Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Oncotarget ; 8(54): 92134-92142, 2017 Nov 03.
Article in English | MEDLINE | ID: mdl-29190903

ABSTRACT

Women with dense breast tissue on mammography are at higher risk of developing breast cancer but the underlying mechanisms are not well understood. De-regulation of microRNAs (miRNAs) has been associated with the onset of breast cancer. miRNAs in the extracellular space participate in the regulation of the local tissue microenvironment. Here, we recruited 39 healthy postmenopausal women attending their mammography-screen that were assessed having extreme dense or entirely fatty breasts (nondense). Microdialysis was performed in breast tissue and a reference catheter was inserted in abdominal subcutaneous fat for local sampling of extracellular compounds. Three miRNAs, associated with tumor suppression, miR-193b, miR-365a, and miR-452 were significantly down-regulated in dense breast tissue compared with nondense breast tissue. In addition, miR-452 exhibited significant negative correlations with several pro-inflammatory cytokines in vivo, which was confirmed in vitro by overexpression of miR-452 in breast cancer cells. No differences were found of miR-21, -29a, -30c, 146a, -148a, -203, or -451 in breast tissue and no miRs were different in plasma. Extracellular miRNAs may be among factors that should be included in studies of novel prevention strategies for breast cancer.

2.
Oncotarget ; 7(49): 81634-81644, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27835571

ABSTRACT

BACKGROUND: Radiotherapy is a well-established anti-cancer treatment. Although radiotherapy has been shown to significantly decrease the local relapse in rectal cancer patients, the rate of distant metastasis is still very high. The aim of this study was to evaluate whether AEG-1 is involved in radiation-enhanced migration and invasion in vitro and in a novel in vivo zebrafish model. RESULTS: Migration and invasion were decreased in all the AEG-1 knockdown cell lines. Furthermore, we observed that radiation enhanced migration and invasion, while AEG-1 knockdown abolished this effect. The results from the zebrafish embryo model confirmed the results obtained in vitro. MMP-9 secretion and expression were decreased in AEG-1 knockdown cells. MATERIALS AND METHODS: We evaluated the involvement of AEG-1 in migration and invasion and, radiation-enhanced migration and invasion by Boyden chamber assay in three colon cancer cell lines and respective stable AEG-1 knockdown cell lines. Furthermore, we injected those cells into zebrafish embryos and evaluated the amount of disseminated cells into the tail. CONCLUSION: AEG-1 knockdown inhibits migration and invasion, as well as radiation-enhanced invasion both in vitro and in vivo. We speculate that this is done via the downregulation of the intrinsic or radiation-enhanced MMP-9 expression by AEG-1 in the cancer cells. This study also shows, for the first time, that the zebrafish is a great model to study the early events in radiation-enhanced invasion.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Movement/radiation effects , Colonic Neoplasms/radiotherapy , Gene Knockdown Techniques , Radiation Tolerance , Animals , Cell Adhesion Molecules/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Matrix Metalloproteinase 9/metabolism , Membrane Proteins , Neoplasm Invasiveness , RNA Interference , RNA-Binding Proteins , Signal Transduction/radiation effects , Transfection , Zebrafish/embryology
3.
Clin Colorectal Cancer ; 13(1): 37-45.e4, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24503111

