Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Cancers (Basel) ; 15(17)2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37686663

ABSTRACT

Anaplastic thyroid carcinoma (ATC) is the most lethal subtype of thyroid cancer, with high invasive and metastatic potential, not responding to conventional treatments. Its aggressiveness may be influenced by macrophages, which are abundant cells in the tumor microenvironment. To investigate the role of macrophages in ATC aggressiveness, indirect co-cultures were established between ATC cell lines and THP-1-derived macrophages. Macrophages significantly increased both the migration and invasion of T235 cells (p < 0.01; p < 0.01), contrasting with a decrease in C3948 (p < 0.001; p < 0.05), with mild effects in T238 migration (p < 0.01) and C643 invasion (p < 0.05). Flow cytometry showed upregulation of CD80 (pro-inflammatory, anti-tumoral) and downregulation of CD163 (anti-inflammatory, pro-tumoral) in macrophages from co-culture with T235 (p < 0.05) and C3948 (p < 0.05), respectively. Accordingly, we found an upregulation of secreted pro-inflammatory mediators (e.g., GM-CSF, IL-1α; p < 0.05) in C3948-macrophage co-cultures. Proteomic analysis showed the upregulation of SPRY4, an inhibitor of the MAPK pathway, in C3948 cells from co-culture. SPRY4 silencing promoted cancer cell invasion, reverting the reduced invasion of C3948 caused by macrophages. Our findings support that macrophages play a role in ATC cell aggressiveness. SPRY4 is a possible modulator of macrophage-ATC cell communication, with a tumor suppressor role relevant for therapeutic purposes.

2.
Endocr Connect ; 12(9)2023 Aug 02.
Article in English | MEDLINE | ID: mdl-37410092

ABSTRACT

Nineteen cases of parathyroid carcinoma in patients with multiple endocrine neoplasia type 1 have been reported in the literature, of which 11 carry an inactivating germline mutation in the MEN1 gene. Somatic genetic abnormalities in these parathyroid carcinomas have never been detected. In this paper, we aimed to describe the clinical and molecular characterization of a parathyroid carcinoma identified in a patient with MEN1. A 60-year-old man was diagnosed with primary hyperparathyroidism during the postoperative period of lung carcinoid surgery. Serum calcium and parathyroid hormone levels were 15.0 mg/dL (8.4-10.2) and 472 pg/mL (12-65), respectively. The patient underwent parathyroid surgery, and histological findings were consistent with parathyroid carcinoma. Analysis of the MEN1 gene by next-generation sequencing (NGS) identified a novel germline heterozygous nonsense pathogenic variant (c.978C>A; p.(Tyr326*)), predicted to encode a truncated protein. Genetic analysis of the parathyroid carcinoma revealed a c.307del, p.(Leu103Cysfs*16) frameshift truncating somatic MEN1 variant in the MEN1 gene, which is consistent with MEN1 tumor-suppressor role, confirming its involvement in parathyroid carcinoma etiology. Genetic analysis of CDC73, GCM2, TP53, RB1, AKT1, MTOR, PIK3CA and CCND1 genes in the parathyroid carcinoma DNA did not detect any somatic mutations. To our knowledge, this is the first report of a PC case presenting both germline (first-hit) and somatic (second-hit) inactivation of the MEN1 gene.

3.
Eur Thyroid J ; 12(3)2023 06 01.
Article in English | MEDLINE | ID: mdl-36976625

ABSTRACT

Background: Treatment of advanced follicular thyroid carcinoma (FTC) is based primarily on indirect evidence obtained with multikinase inhibitors (MKI) in clinical trials in which papillary carcinomas represent the vast majority of cases. However, it should be noted that MKI have a non-negligible toxicity that may decrease the patient's quality of life. Conventional chemotherapy with GEMOX (gemcitabine plus oxaliplatin) is an off-label therapy, which seems to have some effectiveness in advanced differentiated thyroid carcinomas, with a good safety profile, although further studies are needed. Case report: We report a case of a metastatic FTC, resistant to several lines of therapy. However, with a durable response to GEMOX, the overall survival of our patient appears to have been extended significantly due to this chemotherapy. Conclusion: GEMOX may have a role in patients with thyroid cancer unresponsive to MKI.


