Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
2.
J Cell Physiol ; 235(6): 5363-5377, 2020 06.
Article in English | MEDLINE | ID: mdl-31967331

ABSTRACT

Ultrasound (US) offers potentially important opportunities from a therapeutic point of view. Thus, the study of the biological effects of US on cancer cells is important to understand the consequences of these changes on the malignant phenotype. This study aimed to investigate the effects of low-intensity ultrasound (LIPUS) on the phenotype of colorectal cancer cell lines. Cell proliferation was evaluated by viability test and by evaluation of pERK expression, while cell motility using the scratch test. Cell differentiation was evaluated assessing alkaline phosphatase activity. Epithelial mesenchymal transition was assessed by analyzing the expression of Vimentin and E-Cadherin. Release and uptake of extracellular vesicles (EVs) were evaluated by flow cytometry. LIPUS effects on the organization of cytoskeleton were analyzed by confocal microscopy and by evaluation of Rho GTPase expression. No alterations in vitality and clonogenicity were observed when the intermediate (0.4 MPa) and the lowest (0.035 MPa) acoustic intensities were administered while the treatment with high intensity (1 MPa) induced a reduction of both cell viability and clonogenicity in both cell lines in a frequency-dependent manner. LIPUS promoted the differentiation of colon cancer cells, affected epithelial-to-mesenchymal transition, promoted the closure of a wound as well as increased the release of EVs compared with untreated cells. LIPUS-induced increase in cell motility was likely due to a Rho GTPase-dependent mechanism. Overall, the results obtained warrant further studies on the potential combined effect of LIPUS with differentiating agents and on their potential use in a clinical setting.


Subject(s)
Cell Proliferation/radiation effects , Colorectal Neoplasms/radiotherapy , Osteogenesis/radiation effects , Ultrasonic Waves , Cadherins/genetics , Cell Differentiation/radiation effects , Cell Movement/radiation effects , Cell Survival/radiation effects , Cells, Cultured , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/radiation effects , Extracellular Vesicles/genetics , Extracellular Vesicles/radiation effects , Gene Expression Regulation, Neoplastic/radiation effects , HT29 Cells , Humans , Mesenchymal Stem Cells/radiation effects , Signal Transduction/radiation effects , rho GTP-Binding Proteins/genetics
3.
J Cell Physiol ; 234(9): 15459-15471, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30701538

ABSTRACT

Several clinical studies indicated that the daily use of aspirin or acetylsalicylic acid reduces the cancer risk via cyclooxygenases (Cox-1 and Cox-2) inhibition. In addition, aspirin-induced Cox-dependent and -independent antitumor effects have also been described. Here we report, for the first time, that aspirin treatment of human glioblastoma cancer (GBM) stem cells, a small population responsible for tumor progression and recurrence, is associated with reduced cell proliferation and motility. Aspirin did not interfere with cell viability but induced cell-cycle arrest. Exogenous prostaglandin E2 significantly increased cell proliferation but did not abrogate the aspirin-mediated growth inhibition, suggesting a Cox-independent mechanism. These effects appear to be mediated by the increase of p21 waf1 and p27 Kip1 , associated with a reduction of Cyclin D1 and Rb1 protein phosphorylation, and involve the downregulation of key molecules responsible for tumor development, that is, Notch1, Sox2, Stat3, and Survivin. Our results support a possible role of aspirin as adjunctive therapy in the clinical management of GBM patients.

4.
Nanotechnology ; 29(28): 285101, 2018 Jul 13.
Article in English | MEDLINE | ID: mdl-29664420

ABSTRACT

The topical treatment for oral mucosal diseases is often based on products optimized for dermatologic applications; consequently, a lower therapeutic effect may be present. 18-ß-glycyrrhetic acid (GA) is extracted from Glycirrhiza glabra. The first aim of this study was to test the cytotoxicity of GA on PE/CA-PJ15 cells. The second aim was to propose and test two different delivery systems, i.e. nanoparticles and fibers, to guarantee a controlled release of GA in vitro. We used chitosan and poly(lactic-co-glycolic) acid based nanoparticles and polylactic acid fibers. We tested both delivery systems in vitro on PE/CA-PJ15 cells and on normal human gingival fibroblasts (HGFs). The morphology of GA-loaded nanoparticles (GA-NPs) and fibers (GA-FBs) was investigated by electron microscopy and dynamic light scattering; GA release kinetics was studied spectrophotometrically. MTT test was used to assess GA cytotoxicity on both cancer and normal cells. Cells were exposed to different concentrations of GA (20-500 µmol l-1) administered as free GA (GA-f), and to GA-NPs or GA-FBs. ROS production was evaluated using dichlorodihydrofluorescein as a fluorescent probe. Regarding the cytotoxic effect of GA on PE/CA-PJ15 cells, the lowest TC50 value was 200 µmol l-1 when GA was added as GA-NPs. No cytotoxic effects were observed when GA was administered to HGFs. N-acetyl Cysteine reduced mortality induced by GA-f in PE/CA-PJ15 cells. The specific effect of GA on PE/CA-PJ15 cells is mainly due to the different sensitivity of cancer cells to ROS over-production; GA-NPs and GA-FBs formulations increase, in vitro, this toxic effect on oral cancer cells.