ABSTRACT

BACKGROUND: To investigate whether microRNAs are predictive of sensitivity to anti-epidermal growth factor receptor (EGFR) monoclonal antibodies in patients with metastatic colorectal cancer (mCRC). METHODS: A total of 183 mCRC cases from 2 independent cohorts (cohort 1: 74 cases; validation cohort: 109 cases) treated with cetuximab or panitumumab were included in the study. MiRNA arrays were analyzed using Agilent's miRNA platform. RESULTS: The study identified the cluster Let-7c/miR-99a/miR-125b as a signature associated with an outcome different from that of anti-EGFR therapies. In the first cohort, patients with high-intensity signatures had a significantly longer progression-free survival (PFS) (6.1 vs. 2.3 mo; P = .02) and longer overall survival (OS) ( 29.8 vs. 7.0 mo, P = .08) than patients with low-intensity signatures. In the validation cohort, patients with high signature had significantly longer PFS and OS than individuals with low-intensity signatures (PFS 7.8 vs. 4.3 mo, P = .02; OS 12.8 vs. 7.5 mo, P = .02). In the KRAS wild-type population (n = 120), high-intensity signature patients had a significantly longer PFS (7.8 vs. 4.6 mo, P = .016) and longer OS (16.1 vs. 10.9 mo, P = .09) than low-signature individuals, with no difference in KRAS mutated patients. CONCLUSION: The MiR-99a/Let-7c/miR-125b signature may improve the selection of patients with KRAS wild-type mCRC as good candidates for anti-EGFR therapy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal/therapeutic use , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , MicroRNAs/analysis , Adult , Aged , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Cetuximab , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/mortality , Disease-Free Survival , ErbB Receptors/antagonists & inhibitors , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Panitumumab , Proportional Hazards Models , Transcriptome
4.
Int J Oncol ; 43(3): 765-74, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23820752

ABSTRACT

MicroRNAs (miRNAs) play a key role in cancer pathogenesis and are involved in several human cancers, including non-small cell lung cancer (NSCLC). This study evaluated Let-7g and miR-21 expression by quantitative real-time PCR in 80 NSCLC patients and correlated the results with their main clinicopathological and molecular features. MiR-21 expression was significantly higher in NSCLC tissues compared to non-cancer lung tissues (p<0.0001), while no significant changes in Let-7g expression were observed between the tumor and normal lung tissues. Target prediction analysis led to the identification of 26 miR-21 and 24 Let-7g putative target genes that play important roles in cancer pathogenesis and progression. No significant association was observed between the analysed miRNAs and the main clinicopathological or molecular characteristics of the NSCLC patients, although both miRNAs were downregulated in squamous cell carcinomas compared to adenocarcinomas. Noteworthy, we observed a significant association between low Let-7g expression and metastatic lymph nodes at diagnosis (p=0.046), as well as between high miR-21 expression and K-Ras mutations (p=0.0003). Survival analysis did not show any significant correlation between prognosis and the analysed miRNAs, although the patients with a high Let-7g and miR-21 expression showed a significantly lower short-term progression-free survival (p=0.01 and p=0.0003, respectively) and overall survival (p=0.023 and p=0.0045, respectively). In conclusion, we showed that Let-7g and miR-21 expression was deregulated in NSCLC and we demonstrated a strong relationship between miR-21 overexpression and K-Ras mutations. Our data indicate that Let-7g and miR-21 profiling combined with the determination of K-Ras mutational status may be considered a useful biomarker for a more effective molecular characterization and clinical management of NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , MicroRNAs/genetics , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/pathology , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Genes, ras/genetics , Humans , Lung Neoplasms/pathology , Male , MicroRNAs/biosynthesis , Middle Aged , Mutation , Prognosis
5.
Oncol Rep ; 28(5): 1559-66, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22949056

ABSTRACT

Bronchopulmonary neuroendocrine tumours (BP-NETs) comprise a large spectrum of tumours including typical carcinoids (TCs), atypical carcinoids (ACs), large-cell neuroendocrine carcinomas (LCNECs) and small-cell lung carcinomas (SCLCs) that exhibit considerably different biological aggressiveness and clinical behaviours. The phosphatidylinositol-3-kinase α catalytic subunit (PIK3CA) gene is known to be involved in the pathogenesis of several types of human cancers through gene amplification, deletions or somatic missense mutations within the helical and catalytic domains. However, the PIK3CA gene status in BP-NETs has yet to be explored. This study aimed to investigate the PIK3CA gene status in a large series of BP-NETs by direct gene sequencing and to analyse its correlation with the main clinicopathological parameters. To the best of our knowledge, we demonstrated for the first time a high frequency of somatic missense mutations (23.2%) in the PIK3CA gene in a series of 190 BP-NETs, including 75 TCs, 23 ACs, 17 LCNECs and 75 SCLCs. The frequency of the PIK3CA gene mutation in the kinase domain was higher (17.9%) than that in the helical domain (5.3%). When the mutational status of the PIK3CA gene was compared with the main clinical and pathological characteristics of the BP-NET patients, we found a significant association between PIK3CA gene mutations and BP-NET histology (p=0.011). Interestingly, the frequency of PIK3CA gene mutations increased with the biological aggressiveness of all BP-NETs, except LCNECs. In conclusion, our results suggest that PIK3CA gene mutations may play a key role in tumourigenesis and aggressiveness of BP-NETs. The PIK3CA gene may represent a favourable candidate for an effective therapeutic strategy in the treatment of patients with BP-NETs.