Subject(s)
Adenocarcinoma, Follicular , Thyroid Neoplasms , Humans , Gemcitabine , Oxaliplatin/therapeutic use , Quality of Life , Adenocarcinoma, Follicular/drug therapy , Thyroid Neoplasms/drug therapy
4.
Eur Thyroid J ; 12(1)2023 02 01.
Article in English | MEDLINE | ID: mdl-36378538

ABSTRACT

Background: Anaplastic thyroid carcinoma (ATC) is one of the most aggressive solid tumors. ATC is frequently diagnosed at advanced stages with unresectable disease and palliative care is often indicated. Recently, several patient-tailored therapies for ATC are emerging due to advances in molecular profiling of these tumors. Entrectinib is a potent oral selective inhibitor of neutrotrophic tropomyosin receptor kinase (NTRK), ROS1, and anaplastic lymphoma kinase fusions. The experience regarding ATC and other thyroid carcinomas, particularly in the neoadjuvant setting, is minimal. Case report: We present a case of a 51-year-old female patient presenting with a bulky mass of the left thyroid lobe measuring 100 × 108 × 80 mm that was considered surgically unresectable. While waiting for next-generation sequence (NGS) profiling, lenvatinib was initiated. There was an initial clinical and imagiologic response; however, progression occurred after 12 weeks, and at this time NGS identified an ETV6-NTRK3 fusion and entrectinib was started. After 12 weeks, tumor diameters reduced to a minimum of 68×60×49 mm, and the patient underwent total thyroidectomy plus central lymphadenectomy. Histological diagnosis confirmed an ATC (pT4a R2 N1a). Adjuvant radiotherapy (RT) (60 Grays) with weekly paclitaxel (45 mg/m2) was then administered followed by maintenance entrectinib 600 mg daily. Fluorodeoxyglucose positron emission tomography performed 3 months after completion of RT showed only non-specific uptake in the posterior wall of the hypopharynx and larynx, suggestive of inflammation. Conclusion: We report the first case of an ATC with a dramatic response to neoadjuvant therapy with entrectinib, which enabled surgical resection of an ab initio unresectable tumor.


Subject(s)
Thyroid Carcinoma, Anaplastic , Thyroid Neoplasms , Female , Humans , Middle Aged , Thyroid Carcinoma, Anaplastic/genetics , Neoadjuvant Therapy , Protein-Tyrosine Kinases/therapeutic use , Proto-Oncogene Proteins/therapeutic use , Thyroid Neoplasms/diagnostic imaging
6.
J Clin Endocrinol Metab ; 106(10): e3911-e3922, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34134139

ABSTRACT

CONTEXT: The recommendations for radioactive-iodine treatment (RAIT) in metastatic differentiated thyroid cancer (DTC) are mostly based in the experience with papillary histotype and do not consider the differences within the distinct types of DTC, in terms of RAIT uptake and response. OBJECTIVE: This work aims to investigate the association between histology and RAIT avidity and response, and to evaluate whether histotype was an independent prognostic factor in progression-free survival (PFS) and disease-specific survival (DSS) after RAIT for distant metastatic disease. METHODS: A retrospective analysis was conducted of all DTC patients who underwent RAIT for distant metastatic disease, from 2001 to 2018, at a thyroid cancer referral center. We included 126 patients: 42 (33.3%) classical variant papillary thyroid cancer (cvPTC), 45 (35.7%) follicular variant PTC (fvPTC), 17 (13.5%) follicular thyroid cancer (FTC) and 22 (17.5%) Hürthle cell carcinoma. Main outcome measures included RAIT avidity and response. RESULTS: RAIT avidity was independently associated with histology (P < .001) and stimulated thyroglobulin (Tg) at first RAIT for distant lesions (P = .007). Avidity was lowest in HCC (13.6%), intermediate in cvPTC (21.4%), and highest in fvPTC (75.6%) and FTC (76.5%). Regarding RAIT response, HCC and FTC were not different; both showed significantly more often progression after RAIT than fvPTC and cvPTC. Histology influenced PFS (P = .014), but tumor type was not a significant prognostic factor in DSS. Instead, age at diagnosis, resection status, and stimulated Tg at the first RAIT were significantly associated with DSS. CONCLUSION: DTC histotype influenced RAIT avidity and PFS. It is crucial to better detect the metastatic patients that may benefit the most from RAIT.