Subject(s)
Drug Delivery Systems , Glycyrrhetinic Acid/administration & dosage , Glycyrrhetinic Acid/therapeutic use , Mouth Neoplasms/drug therapy , Nanoparticles/chemistry , Cell Death/drug effects , Cell Line, Tumor , Chitosan/chemistry , Delayed-Action Preparations/pharmacology , Delayed-Action Preparations/therapeutic use , Drug Liberation , Dynamic Light Scattering , Fibroblasts/cytology , Fibroblasts/drug effects , Gingiva/cytology , Glycyrrhetinic Acid/pharmacology , Humans , Kinetics , Mouth Neoplasms/pathology , Nanofibers/chemistry , Nanofibers/ultrastructure , Nanoparticles/ultrastructure , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Reactive Oxygen Species/metabolism
5.
Dig Liver Dis ; 48(4): 391-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26804809

ABSTRACT

BACKGROUND: Infliximab is effective in inflammatory bowel disease through several mechanisms, possibly acting at the mucosal level. AIM: To assess the role of infliximab on intestinal mucosa and whether it contributes to mucosal healing. METHODS: Human colonic mucosal biopsies were incubated with or without infliximab. Cultured biopsies were evaluated for histological staining, CD68, CD3, E-cadherin and phospho-extracellular signal-regulated kinases (ERK) expression, and apoptosis. A scratch assay and MTT assay were performed with Caco2 cells in the presence of infliximab and/or tumour necrosis factor (TNF)-α or treated with supernatants obtained from human peripheral blood mononuclear cells or human intestinal fibroblasts treated with TNF-α and infliximab alone or in association. RESULTS: Infliximab-treated biopsies displayed a better histological appearance, reduced inflammation with an increase of E-cadherin, phospho-ERK and apoptosis. Supernatants showed lower TNF-α, IL-17, IL-6 and IL-8 concentration, with an increase in fibroblast-growth-factor. Motility at scratch assay and proliferation at MTT assay of Caco2 cells displayed differential modulation by TNF-α and infliximab, directly or through supernatants of human intestinal fibroblasts and human peripheral blood mononuclear cells exposed to them. CONCLUSION: Infliximab contributes to the mucosal healing process by acting directly at an intestinal mucosal level; infliximab indirectly affects epithelial cell migration and proliferation by acting on both fibroblasts and leukocytes.


Subject(s)
Colitis, Ulcerative/pathology , Gastrointestinal Agents/pharmacology , Infliximab/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Tumor Necrosis Factor-alpha/metabolism , Wound Healing/drug effects , Apoptosis/drug effects , Biopsy , Caco-2 Cells , Cadherins/metabolism , Cell Proliferation/drug effects , Cytokines/metabolism , Fibroblasts/drug effects , Humans , Interleukin-17/metabolism , MAP Kinase Signaling System
6.
Mol Cell Endocrinol ; 412: 205-15, 2015 Sep 05.
Article in English | MEDLINE | ID: mdl-25960164

ABSTRACT

Our group has previously shown that corticotropin releasing factor (CRF) inhibits proliferation of human endocrine-related cancer cell lines via the activation of CRF type-1 receptors (CRF-R1). Tumors originating from the nervous system also express CRF receptors but their role on neoplastic cell proliferation was poorly investigated. Here we investigated the effect of CRF receptor stimulation on nervous system-derived cancer cells, using the SK-N-SH (N) human neuroblastoma cell line as an experimental model. We found that SK-N-SH (N) cells express functionally active CRF-R1, whose activation by CRF and the cognate peptide urocortin (UCN) is associated to reduced cell proliferation and motility, as well as neuronal-like differentiation. UCN did not interfere with cell viability and cell-cycle arrest. Those effects seem to be mediated by a mechanism involving the activation of cAMP/PKA/CREB pathway and the subsequent downstream increase in p27(Kip1) and underphosphorylated retinoblastoma protein levels, as well as reduced c-Myc mRNA accumulation.


