein p53/genetics , Up-Regulation
16.
J Cell Mol Med ; 16(4): 740-51, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21645238

ABSTRACT

The pathways that control mesenchymal stem cells (MSCs) differentiation are not well understood, and although some of the involved transcription factors (TFs) have been characterized, the role of others remains unclear. We used human MSCs from tibial plateau (TP) trabecular bone, iliac crest (IC) bone marrow and Wharton's jelly (WJ) umbilical cord demonstrating a variability in their mineral matrix deposition, and in the expression levels of TFs including Runx2, Sox9, Sox5, Sox6, STAT1 and Slug, all involved in the control of osteochondroprogenitors differentiation program. Because we reasoned that the basal expression level of some TFs with crucial role in the control of MSC fate may be correlated with osteogenic potential, we considered the possibility to affect the hMSCs behaviour by using gene silencing approach without exposing cells to induction media. In this study we found that Slug-silenced cells changed in morphology, decreased in their migration ability, increased Sox9 and Sox5 and decreased Sox6 and STAT1 expression. On the contrary, the effect of Slug depletion on Runx2 was influenced by cell type. Interestingly, we demonstrated a direct in vivo regulatory action of Slug by chromatin immunoprecipitation, showing a specific recruitment of this TF in the promoter of Runx2 and Sox9 genes. As a whole, our findings have important potential implication on bone tissue engineering applications, reinforcing the concept that manipulation of specific TF expression levels may elucidate MSC biology and the molecular mechanisms, which promote osteogenic differentiation.


Subject(s)
Mesenchymal Stem Cells/metabolism , Transcription Factors/physiology , Animals , Apoptosis , Base Sequence , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , Core Binding Factor Alpha 1 Subunit/genetics , DNA Primers , Flow Cytometry , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Mesenchymal Stem Cells/immunology , Polymerase Chain Reaction , Promoter Regions, Genetic , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , SOX9 Transcription Factor/genetics , Sequence Homology, Nucleic Acid , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Mol Cancer ; 9: 300, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21092249

ABSTRACT

BACKGROUND: Basal-like carcinoma are aggressive breast cancers that frequently carry p53 inactivating mutations, lack estrogen receptor-α (ERα) and express the cancer stem cell markers CD133 and CD44. These tumors also over-express Interleukin 6 (IL-6), a pro-inflammatory cytokine that stimulates the growth of breast cancer stem/progenitor cells. RESULTS: Here we show that p53 deficiency in breast cancer cells induces a loss of methylation at IL-6 proximal promoter region, which is maintained by an IL-6 autocrine loop. IL-6 also elicits the loss of methylation at the CD133 promoter region 1 and of CD44 proximal promoter, enhancing CD133 and CD44 gene transcription. In parallel, IL-6 induces the methylation of estrogen receptor (ERα) promoter and the loss of ERα mRNA expression. Finally, IL-6 induces the methylation of IL-6 distal promoter and of CD133 promoter region 2, which harbour putative repressor regions. CONCLUSION: We conclude that IL-6, whose methylation-dependent autocrine loop is triggered by the inactivation of p53, induces an epigenetic reprogramming that drives breast carcinoma cells towards a basal-like/stem cell-like gene expression profile.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Interleukin-6/metabolism , AC133 Antigen , Antigens, CD/genetics , Antigens, CD/metabolism , Blotting, Western , Cell Line, Tumor , DNA Methylation/drug effects , DNA Methylation/genetics , DNA Methylation/physiology , Enzyme-Linked Immunosorbent Assay , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Fluorescent Antibody Technique , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Interleukin-6/genetics , Interleukin-6/pharmacology , Peptides/genetics , Peptides/metabolism , RNA Interference , RNA, Messenger , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/metabolism
18.
Clin Chem Lab Med ; 48(9): 1319-26, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20509833

ABSTRACT

BACKGROUND: Matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) proteomic signature might be of interest for the early detection and staging of hepatocellular carcinoma (HCC). However, published procedures have been criticized for the lack of data about analytical reproducibility, and the use of inadequate data processing. METHODS: MALDI-TOF profiling of peptides bound to serum albumin ("albuminome") was performed using 90 µL of serum from 45 study subjects (HCV-related cirrhosis, small, unifocal HCCs and advanced HCCs). To overcome the large intra-sample variability, a Quality Assurance protocol was implemented, and 4-8 samples for each subject were processed and analyzed. Overall, 522 subject samples and 299 quality-control spectra were analyzed. A machine-learning approach (Random Forest) was applied to analyze the data sets. RESULTS: Mean intra-sample coefficient of variation (CV) of the analytical procedure was 17.6%-30.0%; inter-subject CV was in the range 48.8%-71.3% among the three study groups. The Random Forest procedure correctly classified 433/522 "patient samples" and 295/299 "reference samples"; 43/45 patients were correctly classified following this approach. CONCLUSIONS: Our data suggest that, notwithstanding the large analytical variability found, multiple proteomic profiles obtained from each subject can differentiate cirrhosis with and without HCC, and HCCs with and without vascular invasion, warranting further investigation in a prospective setting.