Subject(s)
Adenocarcinoma, Follicular/pathology , Iodine Radioisotopes/pharmacokinetics , Iodine Radioisotopes/therapeutic use , Adenocarcinoma, Follicular/metabolism , Adenocarcinoma, Follicular/mortality , Adenocarcinoma, Follicular/radiotherapy , Aged , Biological Availability , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/radiotherapy , Carcinoma, Hepatocellular/secondary , Female , Follow-Up Studies , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver Neoplasms/radiotherapy , Liver Neoplasms/secondary , Male , Middle Aged , Neoplasm Metastasis , Portugal/epidemiology , Prognosis , Retrospective Studies , Survival Analysis , Treatment Outcome
7.
Thyroid ; 31(10): 1514-1522, 2021 10.
Article in English | MEDLINE | ID: mdl-34155923

ABSTRACT

Background: Thyrotropin alfa (rhTSH) is not currently approved by the Food and Drug Administration or European Medicines Agency for the preparation of radioactive iodine therapy (RAIT) in patients with distant metastatic papillary thyroid cancer (PTC). There are only a few studies comparing rhTSH with levothyroxine withdrawal (LTW) in this context. Our main aim was to compare the two methods of RAIT preparation in terms of avidity and structural/biochemical response in distant metastatic PTC. We also intended to evaluate whether the two methods of RAIT preparation represented independent prognostic factors for progression-free survival (PFS) and disease-specific survival (DSS) in this subset of patients. Methods: We performed a retrospective analysis of all patients with PTC treated with RAIT for distant metastatic disease between 2006 and 2018. We included 95 PTC patients-27 (28.4%) had LTW and 68 (71.6%) had rhTSH for RAIT. Results: The two groups presented similar clinicopathological characteristics, except for median age at PTC diagnosis, which was higher in the rhTSH group (p = 0.001), but the median age at first RAIT for distant metastatic disease was not different between the two methods of preparation, 63 years old (interquartile range [IQR] 23) in the LTW group versus 70 (IQR 26.75), p = 0.06. Avidity was similar between the two groups (p = 0.973). Median estimate PFS (p = 0.076) and DSS (p = 0.084) were also similar between LTW and rhTSH. Regarding RAIT-related side effects, only 1 (3.7%) patient and 5 (7.4%) patients in the LTW and rhTSH groups, respectively, reported sialadenitis (p = 0.670). Conclusions: There were no differences between the two methods of RAIT preparation regarding avidity and clinical response. rhTSH may be used as an alternative method of preparation for RAIT in patients with known distant lesions, as it presents similar clinical outcomes to LTW and a good safety profile.