Subject(s)
Cell Differentiation , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/physiology , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Movement , Corticotropin-Releasing Hormone/physiology , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Neuroblastoma , Proto-Oncogene Proteins c-myc/genetics , Retinoblastoma Protein/metabolism , Urocortins/physiology
7.
J Exp Clin Cancer Res ; 31: 71, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22964035

ABSTRACT

BACKGROUND: Expression levels of CD133, a cancer stem cell marker, and of the α-subunit of the dystroglycan (α-DG) complex, have been previously reported to be altered in colorectal cancers. METHODS: Expression levels of CD133 and α-DG were assessed by immunohistochemistry in a series of colon cancers and their prognostic significance was evaluated. RESULTS: Scattered cells positive for CD133 were rarely detected at the bases of the crypts in normal colonic mucosa while in cancer cells the median percentage of positive cells was 5% (range 0-80). A significant correlation was observed with pT parameter and tumor stage but not with tumor grade and N status. Recurrence and death from disease were significantly more frequent in CD133-high expressing tumors and Kaplan-Meier curves showed a significant separation between high vs low expressor groups for both disease-free (p = 0.002) and overall (p = 0.008) survival.Expression of α-DG was reduced in a significant fraction of tumors but low α-DG staining did not correlate with any of the classical clinical-pathological parameters. Recurrence and death from the disease were significantly more frequent in α-DG-low expressing tumors and Kaplan-Meier curves showed a significant separation between high vs low expressor tumors for both disease-free (p = 0.02) and overall (p = 0.02) survival. Increased expression of CD133, but not loss of α-DG, confirmed to be an independent prognostic parameters at a multivariate analysis associated with an increased risk of recurrence (RR = 2.4; p = 0.002) and death (RR = 2.3; p = 0.003). CONCLUSIONS: Loss of α-DG and increased CD133 expression are frequent events in human colon cancer and evaluation of CD133 expression could help to identify high-risk colon cancer patients.


Subject(s)
Antigens, CD , Biomarkers, Tumor , Colonic Neoplasms , Dystroglycans , Glycoproteins , Peptides , AC133 Antigen , Aged , Antigens, CD/genetics , Antigens, CD/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Disease-Free Survival , Dystroglycans/genetics , Dystroglycans/metabolism , Female , Gene Expression Regulation, Neoplastic , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Staging , Peptides/genetics , Peptides/metabolism , Prognosis , Treatment Outcome
8.
Scand J Gastroenterol ; 47(10): 1211-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22856425

ABSTRACT

OBJECTIVE: Stage I colorectal carcinomas display a highly variable behavior which is not accurately predicted by the available prognostic markers. CD133 is considered a useful marker to identify the so-called cancer stem cells in colorectal cancers (CRCs) and its expression has been shown to have prognostic significance in CRC patients. This study aimed to verify whether immunohistochemical evaluation of CD133 might correlate with the progression risk of stage I CRC patients. MATERIAL AND METHODS: Expression levels of the CD133 molecule were analyzed and compared in two series of stage I surgically resected CRC patients showing disease progression and death for the disease and patients with no evidence of disease progression after at least 6 years after surgery. RESULTS: A positive staining for CD133 was detected in 52% of the cases with poor prognosis and only in 9% of the group with good prognosis, and this difference was highly significant (p < 0.001). A significant correlation was detected between CD133 expression and histological parameters, such as tumor budding, vascular invasion, and presence of lymph node micrometastases but not tumor grading, gender, and age. Disease-free survival and cancer-specific survival of CD133 negative tumors were significantly longer compared to positive cases. In multivariate analyses, CD133 staining confirmed to be a predictor of shorter survival independent from vascular invasion but not from lymph nodes micrometastases. CONCLUSIONS: These findings demonstrate that CD133 immunostaining is a useful predictor of high risk progression in stage I CRC patients and might help to identify patients eligible for adjuvant chemotherapy.


Subject(s)
Adenocarcinoma/secondary , Antigens, CD , Biomarkers, Tumor , Colorectal Neoplasms , Glycoproteins , Neoplastic Stem Cells/metabolism , Peptides , AC133 Antigen , Aged , Antigens, CD/analysis , Antigens, CD/metabolism , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Chemotherapy, Adjuvant/methods , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Disease Progression , Disease-Free Survival , Female , Glycoproteins/analysis , Glycoproteins/metabolism , Humans , Immunohistochemistry/methods , Kaplan-Meier Estimate , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Patient Selection , Peptides/analysis , Peptides/metabolism , Predictive Value of Tests , Prognosis , Risk Assessment/methods
9.
Magnes Res ; 24(3): S122-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21951649