Subject(s)
Lung Neoplasms/genetics , Neuroendocrine Tumors/genetics , Phosphatidylinositol 3-Kinases/genetics , Adult , Aged , Aged, 80 and over , Class I Phosphatidylinositol 3-Kinases , Female , Gene Amplification , Humans , Lung Neoplasms/metabolism , Male , Middle Aged , Mutation , Mutation, Missense , Neuroendocrine Tumors/metabolism , Neurosecretory Systems/pathology , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Young Adult
6.
Exp Ther Med ; 2(5): 787-792, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22977576

ABSTRACT

Bronchopulmonary neuroendocrine tumors (BP-NETs) are separated into four subgroups: typical carcinoid tumor (TC), atypical carcinoid tumor (AC), large-cell neuroendocrine carcinoma (LCNEC) and small-cell lung carcinoma (SCLC). The signaling pathway involving AKT/mammalian target of rapamycin (mTOR) is crucial to the regulation of cell growth, proliferation and survival, and is frequently activated in human cancers. Consequently, mTOR is considered an attractive target for anticancer agents. The present study aimed to evaluate the expression of phosphorylated AKT and mTOR in a series of BP-NETs, and to analyze the correlations with clinicopathological parameters. p-AKT and p-mTOR levels were determined by immunohistochemistry in a series of 210 BP-NETs, including 85 SCLCs, 17 LCNECs, 26 ACs, 75 TCs and 7 tumorlets. Higher p-AKT and p-mTOR expression levels were identified in the majority of tumorlets and carcinoids in comparison to the LCNECs (P=0.0001) and SCLCs (P=0.0002). Furthermore, a significant association was observed between p-mTOR expression and tumor size (T) in SCLCs (P=0.04) and LCNECs (P=0.03): T3-T4 tumors exhibited significantly lower p-mTOR expression compared to T1-T2 tumors. In conclusion, most of the BP-NETs examined in this study expressed p-AKT and p-mTOR, suggesting that the AKT/mTOR pathway plays an important role in these tumors. Additionally, our results confirm that low- to intermediate-grade tumors are more closely associated to each other than to high-grade tumors, despite sharing common classification and a common origin from neuroendocrine cells. These findings improve our knowledge of the biological characterization of these tumors and indicate new therapeutic opportunities for the treatment of BP-NETs.

7.
Hum Pathol ; 40(10): 1408-17, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19428048

ABSTRACT

The phosphoinositide 3-kinase/Akt pathway is involved in the pathogenesis of several human cancers. In this study, the biological and prognostic value of phosphoinositide 3-kinase/Akt pathway dysregulation was assessed by immunohistochemistry in a well-characterized series of 72 patients with node-negative breast cancer with a long-term follow-up. Phosphorylated Akt and PTEN expression was reduced in 32% and 12.5% of the tumors, respectively. Phosphorylated Akt or PTEN status was not associated with the main clinicopathologic and biological parameters, whereas their expression was tightly related to their downstream targets cyclin D1 and p27(Kip1) which are involved in cell proliferation. Survival analysis showed a strong association between a shorter disease-free survival and the dysregulated expression of phosphorylated Akt (P = .036), PTEN (P = .003), p27(Kip1) (P = .008), and Ki67 (P = .0007), or the distinct subtypes of breast tumors (luminal, HER2 overexpressing, and basal-like; P = .03). Moreover, multivariate analysis using the Cox proportional-hazards regression model showed that PTEN and Ki67 were independent predictive factors of disease recurrence and that their simultaneous dysregulation strongly increased the hazards ratio of the patients with node-negative breast cancer (hazards ratio, 38.30; P = .0014). In conclusion, our results show that the dysregulation of the phosphoinositide 3-kinase/Akt/PTEN pathway is relevant to the prognosis in node-negative breast carcinoma and that the evaluation of key components of this pathway might be a useful tool to identify the patients with node-negative breast cancer at high-risk of disease recurrence.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Adult , Aged , Aged, 80 and over , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Disease-Free Survival , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lymphatic Metastasis/pathology , Middle Aged , Neoplasm Staging , Prognosis
8.
Cancer Sci ; 100(6): 1026-33, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19385967