Subject(s)
Iodine Radioisotopes/therapeutic use , Neoplasm Metastasis/radiotherapy , Radiopharmaceuticals/therapeutic use , Radiotherapy/methods , Thyroid Cancer, Papillary/radiotherapy , Thyroid Neoplasms/radiotherapy , Thyrotropin Alfa , Thyroxine , Aged , Disease-Free Survival , Female , Humans , Male , Middle Aged , Retrospective Studies , Thyroid Cancer, Papillary/mortality , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/mortality , Thyroid Neoplasms/pathology , Treatment Outcome
8.
Endocrine ; 73(3): 588-597, 2021 09.
Article in English | MEDLINE | ID: mdl-33683595

ABSTRACT

PURPOSE: Germline mutations in DNA repair-related genes have been recently reported in cases with familial non-medullary thyroid carcinoma (FNMTC). A Portuguese family from the Roma ethnic group with four members affected with papillary thyroid carcinoma (PTC), and three members with multinodular goiter (MNG) was identified. The aim of this study was to investigate the involvement of DNA repair-related genes in the etiology of FNMTC in this family and in the Roma ethnic group. METHODS: Ninety-four hereditary cancer predisposition genes were analyzed through next-generation sequencing. Sanger sequencing was used for variant confirmation and screening. Twelve polymorphic markers were genotyped for haplotype analysis in the CHEK2 locus. RESULTS: A germline pathogenic frameshift variant in the CHEK2 gene [c.596dupA, p.(Tyr199Ter)] was detected in homozygosity in the proband (PTC) and in his brother (MNG), being heterozygous in his mother (PTC), two sisters (PTC), and one nephew (MNG). This variant was absent in 100 general population controls. The screening of the CHEK2 variant was extended to other Roma individuals, being detected in 2/33 Roma patients with thyroid cancer, and in 1/15 Roma controls. Haplotype segregation analysis identified a common ancestral core haplotype (Hcac), covering 10 Mb in the CHEK2 locus, shared by affected CHEK2 variant carriers. Analysis of 62 individuals CHEK2 wild-type indicated that none presented the Hcac haplotype. The estimated age for this variant suggested that it was transmitted by a relatively recent common ancestor. CONCLUSIONS: We identified a founder CHEK2 pathogenic variant, which is likely to underlie thyroid cancer and other cancer manifestations in the Roma population.


Subject(s)
Roma , Thyroid Neoplasms , Checkpoint Kinase 2/genetics , Ethnicity , Founder Effect , Genetic Predisposition to Disease , Humans , Male , Pedigree , Portugal , Thyroid Neoplasms/genetics
9.
Endocrine ; 49(1): 204-14, 2015 May.
Article in English | MEDLINE | ID: mdl-25381600

ABSTRACT

The familial forms of non-medullary thyroid carcinoma (FNMTC) represent approximately 5 % of thyroid neoplasms. Nine FNMTC susceptibility loci have been mapped; however, only the DICER1 and SRGAP1 susceptibility genes have been identified. The transcription factors NKX2-1, FOXE1, PAX8, and HHEX are involved in the morphogenesis and differentiation of the thyroid. Recent studies have identified NKX2-1 germline mutations in FNMTC families. However, the role of high-penetrant FOXE1 variants in FNMTC etiology remains unclear. The aim of this study was to investigate the role of FOXE1 germline mutations in the pathogenesis of FNMTC. We searched for molecular changes in the FOXE1 gene in the probands from 60 Portuguese families with FNMTC. In this series, we identified nine polymorphisms and one variant (c.743C>G, p.A248G) which was not previously described. This variant, which involved an amino acid residue conserved in evolution, segregated with disease in one family, and was also detected in an apparently unrelated case of sporadic NMTC. Functional studies were performed using rat normal thyroid cells (PCCL3) clones and human papillary thyroid carcinoma cell line (TPC-1) pools, expressing the wild type and mutant (p.A248G) forms of FOXE1. In these experiments, we observed that the p.A248G variant promoted cell proliferation and migration, suggesting that it may be involved in thyroid tumorigenesis. Additionally, somatic p.V600E BRAF mutations were also detected in the thyroid tumors of two members of the family carrying the p.A248G variant. This study represents the first evidence of involvement of a germline FOXE1 rare variant in FNMTC etiology and suggests that mutations in MAPK pathway-related genes may contribute to tumor development in these familial cases.