ABSTRACT

The epithelial Mg(2+) channel TRPM6 is considered a pivotal component in active Mg(2+)absorption and re-absorption in the intestine and kidney, but its expression and function in other tissues are largely unknown. We have previously demonstrated that extracellular Mg(2+) availability modulates TRPM6, but not the ubiquitous TRPM7, in cultured mammary epithelial cells; in addition, TRPM6 protein expression correlated to Mg(2+) influx capacities. Our results closely remind the modulation of TRPM6 described by others in murine kidney and colon following Mg(2+) dietary restriction. We sought to validate our observations by investigating whether TRPM6 modulation by extracellular Mg(2+)also occurs in vivo. To this aim, we exploited a model consisting of rats fed either with a Mg(2+)-deficient or a Mg(2+)-enriched diet, and studied TRPM6 expression in breast and kidney tissues. Immunohistochemical and western blot analyses confirmed that rat mammary tissues express TRPM6 protein levels similar to those found in the kidney, and that protein expression is modulated by dietary Mg(2+). In particular, Mg(2+) restriction upregulated TRPM6 expression, while Mg(2+) supplementation resulted in a significant decrease in protein levels. This work confirms and extends our previous results on TRPM6 modulation by Mg(2+) availability in mammary tissues. Further studies are required to clarify the functional significance of these findings, and the role of TRPM6 in tissue-specific magnesium homeostasis.


Subject(s)
Dietary Supplements , Epithelial Cells/metabolism , Magnesium/pharmacology , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , TRPM Cation Channels/metabolism , Animals , Blotting, Western , Body Weight/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Immunohistochemistry , Kidney/drug effects , Kidney/metabolism , Magnesium/blood , Mammary Glands, Animal/drug effects , Organ Size/drug effects , Rats , Rats, Sprague-Dawley
10.
J Pathol ; 225(2): 305-14, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21826669

ABSTRACT

Several in vitro assays have been proposed to identify cancer stem cells (CSCs), including immunophenotyping, sphere assay and side population (SP) assay. CD133 antigen has been proposed as a CSC marker in colon cancer (CC). However, no functional data are available to date and conflicting results have been reported regarding its role as true CSC marker. Here we set out to identify a molecular signature associated with potential CSC. CD133(+) cells isolated from the CaCo-2 CC cell line were analysed by microarray molecular profiling compared to CD133(-) counterparts. Various differentially expressed genes were identified and the most relevant transcripts found to be over-expressed in CD133(+) cells were evaluated by quantitative RT-PCR in the CD133(+) fractions isolated from several CC cell lines. In the attempt to find a correlation between putative CSCs, isolated by means of CD133 immunophenotyping and the SP approach, we demonstrated a significant enrichment of CD133(+) cells within the SP fraction of CC cells, and comparison of the gene expression profiles revealed that Endothelin-1 (END-1) and nuclear receptor subfamily 4, group A, member 2 (NR4A2) transcripts are highly expressed in both CD133(+) and SP fractions of CC cells. Moreover, depletion of CD133 by siRNA induced a significant attenuation of END-1 and NR4A2 expression levels in CaCo-2 cells, while expression of all three molecules decreased during sodium butyrate-induced differentiation. In conclusion, we have identified a molecular signature associated with potential CSCs and showed for the first time the existence of a functional relationship between CD133, END-1 and NR4A2 expression in colon cancer cells.


Subject(s)
Antigens, CD/genetics , Colonic Neoplasms/genetics , Endothelin-1/genetics , Gene Expression Regulation, Neoplastic/genetics , Glycoproteins/genetics , Neoplastic Stem Cells/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Peptides/genetics , AC133 Antigen , Antigens, CD/metabolism , Blotting, Western , Caco-2 Cells , Cell Separation , Colonic Neoplasms/metabolism , Endothelin-1/metabolism , Flow Cytometry , Gene Expression Profiling , Glycoproteins/metabolism , Humans , Immunophenotyping , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Peptides/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
Carcinogenesis ; 31(10): 1813-21, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20699249