ABSTRACT

Anthracycline-based chemotherapy represents a milestone in the treatment of breast cancer. We previously demonstrated in an in vitro model that cyclin E overexpression is associated with increased expression of manganese superoxide dismutase (MnSOD) and resistance to doxorubicin. In the present study, immunohistochemical expression of cyclin E and MnSOD was evaluated in 134 early breast cancer patients receiving adjuvant epirubicin-based chemotherapy regimens containing epirubicin. Both parameters were correlated with the available clinicopathological parameters and with the outcome of patients. Overexpression of cyclin E and MnSOD was detected in 46 (34.3%) and 56 (41.8%) patients, respectively, and expression levels of the two proteins were related. Disease-free and alive patients displayed a lower mean percentage of cyclin E-expressing cells than relapsed and dead patients, respectively. Kaplan-Meier survival analysis demonstrated a significant separation between high versus low cyclin E-expressing tumors in terms of overall survival (P = 0.038 by log-rank). Similar results were obtained considering the subset of node-negative patients separately. No significant relationship with patient outcome was observed for MnSOD expression levels. At multivariate analysis cyclin E failed to demonstrate an independent prognostic value. In conclusion, the results of the present study support previous evidence that increased cyclin E expression is associated with higher MnSOD expression levels and poorer outcome, at least as evaluated in terms of overall survival. Further studies are warranted to evaluate the usefulness of cyclin E as a prognostic marker to identify breast cancer patients at higher risk of death from the disease when treated with adjuvant anthracycline-based therapy.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cyclin E/metabolism , Epirubicin/therapeutic use , Superoxide Dismutase/metabolism , Adult , Aged , Antibiotics, Antineoplastic/therapeutic use , Breast Neoplasms/enzymology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Combined Modality Therapy , Disease-Free Survival , Female , Humans , Immunohistochemistry , Middle Aged , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Survival Rate
9.
Pancreas ; 38(2): 184-96, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19002021

ABSTRACT

OBJECTIVES: Pancreatic cancer still remains a challenge for its biological complexity and lack of effective therapeutic strategies. Establishing new pancreatic cancer cell lines is therefore of paramount importance to clarify its biology. METHODS: We established and characterized 4 new pancreatic cancer cell lines (PP78, PP109, PP117, and PP161) according to their genetic (K-Ras, TP53, CDKN2A, and MADH4; DNA fingerprinting; karyotype), cytostructural (cytokeratins 7, 8, 18, and 19 vimentin, and ezrin), and functional profiles (doubling time; migration assay). RESULTS: K-Ras, TP53, and CDKN2A gene alterations were detected in all 4 of them. Each cell line had a unique DNA profile revealed by DNA fingerprinting. A complex karyotype with numerous structural and numeric chromosomal abnormalities was present in each cell line. All 4 cell lines showed positivity for cytokeratins 7, 8, and 18. All but PP78 expressed cytokeratin 19, whereas vimentin was expressed only in PP117 and PP78 cells. A different ezrin cellular distribution was noticed in PP78 and PP117, being mostly located at membrane ruffles. This peculiar distribution was associated with the strongest migratory capability. CONCLUSIONS: Our results seem to confirm the pancreatic ductal adenocarcinoma heterogeneity; in fact, the same genetic abnormalities (K-Ras, TP53, and CDKN2A) may have different effects on tumor biology depending on cellular differentiation.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Pancreatic Neoplasms/pathology , Adenocarcinoma/chemistry , Adenocarcinoma/genetics , Aged , Aged, 80 and over , Carcinoma, Pancreatic Ductal/chemistry , Carcinoma, Pancreatic Ductal/genetics , Cell Movement , Cell Proliferation , DNA Fingerprinting , Female , Genes, p16 , Genes, p53 , Genes, ras , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Karyotyping , Keratins/analysis , Male , Middle Aged , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/genetics , Phenotype , Smad4 Protein/genetics , Vimentin/analysis
10.
Thyroid ; 17(9): 861-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17705697