Subject(s)
Forkhead Transcription Factors/genetics , Germ-Line Mutation/genetics , Thyroid Neoplasms/genetics , Adult , Aged , Animals , Cells, Cultured , Female , Humans , Male , Middle Aged , Pedigree , Polymorphism, Genetic , Portugal , Rats , Young Adult
10.
J Clin Endocrinol Metab ; 99(3): E497-507, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24423316

ABSTRACT

BACKGROUND: Anaplastic thyroid carcinomas (ATCs) are among the most lethal malignancies, for which there is no effective treatment. OBJECTIVE: In the present study, we aimed to elucidate the molecular alterations contributing to ATC development and to identify novel therapeutic targets. DESIGN: We profiled the global gene expression of five ATCs and validated differentially expressed genes by quantitative RT-PCR in an independent set of tumors. In a series of 26 ATCs, we searched for pathogenic alterations in genes involved in the most deregulated cellular processes, including the hot spot regions of RAS, BRAF, TP53, CTNNB1 (ß-catenin), and PIK3CA genes, and, for the first time, a comprehensive analysis of components involved in the cell cycle [cyclin-dependent kinase (CDK) inhibitors (CDKI): CDKN1A (p21(CIP1)); CDKN1B (p27(KIP1)); CDKN2A (p14(ARF), p16(INK4A)); CDKN2B (p15(INK4B)); CDKN2C (p18(INK4C))], cell adhesion (AXIN1), and proliferation (PTEN). Mutational analysis was also performed in 22 poorly differentiated thyroid carcinomas (PDTCs). RESULTS: Expression profiling revealed that ATCs were characterized by the underexpression of epithelial components and the up regulation of mesenchymal markers and genes from TGF-ß pathway, as well as, the overexpression of cell cycle-related genes. In accordance, the up regulation of the SNAI2 gene, a TGF-ß-responsive mesenchymal factor, was validated. CDKN3, which prevents the G1/S transition, was significantly up regulated in ATCs and PDTCs and aberrantly spliced in ATCs. Mutational analysis showed that most mutations were present in TP53 (42% of ATCs; 27% of PDTCs) or RAS (31% of ATCs; 18% of PDTCs). TP53 and RAS alterations showed evidence of mutual exclusivity (P = .0354). PIK3CA, PTEN, and CDKI mutations were present in 14%-20% of PDTCs, and in 10%-14% of ATCs. BRAF, CTNNB1, and AXIN1 mutations were rarely detected. CONCLUSION: Overall, this study identified crucial roles for TP53, RAS, CDKI, and TGF-ß pathway, which may represent feasible therapeutic targets for ATC and PDTC treatment.


Subject(s)
Cell Cycle/genetics , Genes, p53 , Genes, ras , Mutation , Thyroid Neoplasms/genetics , Cell Line, Tumor , DNA Mutational Analysis , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Microarray Analysis , Thyroid Carcinoma, Anaplastic , Thyroid Neoplasms/pathology
11.
Clin Endocrinol (Oxf) ; 76(1): 33-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21790700