ABSTRACT

Several evidences suggest that cancer cells have abnormal cholesterol biosynthetic pathways and prenylation of small guanosine triphosphatase proteins. Tomato lycopene has been suggested to have beneficial effects against certain types of cancer, including that of prostate, although the exact molecular mechanism(s) is unknown. We tested the hypothesis that lycopene may exert its antitumor effects through changes in mevalonate pathway and in Ras activation. Incubation of the Ras-activated prostatic carcinoma LNCaP cells with a 24 h lycopene treatment (2.5-10 µM) dose dependently reduced intracellular total cholesterol by decreasing 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase expression and by inactivating Ras, as evidenced by its translocation from cell membranes to cytosol. Concomitantly, lycopene reduced the Ras-dependent activation of nuclear factor-kappaB (NF-κB). Such a reduction was parallel to an inhibition of reactive oxygen species production and to a decrease in the phosphorylation ofc-jun N-terminal kinase, extracellular signal-regulated kinase 1/2 and p38. These effects were also accompanied by an arrest of cell cycle progression and by apoptosis induction, as evidenced by a decrease in cyclin D1 and phospho-AKT levels and by an increase in p21, p27 and p53 levels and in Bax:Bcl-2 ratio. The addition of mevalonate prevented the growth-inhibitory effects of lycopene as well as its increase in Ras cytoplasmatic accumulation and the subsequent changes in NF-κB. The ability of lycopene in inhibiting HMG-CoA reductase expression and cell growth and in inactivating Ras was also found in prostate PC-3, colon HCT-116 and HT-29 and lung BEN cancer cells. These findings provide a novel mechanistic insight into the growth-inhibitory effects of lycopene in cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carotenoids/pharmacology , Mevalonic Acid/metabolism , Neoplasms/drug therapy , Signal Transduction/drug effects , ras Proteins/physiology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lycopene , Male , NF-kappa B/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Protein Transport/drug effects , Reactive Oxygen Species/metabolism
12.
Cancer Sci ; 101(9): 2080-6, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20626751

ABSTRACT

Expression levels of p27(kip1) , a negative regulator of the G1 phase of the cell cycle, and 8-hydroxydeoxyguanosine (8-OHdG), a marker of oxidative DNA damage, were assessed by immunostaining in a series of renal cell carcinomas (RCCs) and their prognostic significance was evaluated. Expression of p27(kip1) as well as of the α-subunit of the dystroglycan (DG) complex, previously reported to be altered in RCC, was also evaluated by western blot analysis. Nuclear expression of p27(kip1) was reduced in a significant fraction of tumors and low p27(kip1) staining correlated with higher tumor grade (P < 0.01). Recurrence and death from clear cell RCCs were significantly more frequent in p27(kip1) -low expressing tumors and Kaplan-Meier curves showed a significant separation between high vs low expressor groups for both disease-free (P = 0.011) and overall (P = 0.002) survival. Low nuclear expression of p27(kip1) as well as loss of α-DG were confirmed to be independent prognostic parameters at a multivariate analysis and the simultaneous loss of both molecules defined a "high-risk" group of patients with increased risk of recurrence (RR = 28.7; P = 0.01) and death (RR = 12.9; P = 0.03). No significant correlation with clinical or pathological parameters was found for 8-OHdG staining. Western blot analyses suggested a post-translational mechanism for the loss of α-DG expression and demonstrated that cytoplasmic dislocation of the protein contributes to the loss of active nuclear p27(kip1) . Loss of nuclear p27(kip1) is a frequent event in human RCCs and is a powerful predictor of poor outcome which, in combination with low DG expression, could help to identify high-risk patients with clear cell RCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Dystroglycans/metabolism , Kidney Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Carcinoma, Renal Cell/pathology , Cell Nucleus/metabolism , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Outcome Assessment, Health Care , Predictive Value of Tests , Prognosis , Young Adult
13.
J Exp Clin Cancer Res ; 29: 77, 2010 Jun 17.
Article in English | MEDLINE | ID: mdl-20565793

ABSTRACT

BACKGROUND: Total thyroidectomy is the treatment of choice in the majority of thyroid malignancies, preventing the risk of reoperative surgery due to recurrences. In order to assess the usefulness of such an approach, expression levels of inflammatory and proliferative markers were evaluated immunohistochemically in non-lesional adjacent thyroid tissues from a group of patients who underwent total thyroidectomy for different thyroid diseases. METHODS: Nineteen consecutive patients treated by total thyroidectomy for different thyroid diseases entered the study. IL-6Rb gp130 component of the IL-6 cytokine family members receptor complexes, STAT3 cytokine signalling transduction and transcription activation factor, p53 as tumour suppressor and CK19 cytokeratin as proliferation marker were analyzed in non-lesional thyroid tissues. RESULTS: Gp 130 expression was detected in all tissue samples with a scattered distribution while STAT3 and p53 positivity was observed in 17 out of 19 patients with a prevailing cytoplasmic localization. Cytokeratin 19 positivity was found in patients with papillary carcinoma, in one case of follicular adenoma, 3 multinodular goiters and one Basedow disease. CONCLUSION: Based on the results of this preliminary study, it may be concluded that the presence of a persisting cytokine-mediated activation associated with cytoplasmic localization of p53 is frequently observed in different thyroid diseases. Such a process seems to occur in the thyroid gland as a whole. Moreover, STAT3 activation as well as mutant p53 are risk factors for the development of neoplastic diseases. Total thyroidectomy may be supported as an adequate therapeutic approach for all the patients in whom overexpression of cytokine-dependent markers is detected.