ABSTRACT

Loss-of-function mutations of the thyrotropin receptor (TSHr) may be responsible for congenital hypothyroidism or isolated hyperthyreotropinemia. To study cell surface expression of inactivating TSHr mutations detected in patients with isolated hyperthyreotropinemia (L252P, Q8fsX62, P27T, E34K, R46P, D403N, W488R, and M527T), we used the Agilent 2100 bioanalyzer to perform microchip flow cytometry analysis. The previously described TSHr inactivating mutation T477I was used as control. The level of receptor expression in COS-7 cells transfected with the T477I measured by binding assay was four times lower with respect to the wild-type TSHr. The very low expression of T477I was confirmed by fluorescence-activated cell sorting (FACS) analysis and by microchip flow cytometry analysis, suggesting that this method can be a reliable system to measure receptor cell surface expression. Other inactivating TSHr mutations were expressed in COS-7 cells for binding studies, FACS analysis, and microchip flow cytometry analysis. Binding studies showed that L252P, Q8fsX62, P27T, E34K, R46P, D403N, W488R, and M527T mutants had a low expression at the cell surface, as demonstrated by Bmax values. Data obtained by binding studies were in good agreement with data obtained by FACS analysis and microchip flow cytometry analysis. In conclusion, the low number of cells required for analysis and the ease of use make the microchip flow cytometry analysis a very reliable and favorable system to study cell surface expression of TSHr mutations.


Subject(s)
Flow Cytometry/methods , Microchip Analytical Procedures/methods , Receptors, Thyrotropin/biosynthesis , Animals , COS Cells , Chlorocebus aethiops , Humans , Mutation , Receptors, Thyrotropin/analysis , Receptors, Thyrotropin/genetics
11.
Int J Cancer ; 120(6): 1169-78, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17163420

ABSTRACT

BRMS1 is regarded as a metastasis suppressor gene for its ability to reduce metastatic potential of human and murine breast cancer cells as well as human melanoma cells. However, BRMS1 association to human tumor progression is not clearly understood. In the present study we analyzed BRMS1 mRNA expression in tumor progression and its potential prognostic value for breast carcinoma. BRMS1 mRNA expression level was quantified by real-time PCR in 47 tumoral, in 14 peritumoral and in 15 metastatic microdissected cellular populations from 47 breast cancer patients with 10-year follow up. We found BRMS1 expression to be higher in carcinoma cells than in matching normal epithelial cell populations in 10 out of 14 cases (p = 0.0005), while lymph-nodal carcinoma cells showed lower BRMS1 expression in 9 out of 15 cases (p = 0.001). Using both in vivo (human mammary breast carcinomas) and in vitro systems (breast cancer cell lines) we were able to demonstrate that BRMS1 overexpression was not a bias effect induced by cell proliferation rate. BRMS1 expression levels did not correlate with standard breast cancer prognostic factors but BRMS1 higher expression was associated with patient shorter disease-free and overall survival. Our findings are apparently inconsistent with the concept of BRMS1 as a metastasis suppressor gene. One possible explanation is that epithelial cells increase their BRMS1 expression as a compensatory response to tumor formation or metastasis progression, which is elevated in proportion to tumor aggressiveness, whereas those cells of the primary tumor that cannot upregulate BRMS1 escape to form metastasis.


Subject(s)
Breast Neoplasms/diagnosis , Carcinoma/diagnosis , Genes, Tumor Suppressor , Neoplasm Proteins/genetics , RNA, Messenger/analysis , Breast Neoplasms/pathology , Carcinoma/secondary , Epithelium/pathology , Female , Gene Expression , Humans , Repressor Proteins , Tumor Cells, Cultured
12.
Anticancer Res ; 26(6B): 4415-21, 2006.
Article in English | MEDLINE | ID: mdl-17201163