ABSTRACT

OBJECTIVE: Germline mutations in the HRPT2 gene are associated with the hereditary hyperparathyroidism-jaw tumour syndrome (HPT-JT) and a subset of familial isolated hyperparathyroidism (FIHP). Somatic HRPT2 mutations are detected in sporadic parathyroid carcinomas and less frequently in cystic adenomas. The purpose of this study was to investigate the underlying HRPT2 defect in a young patient with symptomatic hyperparathyroidism due to an apparently sporadic parathyroid adenoma with cystic features. DESIGN AND METHODS: HRPT2 mutations in the patient's genomic and parathyroid tumour DNA were screened by PCR-based sequencing. Tumour loss of heterozygosity (LOH) at the HRPT2 locus was assessed with microsatellite markers. A large germline HRPT2 deletion was investigated by real-time quantitative PCR analysis (qPCR). Genomic DNA losses were also appraised by chromosomal comparative genomic hybridization (cCGH). RESULTS: No germline HRPT2 point mutation was detected by direct sequencing. A novel hemizygous HRPT2 somatic mutation (c.32delA) was identified in the tumour. Apparent constitutional homozygosity for HRPT2 flanking microsatellite markers, and absence of LOH at a distal marker, suggested a large germline deletion. Gene dose mapping by qPCR unveiled a de novo deletion of the whole HRPT2 gene and adjacent loci (<9·3 Mb in size). cCGH confirmed germline DNA loss involving the HRPT2 locus. CONCLUSIONS: We report the first large germline deletion of the HRPT2 gene, which was not detectable by conventional PCR-based sequencing methods. This finding emphasizes that qPCR should be implemented in HRPT2 molecular analysis, which may improve genetic assessment and clinical management of patients with FIHP and HPT-JT.


Subject(s)
Gene Deletion , Germ-Line Mutation , Hyperparathyroidism/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Adult , Base Sequence , DNA/genetics , Humans , Male
12.
Endocr Pathol ; 22(1): 44-52, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21360064

ABSTRACT

The diagnosis of parathyroid carcinomas is often difficult. HRPT2 mutations have been identified in familial [hyperparathyroidism-jaw tumor (HPT-JT) syndrome] and sporadic parathyroid carcinomas, supporting that HRPT2 mutations may confer a malignant potential to parathyroid tumors. In this study, we report the clinical, histopathological, and genetic investigation of two unrelated cases, whom had apparently sporadic malignant parathyroid tumors, initially diagnosed as adenomas. In one case, the differential diagnosis was complicated by cervical seeding of parathyroid tumor cells. Genetic studies identified de novo HRPT2 germline mutations in cases 1 (c.518_521delTGTC [p.Ser174LysfsX27]) and 2 (c.226 C > T [p.Arg76X]), unveiling the hereditary HPT-JT syndrome in both patients. Furthermore, the identification of somatic mutations in the patients? parathyroid tumors provided evidence for complete inactivation of the HRPT2 gene, which was consistent with the tumor malignant features. The sensitivity of parafibromin immunostaining to detect HRPT2 mutations was limited. The present data suggests that patients with apparently sporadic parathyroid carcinomas, or parathyroid tumors with atypical histological features, should undergo molecular genetic testing, as it may detect germline HRPT2 mutations. Establishing the diagnosis of hereditary HPT-JT syndrome is relevant for clinical counseling and management of the carriers and their relatives.


Subject(s)
Adenocarcinoma/genetics , Germ-Line Mutation , Parathyroid Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Adenocarcinoma/diagnosis , Adenocarcinoma/therapy , Adult , Combined Modality Therapy , DNA Mutational Analysis , DNA, Neoplasm/analysis , Female , Humans , Parathyroid Neoplasms/diagnosis , Parathyroid Neoplasms/therapy , Treatment Outcome , Young Adult
13.
Endocrine ; 37(3): 408-14, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20960161

ABSTRACT

Patients with pseudohypoparathyroidism type Ib (PHP-Ib) present hypocalcemia and hyperphosphatemia, as a consequence of a resistance to PTH action, through its G-protein-coupled receptor, in the renal tubules. This resistance results from tissue-specific silencing of the G-protein alpha-subunit (G(s)α), due to imprinting disruption of its encoding locus--GNAS. In familial PHP-Ib, maternally inherited deletions at the STX16 gene are associated to a regional GNAS methylation defect. In sporadic PHP-Ib, broad methylation changes at GNAS arise from unknown genetic causes. In this study, we describe the clinical presentation of PHP-Ib in four Portuguese patients (two of whom were siblings), and provide further insight for the management of patients with this disease. The diagnosis of PHP-Ib was made after detection of GNAS imprinting defects in each of the cases. In the siblings, a regional GNAS methylation change resulted from a known 3.0 kb STX16 deletion. In the other two patients, the broad methylation defects at GNAS, which were absent in their relatives, resulted from genetic alterations that remain to be identified. We report the first clinical and genetic study of Portuguese patients with PHP-Ib. The genetic identification of a hereditary form of this rare disease allowed an early diagnosis, and may prevent hypocalcemia-related complications.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/genetics , Pseudohypoparathyroidism/diagnosis , Adult , Child , Chromogranins , Female , Humans , Portugal , Pseudohypoparathyroidism/genetics , Syntaxin 16/genetics , Young Adult
14.
Thyroid ; 17(6): 497-509, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17614769