Subject(s)
Adenoma/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Papillary/metabolism , Thyroid Neoplasms/metabolism , Thyroidectomy , Adenoma/pathology , Adenoma/surgery , Adult , Carcinoma, Papillary/pathology , Carcinoma, Papillary/surgery , Cytokine Receptor gp130/metabolism , Female , Humans , Immunoenzyme Techniques , Keratin-19/metabolism , Middle Aged , Pilot Projects , STAT3 Transcription Factor/metabolism , Thyroid Neoplasms/pathology , Thyroid Neoplasms/surgery , Tumor Suppressor Protein p53/metabolism , Young Adult
14.
Toxicol Ind Health ; 26(5): 309-17, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20371633

ABSTRACT

Cement is widely used for construction and several reports have suggested a potential toxicity of cement dusts although it has never been definitively assessed. To determine the cytotoxic and bioactive effects of cement dusts, cultures of normal rat fibroblasts were exposed to different types of cements and cell growth parameters, apoptosis and the occurrence of DNA damage (both in terms of DNA breaks and oxidative damage) were analyzed. Cells were exposed to cement extracts or cultured in direct contact with cement dusts and the results obtained were compared to cells cultured in fresh medium. A dose-dependent decrease in viable cells was observed with all tested cements. Different results were obtained in the cell-cement direct contact tests compared to the indirect contact tests performed using extracts. Inhibition of cell growth was associated in most cases with an accumulation of cells in the S-phase of the cell-cycle and the appearance of an apoptotic peak. DNA strand breaks, assessed by comet test, and increase in the levels of 8-OHdG, an important marker of DNA oxidative damage, always occurred by incubating cells in the presence of cement extracts or dusts. However, after removal of cement, a rapid damage repair was generally observed with an almost complete recovery within 12 hours. In conclusion, all cements analyzed in this study displayed a limited toxicity in vitro without significant differences amongst them. Overall, the results obtained indicate that cements should be treated as hazardous materials but they do not allow to make accurate predictions regarding the in vivo effects. Further studies are warranted to reach a better understanding of the potential toxic effects of cements, to identify the responsible mechanisms and to evaluate the possibility of modulating and/ or preventing them.


Subject(s)
Construction Materials/toxicity , DNA Damage/drug effects , Dust , Toxicity Tests/methods , Animals , Cell Cycle/drug effects , Cell Line, Transformed , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Environmental Exposure , Fibroblasts , Inhibitory Concentration 50 , Oxidative Stress/drug effects , Rats
15.
J Nutr Biochem ; 21(1): 34-46, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19157829

ABSTRACT

The present study was undertaken to examine whether lycopene is able to counteract 7-ketocholesterol (7-KC)-induced oxidative stress and apoptosis in human macrophages. Human THP-1 macrophages were exposed to 7-KC (10-25 microM) alone and in combination with lycopene (0.5-2 microM), and we monitored changes in cell oxidative status [reactive oxygen species (ROS) production, NOX-4, hsp70 and hsp90 expressions, 8-OHdG formation] and in cell proliferation and apoptosis. After 24 h of treatment, lycopene significantly reduced the increase in ROS production and in 8-OHdG formation induced by the oxysterol in a dose-dependent manner. Moreover, the carotenoid strongly prevented the increase of NOX-4, hsp70 and hsp90 expressions as well as the phosphorylation of the redox-sensitive p38, JNK and ERK1/2 induced by the oxysterol. The attenuation of 7-KC-induced oxidative stress by lycopene coincided with a normalization of cell growth in human macrophages. Lycopene prevented the arrest in G0/G1 phase of cell cycle induced by the oxysterol and counteracted the increased expression of p53 and p21. Concomitantly, it inhibited 7-KC-induced apoptosis, by limiting caspase-3 activation and the modulatory effects of 7-KC on AKT, Bcl-2, Bcl-xL and Bax. Comparing the effects of lycopene, beta-carotene and (5Z)-lycopene on ROS production, cell growth and apoptosis show that lycopene and its isomer were more effective than beta-carotene in counteracting the dangerous effects of 7-KC in human macrophages. Our study suggests that lycopene may act as a potential antiatherogenic agent by preventing 7-KC-induced oxidative stress and apoptosis in human macrophages.