Subject(s)
Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/pathology , Early Detection of Cancer/methods , Liver Neoplasms/diagnosis , Liver Neoplasms/pathology , Proteomics/methods , Serum Albumin/metabolism , Aged , Aged, 80 and over , Artificial Intelligence , Biomarkers, Tumor/blood , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/metabolism , Blood Vessels/pathology , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/complications , Data Interpretation, Statistical , Disease Progression , Female , Hepatitis C/complications , Humans , Liver Cirrhosis/blood , Liver Cirrhosis/complications , Liver Neoplasms/blood , Liver Neoplasms/complications , Male , Middle Aged , Molecular Weight , Neoplasm Invasiveness , Neoplasm Staging , Peptides/blood , Peptides/chemistry , Peptides/metabolism , Protein Binding , Reproducibility of Results , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
J Cell Physiol ; 225(3): 682-91, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20509143

ABSTRACT

Extracellular and intracellular mediators of inflammation, such as tumor necrosis factor alpha (TNFα) and NF-kappaB (NF-κB), play major roles in breast cancer pathogenesis, progression and relapse. SLUG, a mediator of the epithelial-mesenchymal transition process, is over-expressed in CD44(+)/CD24(-) tumor initiating breast cancer cells and in basal-like carcinoma, a subtype of aggressive breast cancer endowed with a stem cell-like gene expression profile. Cancer stem cells also over-express members of the pro-inflammatory NF-κB network, but their functional relationship with SLUG expression in breast cancer cells remains unclear. Here, we show that TNFα treatment of human breast cancer cells up-regulates SLUG with a dependency on canonical NF-κB/HIF1α signaling, which is strongly enhanced by p53 inactivation. Moreover, SLUG up-regulation engenders breast cancer cells with stem cell-like properties including enhanced expression of CD44 and Jagged-1 in conjunction with estrogen receptor alpha down-regulation, growth as mammospheres, and extracellular matrix invasiveness. Our results reveal a molecular mechanism whereby TNFα, a major pro-inflammatory cytokine, imparts breast cancer cells with stem cell-like features, which are connected to increased tumor aggressiveness.


Subject(s)
Breast Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation Mediators/metabolism , NF-kappa B/metabolism , Neoplastic Stem Cells/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Estrogen Receptor alpha/metabolism , Extracellular Matrix/metabolism , Female , Humans , Hyaluronan Receptors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/metabolism , NF-kappa B/genetics , Neoplasm Invasiveness , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Phenotype , RNA Interference , Recombinant Proteins/metabolism , Serrate-Jagged Proteins , Signal Transduction , Snail Family Transcription Factors , Spheroids, Cellular , Transcription Factors/genetics , Transfection , Tumor Suppressor Protein p53/metabolism , Up-Regulation
20.
Cancer Res ; 70(12): 5184-93, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20501828

ABSTRACT

MicroRNAs (miRNA) have rapidly emerged as modulators of gene expression in cancer in which they may have great diagnostic and therapeutic import. MicroRNA-199a-3p (miR-199a-3p) is downregulated in several human malignancies including hepatocellular carcinoma (HCC). Here, we show that miR-199a-3p targets mammalian target of rapamycin (mTOR) and c-Met in HCC cells. Restoring attenuated levels of miR-199a-3p in HCC cells led to G(1)-phase cell cycle arrest, reduced invasive capability, enhanced susceptibility to hypoxia, and increased sensitivity to doxorubicin-induced apoptosis. These in vitro findings were confirmed by an analysis of human HCC tissues, which revealed an inverse correlation linking miR-199a-3p and mTOR as well as a shorter time to recurrence after HCC resection in patients with lower miR-199a-3p expression. These results suggest that tactics to regulate mTOR and c-Met by elevating levels of miR-199a-3p may have therapeutic benefits in highly lethal cancers such as HCC.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Carcinoma, Hepatocellular/drug therapy , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Intracellular Signaling Peptides and Proteins/genetics , MicroRNAs/physiology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-met/genetics , Aged , Aged, 80 and over , Apoptosis/drug effects , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Luciferases/metabolism , Male , Middle Aged , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-met/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , TOR Serine-Threonine Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...