ABSTRACT

OBJECTIVE: Paired box gene 8/peroxisome proliferator-activated receptor gamma (PAX8PPARgamma) translocation is a molecular event associated with follicular thyroid tumorigenesis and is generated by a chromosomal rearrangement between PAX8 and PPARgamma genes. In this study, we investigated the effects of PAX8PPARgamma fusion protein on cell growth and on thyroid-specific gene expression in immortalized human thyroid cells (Nthy-ori 3-1). METHODS: PAX8PPARgamma-, PAX8-, and thyroid transcription factor-1 (TTF-1)-transfected cell culture models; count of live and dead cells; mRNA analysis by reverse transcription-polymerase chain reaction (RT-PCR) and quantitative RT-PCR; and protein analysis by western blotting and gel shift assays. RESULTS: Cells transfected with the PAX8PPARgamma fusion gene showed higher cell viability at 24, 48, and 72 hours after transfection than cells transfected with control vectors. A PAX8 expression vector increased thyroglobulin (Tg), sodium/iodide symporter (NIS), and thyroid-stimulating hormone (thyrotropin) receptor (TSHR) mRNA levels in a dose-dependent manner. TTF-1 expression vector promoted a significant increase of Tg mRNA level, but had no effect on NIS and TSHR mRNA levels. PAX8PPARgamma transfectants presented a significant decrease in TSHR mRNA level compared to empty vector, but had no effect on Tg and NIS mRNA levels. PAX8 plus PAX8PPARgamma significantly lowered Tg and TSHR mRNA expression levels, but upregulated NIS mRNA level, compared to PAX8 plus control vector. CONCLUSION: The results obtained with this in vitro system demonstrated that PAX8PPARgamma increases thyroid cell viability and has opposite effects on thyroid-specific gene expression, suggesting that the presence of this rearrangement may contribute to the malignant transformation of thyroid follicular cells.


Subject(s)
Oncogene Proteins, Fusion/physiology , PPAR gamma/physiology , Paired Box Transcription Factors/physiology , Thyroid Gland/cytology , Translocation, Genetic/physiology , Cell Line , Cell Survival , Cell Transformation, Neoplastic/genetics , Humans , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Oncogene Proteins, Fusion/genetics , PAX8 Transcription Factor , PPAR gamma/genetics , Paired Box Transcription Factors/genetics , Thyroid Gland/physiology , Thyroid Neoplasms/genetics , Thyroid Nuclear Factor 1 , Transcription Factors/genetics , Transcription Factors/physiology , Transfection
15.
J Endocrinol ; 191(1): 229-38, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17065406

ABSTRACT

The chemokine receptor CCR7 plays a critical role in lymphocyte and dendritic cell trafficking into and within lymph nodes, the preferential metastatic site for papillary (PTC) and medullary (MTC) thyroid carcinomas. In order to determine a possible role for CCR7 in mediating the metastatic behaviour of thyroid carcinomas, we analysed its expression in normal and tumoral thyroid tissues of different histotypes and studied the in vitro effects of its activation by the CCR7 ligand, CCL21. Using real-time quantitative-PCR, we observed that CCR7 expression was higher in PTCs and MTCs than in follicular and poorly differentiated thyroid carcinomas. CCR7 expression was ninefold higher in classic compared with follicular variants of PTCs, and its expression in MTCs was significantly correlated with lymph node metastases. Immunohistochemical staining for CCR7 showed protein expression in neoplastic thyroid cells, with higher intensity in PTCs, MTCs and their lymph node metastases (LNMs). We further showed that CCL21 stimulation of a CCR7-expressing thyroid tumour cell line (TPC-1) promotes cell proliferation and migration, and the chemotactic effect of CCL21 in these cells involves actin polymerization, increased beta1-integrin expression and increased matrix metalloproteinase secretion. Taken together, our results demonstrate that CCR7 activation on thyroid carcinoma cells by CCL21 - a chemokine abundantly expressed in lymph nodes - favours tissue invasion and cell proliferation, and therefore may promote thyroid carcinoma growth and LNM.


Subject(s)
Carcinoma, Papillary/metabolism , Lymph Nodes/metabolism , Receptors, Chemokine/metabolism , Thyroid Neoplasms/metabolism , Actins/analysis , Carcinoma, Papillary, Follicular/metabolism , Cell Line, Tumor , Cell Membrane/chemistry , Cell Movement/drug effects , Chemokine CCL21 , Chemokines, CC/pharmacology , Flow Cytometry , Humans , Immunohistochemistry/methods , Integrin beta1/analysis , Lymphatic Metastasis , Matrix Metalloproteinases/analysis , Receptors, CCR7 , Receptors, Chemokine/analysis , Receptors, Chemokine/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Gland/chemistry , Thyroid Gland/metabolism
16.
Eur J Endocrinol ; 153(5): 701-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16260429

ABSTRACT

OBJECTIVE: The aim of this study was to clarify the role of vascular endothelial growth factor (VEGF) and VEGF receptor (VEGFR) pathways in thyroid tumourigenesis. METHODS: We examined VEGF, VEGFR-1 and VEGFR-2 expression on 34 papillary thyroid carcinomas (PTCs), 18 follicular thyroid carcinomas (FTCs), eight poorly differentiated thyroid carcinomas (PDTCs) and on a thyroid tumour-derived cell line (NPA'87) by immunohistochemistry, reverse transcriptase PCR, immunofluorescence and Western blotting. RESULTS: We have demonstrated that VEGF expression was significantly (P < 0.05) more prevalent in PTCs (79%) than in FTCs (50%) or PDTCs (37%). Similarly, 76% of PTCs, 83% of FTCs and 25% of PDTCs expressed VEGFR-1, whereas 68% of PTCs, 56% of FTCs and 37% of PDTCs expressed VEGFR-2. Coexpression of VEGF and its receptors was observed in 50% of PTCs, 39% of FTCs and 12% of PDTCs, raising the possibility that VEGF may signal in an autocrine loop in these neoplasias, as observed previously for other types of cancer. In agreement with the idea that autocrine VEGF signalling plays an important role in thyroid carcinogenesis, the blockade of either VEGF or its receptors with neutralizing antibodies significantly reduced cell viability and increased apoptosis levels of the VEGFR-positive thyroid tumour cell line NPA'87. CONCLUSIONS: Our results highlight a previously undefined VEGF autocrine action in thyroid carcinomas which could play a crucial role in tumour cell survival and could represent a useful therapeutic target for thyroid tumours.


Subject(s)
Adenocarcinoma, Follicular/metabolism , Autocrine Communication , Receptors, Vascular Endothelial Growth Factor/metabolism , Thyroid Neoplasms/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adenocarcinoma, Follicular/genetics , Adenocarcinoma, Follicular/pathology , Adenocarcinoma, Follicular/physiopathology , Apoptosis , Cell Line , Cell Survival , Genetic Testing , Humans , Mutation , Proto-Oncogene Proteins B-raf/genetics , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...