Subject(s)
Apoptosis/drug effects , Carotenoids/pharmacology , Ketocholesterols/antagonists & inhibitors , Oxidative Stress/drug effects , 8-Hydroxy-2'-Deoxyguanosine , Cell Cycle/drug effects , Cells, Cultured , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/biosynthesis , HSP70 Heat-Shock Proteins/biosynthesis , HSP90 Heat-Shock Proteins/biosynthesis , Humans , Ketocholesterols/metabolism , Lycopene , Macrophages/drug effects , NADPH Oxidase 4 , NADPH Oxidases/biosynthesis , Reactive Oxygen Species/metabolism
16.
Cancer Lett ; 283(1): 108-17, 2009 Sep 28.
Article in English | MEDLINE | ID: mdl-19423215

ABSTRACT

The growth-inhibitory effects of the astaxanthin-rich Haematococcus pluvialis were studied in HCT-116 colon cancer cells. H. pluvialis extract (5-25 microg/ml) inhibited cell growth in a dose- and time-dependent manner, by arresting cell cycle progression and by promoting apoptosis. At 25 microg/ml of H. pluvialis extract, an increase of p53, p21(WAF-1/CIP-1) and p27 expression (220%, 160%, 250%, respectively) was observed, concomitantly with a decrease of cyclin D1 expression (58%) and AKT phosphorylation (21%). Moreover, the extract, at the same concentration, strongly up-regulated apoptosis by modifying the ratio of Bax/Bcl-2 and Bcl-XL, and increased the phosphorylation of p38, JNK, and ERK1/2 by 160%, 242%, 280%, respectively. Growth-inhibitory effects by H. pluvialis were also observed in HT-29, LS-174, WiDr, SW-480 cells. This study suggests that H. pluvialis may protect from colon cancer.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Eukaryota , Gene Expression/drug effects , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , Chromatography, High Pressure Liquid , Eukaryota/chemistry , Humans , In Situ Nick-End Labeling , Xanthophylls/isolation & purification , Xanthophylls/pharmacology
17.
Biochim Biophys Acta ; 1794(4): 615-26, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19336041

ABSTRACT

Drug induced cell differentiation represents a promising experimental model for proteomic analysis of cancer cells. In fact, by modulating and monitoring neoplastic cell differentiation it could be possible to identify cytodifferentiation related protein expression changes that can be subsequently utilized in vivo as potential cancer biomarkers. One main advantage of this approach is the significant reduction of biological variability normally observed in clinical biomarker research, with important implications also in prognosis and therapy. At this regard, a new class of differentiating agents is emerging, the so called PPAR-ligands, which however are characterized by a debated mechanism of action that has not been yet studied through a proteomic approach. To this aim, we investigated ciglitazone-induced differentiation of a human hepatocarcinoma HepG2 cell line, by monitoring biochemical and cellular parameters of cytodifferentiation and modifications of cellular protein profiles through 2-DE and MALDI-TOF analysis. Independent of the hypothesized mechanism of action of this intriguing PPARgamma agonist, results indicated that ciglitazone is a strong differentiating agent for the HepG2 cell line and that this process is associated with modifications of protein expression related to cell antioxidant systems, the cell cycle apparatus, signal transduction pathways, cellular stress and invasiveness. At last, considering these and other published data, a proteomic profile related to the cancer aggressiveness is beginning to emerge.


Subject(s)
Cell Differentiation/drug effects , Proteomics/methods , Thiazolidinediones/pharmacology , Analysis of Variance , Apoptosis/drug effects , Biomarkers, Tumor/analysis , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Electrophoresis, Gel, Two-Dimensional , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , PPAR gamma/agonists , PPAR gamma/pharmacology , Proteins/analysis , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
18.
Nutr Cancer ; 61(1): 131-6, 2009.
Article in English | MEDLINE | ID: mdl-19116883

ABSTRACT

Low Mg availability reversibly inhibited the growth of mammary epithelial HC11 cells by increasing the number of cells in the G0/G1 phase of the cell cycle. Because low Mg has been reported to promote oxidative reactions, we considered that low Mg-dependent growth arrest was mediated by oxidative stress. Surprisingly, both dichlorofluorescein-detectable reactive oxygen species and hydrogen peroxide-induced oxidative DNA damage were found to be lower in cells cultured in low Mg than in cells grown under control or high-Mg conditions. Gene expression profiling of low- and high-Mg cells showed the modulation of several genes, some regulating cell proliferation. In addition, low Mg cells also displayed overexpression of glutathione S-transferase (GST), leading to increased enzymatic activity. Of note, GST has been shown to modulate cell growth; therefore, we suggest that in low-Mg cells, GST upregulation might have a dual role in protecting against oxidative stress and in modulating cell proliferation.


Subject(s)
Cell Division/drug effects , Epithelial Cells/cytology , Magnesium Deficiency/pathology , Magnesium/pharmacology , Oxidative Stress/drug effects , Cell Division/physiology , Cell Line , Comet Assay , DNA Damage , Dose-Response Relationship, Drug , G1 Phase/drug effects , G1 Phase/physiology , Gene Expression Profiling , Glutathione Transferase/metabolism , Humans , Magnesium Deficiency/physiopathology , Oligonucleotide Array Sequence Analysis , Oxidative Stress/physiology , Reactive Oxygen Species , Resting Phase, Cell Cycle/drug effects , Resting Phase, Cell Cycle/physiology , Up-Regulation
19.
Br J Nutr ; 102(2): 207-14, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19105854

ABSTRACT

Lycopene beta-cyclase (tlcy-b) tomatoes, obtained by modulating carotenogenesis via genetic engineering, contain a large amount of beta-carotene, as clearly visible by their intense orange colour. In the present study we have subjected tlcy-b tomatoes to an in vitro simulated digestion and analysed the effects of digestate on cell proliferation. To this aim we used HT-29 human colon adenocarcinoma cells, grown in monolayers, as a model. Digested tomatoes were diluted (20 ml, 50 ml and 100 ml/l) in culture medium and added to the cells for different incubation times (24 h, 48 h and 72 h). Inhibition of cell growth by tomato digestate was dose-dependent and resulted from an arrest of cell cycle progression at the G0/G1 and G2/M phase and by apoptosis induction. A down-regulation of cyclin D1, Bcl-2 and Bcl-xl expression was observed. We also found that heat treatment of samples before digestion enhanced beta-carotene release and therefore cell growth inhibition. To induce with purified beta-carotene solubilised in tetrahydrofuran the same cell growth inhibition obtained with the tomato digestate, a higher amount of the carotenoid was necessary, suggesting that beta-carotene micellarised during digestion is utilised more efficiently by the cells, but also that other tomato molecules, reasonably made available during digestion, may be present and cooperate with beta-carotene in promoting cell growth arrest.


Subject(s)
Adenocarcinoma/drug therapy , Colonic Neoplasms/drug therapy , Down-Regulation , Intramolecular Lyases/therapeutic use , Solanum lycopersicum/enzymology , beta Carotene/therapeutic use , Adenocarcinoma/pathology , Analysis of Variance , Animals , Apoptosis/drug effects , Biomarkers/analysis , Caspase 3/analysis , Colonic Neoplasms/pathology , Cyclin D1/genetics , Digestion/physiology , Genes, bcl-2 , Genetic Markers , HT29 Cells , Humans , Interphase/drug effects , Solanum lycopersicum/genetics , Plants, Genetically Modified , Swine , bcl-X Protein/genetics , beta Carotene/analysis
20.
Apoptosis ; 13(9): 1172-83, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18679798

ABSTRACT

Different agents able to modulate apoptosis have been shown to modify the expression of the MAP-kinase-phosphatase-1 (MKP-1). The expression of this phosphatase has been considered a potential positive prognostic factor in lung cancer, and smoke was shown to reduce the levels of MKP-1 in ferret lung. Our aim was to assess whether the n-3 polyunsaturated fatty acid docosahexaenoic acid (DHA), known to inhibit the growth of several cancer cells mainly inducing apoptosis, may exert pro-apoptotic effect in lung cancer cells by modifying MKP-1 expression. We observed that DHA increased MKP-1 protein and mRNA expression and induced apoptosis in different lung cancer cell lines (mink Mv1Lu adenocarcinoma cells, human A549 adenocarcinoma and human BEN squamous carcinoma cells). We inhibited the pro-apoptotic effect of DHA by treating the cells with the phosphatase inhibitor Na(3)VO(4) or by silencing the MKP-1 gene with the specific siRNA. This finding demonstrated that the induction of apoptosis by DHA involved a phosphatase activity, specifically that of MKP-1. DHA reduced also the levels of the phosphorylated MAP-kinases, especially ERK1/2 and p38. Such an effect was not observed when the MKP-1 gene was silenced. Altogether, the data provide evidence that the DHA-induced overexpression of MKP-1 and the resulting decrease of MAP-kinase phosphorylation by DHA may underlie the pro-apoptotic effect of this fatty acid in lung cancer cells. Moreover, they support the hypothesis that DHA may exert chemopreventive action in lung cancer.


Subject(s)
Apoptosis/drug effects , Docosahexaenoic Acids/pharmacology , Down-Regulation/drug effects , Dual Specificity Phosphatase 1/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Lung Neoplasms/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Line, Tumor , Drug Screening Assays, Antitumor , Dual Specificity Phosphatase 1/antagonists & inhibitors , Dual Specificity Phosphatase 1/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphoproteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Vanadates/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...