ABSTRACT

BACKGROUND: Available prognostic factors do not accurately identify node-negative breast cancer patients at high risk of disease recurrence and progression. PATIENTS AND METHODS: Cyclin A and E2F1 expression levels were evaluated in 75 consecutive node-negative breast cancer patients with a median follow-up of 10 years. Both parameters were tested for correlation with all the available clinicopathological parameters and with the clinical evolution of the disease. RESULTS: Cyclin A was overexprNed in 45.3% of patients and significantly related to large tumor size, high Ki67 and high E2F1 expression levels. No relationship was observed between cyclin A and tumor estrogen receptor (ER) status, grading or patient age. Seventeen patients relapsed within 5 years from diagnosis. Twelve (71%) of them showed cyclin A overexpression in comparison with 22 (38%) out of the 58 who did not relapse (p = 0.02). Disease-free survival (DFS) was significantly shorter in patients with cyclin A-overexpressing tumors compared to non-overexpressing ones (p = 0.01). DFS was also significantly longer in low vs. high Ki67 expression (p = 0.003) and in low vs. high E2F1 expression (p = 0.02). On multivariate analysis, the simultaneous high expression of all three parameters (cyclin A, Ki67 and E2Fl) was a strong independent prognostic factor for shorter DFS (HR 13.4). CONCLUSION: These findings suggest that assessment of cyclin A and/or E2F1 expression levels, associated with Ki67, might be useful for a better prognostic evaluation of node-negative breast cancer patients and support the need for further studies to evaluate their suitability for use in the routine clinical management of these patients.


Subject(s)
Breast Neoplasms/metabolism , Cyclin A/metabolism , E2F1 Transcription Factor/metabolism , Disease-Free Survival , Female , Humans , Immunohistochemistry , Middle Aged
13.
J Clin Endocrinol Metab ; 90(4): 2403-11, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15623821

ABSTRACT

Conventional chemotherapy and radiotherapy are ineffective for the treatment of advanced thyroid tumors like poorly differentiated papillary, anaplastic, and medullary thyroid cancer. In the attempt to evaluate the possibility of using retinoic acid (RA) in the treatment of thyroid cancer refractory to conventional therapy, we studied the effect of all-trans-RA treatment on five human thyroid cancer cell lines. We found that WRO and NPA, derived from follicular and poorly differentiated human thyroid carcinoma, respectively, showed a growth inhibition after 25 and 21 d of RA treatment. Both apoptosis and a decrease in DNA synthesis were observed as mechanisms of growth inhibition. In the NPA cell line, a delay of cell-cycle progression has also been observed. On the contrary, we did not observe any recovery of mRNA expression of thyroid-specific genes and in particular of the sodium iodide symporter gene. The lack of recovery of radioiodide uptake after all-trans-RA treatment confirmed the inability to reexpress sodium iodide symporter mRNA. The main difference between the all-trans-RA responding cells (WRO and NPA) and the nonresponding cells [ARO, FRO (derived from human anaplastic thyroid tumors) and TT (derived from human medullary thyroid tumor)] was the basal and all-trans-RA induced RA receptor (RAR)beta mRNA expression. Interestingly, 14 thyroid tumors (10 papillary and four anaplastic) showed a significant lower expression of RARbeta mRNA when compared with normal thyroid tissues. In agreement with this result, only 30% of papillary thyroid carcinomas analyzed were positive for RARbeta protein expression with a degree of expression that was much lower than that found in normal thyroid tissue. In conclusion we found that all-trans-RA treatment can determine a significant in vitro growth inhibition especially in differentiated thyroid tumor-derived cell lines but it seems unable to reinduce the expression of thyroid-specific genes and in particular to reinduce the ability to take up iodine. The growth inhibition is likely due to apoptosis in an early phase and to a decrease of DNA synthesis later. In some cases, a delay of the cell-cycle progression also may be responsible for the growth inhibition. The finding of a basal and RA-induced RARbeta mRNA expression only in cell lines responding to all-trans-RA suggests that the growth inhibition might be mediated by RARbeta.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , RNA, Messenger/analysis , Receptors, Retinoic Acid/genetics , Thyroid Gland/metabolism , Thyroid Neoplasms/drug therapy , Tretinoin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Immunohistochemistry , Receptors, Retinoic Acid/analysis , Receptors, Retinoic Acid/physiology , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL