• Non ci sono risultati.

ABNORMALITIES OF NUCLEAR RECEPTORS IN THYROID CANCER

N/A
N/A
Protected

Academic year: 2021

Condividi "ABNORMALITIES OF NUCLEAR RECEPTORS IN THYROID CANCER"

Copied!
14
0
0

Testo completo

(1)

SHEUE-YANN CHENG, P

H

.D.

Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-4264

INTRODUCTION

Nuclear receptors comprise a large family of ligand-inducible transcription factors that are critically important for growth, differentiation, development, and maintenance of metabolic homeostasis. They regulate the expression of target genes by binding to the specific DNA sequences at the promoters to mediate the biological effects. Many nuclear receptors have multiple isoforms with over-lapping functions or isoform- specific functions (1, 2). The expression of these receptor isoforms is regulated in a tissue- and development-dependent manner. A host of coregulatory proteins that influence the ligand selectivity and DNA binding capacity further modulates the tran- scriptional activities (3, 4).

Abnormal expression and/or aberrant functions of sex steroid nuclear receptors

are known to be involved in the development and progression of such endocrine

cancers as breast, ovarian, endometrium, and prostate, but less is known about the

role of nuclear receptors in the carcinogenesis of the thyroid. Progress in this area

has recently been made as a result of the discoveries of the fusion gene of PAX8 with

the peroxisome proliferator-activated receptor in follicular

thyroid carcinoma and of the spontaneous development of follicular thyroid carcinoma

in the homozygous knock-in mutant mice harboring a mutated thyroid hormone

receptor This review will first examine the latest findings on the possible roles

of several sex steroid nuclear receptors in thyroid carcinogenesis. It will then discuss

the molecular actions of the mutant in carcinogenesis, particularly in relation to

a unique knock-in mouse model of thyroid cancer.

(2)

ABNORMAL EXPRESSION OF ESTROGEN AND PROGESTERONE RECEPTORS IN THYROID CANCER

Thyroid carcinoma is more common in women than in men (5). For 2003, the esti- mate of new cases of thyroid cancer has a female predominance with a 2.9:1 ratio (6).

This predominance suggests that estrogens may play a critical role in the development of thyroid carcinoma. In the past two decades, efforts have been made to demonstrate the presence of estrogen receptors (ERs) in thyroid tumors and to correlate tumor malignancy with ER expression. Using a dextran-coated charcoal method and analysis by the method of Scatchard, Miki et al. did not detect ER in the cytosol of normal thyroids, but they found a significantly higher ER in the neoplastic and hyperplastic thyroid tissues (7). Using different biochemical methods, Mizukami et al. (8) and Yane et al. (9) also showed a higher expression of ER in neoplastic thyroid lesions than in normal thyroids or in adjacent normal tissues. Lewy-Trenda examined 72 thyroid glands for the expression of ER by using immunochemical assays with anti-ER anti- bodies. Positive staining occurred in the nuclei of differentiated thyroid cancer cells (24%), but not in non-neoplastic cells. A small number of oxyphillic (4%) and follicular adenomas (6%) also stained positive for ERs (10).

Consistent with these findings, several studies showed that estrogens stimulate the proliferation of thyroid carcinoma cells (11–13), whereas the antiestrogen, tamoxifen, inhibits the proliferation of a tumor cell line derived from medullary thyroid carci- noma (11). These studies clearly showed that cell proliferation induced by estrogens is mediated by ERs, but little is known about the specific molecular pathways. One study suggested that activation of the mitogen-activated protein kinase by phosphorylation might be one of the key steps in the estrogen-mediated cell proliferation of thyroid cancer cells (13).

The relevance of the increased expression of ERs in thyroid tumorigenesis is not obvious, particularly given that there is no clear correlation in the extent of expression of ER to age, sex, presenting clinical or pathological features, or, in cases of carcinoma, to subsequent metastatic potential (10, 14, 15). Furthermore, the failure of several stud- ies to detect a greater expression of ERs in thyroid tumors than in normal tissues casts further doubt on the significance of expression of ERs in thyroid tumor development and progression (16–19). It is unclear whether the discrepancy among studies is due to the sensitivity of the detection or the intrinsic variability in the expression of ERs in tumor samples. Plainly, more studies are needed to understand whether estrogens and ERs are the major factors that contribute to thyroid cancers predominance in females.

Fewer studies have investigated the roles of progesterone receptors (PRs) in thyroid

carcinogenesis. Because of the interest in understanding thyroid cancer’s predominance

in females, the expression of PRs in thyroid tumors has been evaluated by means

of ligand binding assays, enzyme immunoassays, and/or immunohistochemistry. In

a few limited studies, the presence of PR and ER was assessed concurrently in the

same samples. In 135 thyroid lesions that included papillary, follicular, medullary, and

Hurthle cell carcinomas, van Hoeven detected the presence of PR in 51% of the cases,

with the highest abundance in papillary carcinomas, particularly in male patients and

women older than 50 years (15). In that same study, ER was found in 46% of the

(3)

samples. In other studies, however, a higher frequency of ER than PR was found in papillary carcinomas (7, 10, 14). Similar to the findings for ER, no correlations were observed between the expression of PR and age, sex, tumor size, presence of capsular or vascular invasion, or lymph node status (10, 14). Still, how the expression of PR is involved in the development of thyroid cancer has not been assessed.

ALTERED EXPRESSION OF THE RETINOIC ACID RECEPTORS IN THYROID CANCER

Retinoic acids (RAs) are essential for many biological processes including prolifer- ation, development, differentiation, carcinogenesis, and apoptosis. These biological effects are mediated through their receptors (RARs). The retinoids, both the natural and synthetic analogs, have been shown to be effective in preventing several cancers in experimental animals and in reversing pre-neoplastic lesions in humans (20, 21).

Whether the retinoids could be effective in re-differentiating thyroid cancer cells to be amenable to radioiodide or TSH-suppressive T4 therapy has prompted several investi- gators to study the expression of RAR in cancer cell lines and tissues. Using Northern blot analysis, del Senno found that the expression of was lower in thyroid carcinoma cells than in normal thyroid follicular cells. Moreover, del Senno demonstrated that RA reduces the proliferation and function of thyroid follicular cells (22, 23). These findings were confirmed in a larger study. Using immunohistochem- istry and Western blotting, Rochaix et al. compared the expression of in 40 normal/benign tissues, 16 papillary carcinomas, and two follicular carcinomas, immunostaining was detected in the nuclei, but was limited to the normal epithelial thyroid tissue. A dramatic decrease in immunostaining was observed in all 16 papillary carcinomas, but in only one follicular carcinoma (24).

Because the feasibility of retinoid-induced differentiation therapy in thyroid cancer

hinges on functional RARs, Schmutzler et al. not only examined the expression of

mRNA in several human thyroid carcinoma cell lines and tissues, but also assessed

the ligand and DNA binding activities (25). Functional RARs were clearly detectable

in the two human thyroid carcinoma cell lines (FTC-133 and FTC-238) and two

anaplastic thyroid carcinoma cell lines (HTH74 and C643). Intriguingly, variable lev-

els of mRNA were observed in these cell lines, an observation probably indicative

of dysregulation of receptor expression in thyroid cancer (25). These results suggest

the heterogeneity in the expression of RARs and the association of the dysregula-

tion of the expression of RAR with thyroid carcinogenesis. However, the available

expressed functional RAR seems to be able to respond to RA treatment. In a pilot

study, patients with advanced thyroid cancer and without the therapeutic options of

operation or radioiodide therapy were treated with 13-cis-retinoic acid (1.5 mg/kg

body weight daily for 5 weeks). Overall, tumor regression was observed in 19 patients

(38%). However, response to retinoid therapy did not always correlate with increased

radioiodine uptake (a re-differentiation marker), and so other direct antiproliferative

effects could also be involved (26). These encouraging clinical findings warrant addi-

tional studies on the RA-based treatment of thyroid cancer. At present, however, little

is known about either the molecular mechanisms by which the expression of RAR

(4)

is dysregulated during thyroid carcinogenesis or the RA-induced–redifferentiation of follicular cells. Elucidation of these mechanisms should help in the design of an effective treatment of thyroid carcinomas that uses the retinoids.

ABNORMALITIES OF THYROID HORMONE RECEPTORS IN THYROID CANCER The thyroid hormone receptors (TRs) mediate the pleiotropic activities of the thy- roid hormone (T3) in growth, development, and differentiation and in maintaining metabolic homeostasis. The two TR genes, and are located on human chromo- somes 17 and 3, respectively. Alternative splicing of the primary transcripts gives rise

to five major TR isoforms and and

differ in their lengths and amino acid sequences at the amino terminal A/B domain, but they bind T3 with high affinity to mediate gene regulatory activity. By contrast, which differs from the other TR isoforms in the C-terminus, does not bind T3, and its precise functions have yet to be elucidated. The expression of TR isoforms is tissue-dependent and developmentally regulated (1, 2).

Early evidence to suggest that mutated TR could be involved in carcinogenesis came from the discovery that is the cellular counterpart of the retroviral v-erbA that is involved in the neoplastic transformation leading to acute erythroleukemia and sarcomas (27, 28). The oncogenic role of v-erbA was subsequently demonstrated in mammals in that male transgenic mice overexpressing v-erbA developed hepatocellular carcinoma (29).

In recent years, increasing evidence suggests that aberrant expression and mutation of the TR genes could be associated with human neoplasias. Somatic point mutations of and were found in 65% (11/17 tumors) and 76% (13/17 tumors), respectively, of human hepatocellular carcinomas. Many of these mutated TRs have lost T3-binding activity and exhibit aberrant DNA-binding activity (30). Aberrant expression and mutations of TR genes were also found in renal clear cell carcinomas (31). Cloning of TRs from 22 renal clear cell carcinomas and 20 surrounding normal tissues identified somatic mutations in 32% and 14% of cloned and cDNAs, respectively (32). Most of the mutations were localized in the hormone- binding domain that leads to loss of T3-binding activity and/or impairment in binding to TREs. Similar to the mutated TRs detected in hepatocellular carcinoma (30, 33), the mutated TRs identified in renal clear cell carcinomas exhibit dominant negative activity (32). These studies suggest that mutated TR plays an important role in the development of these human cancers.

Abnormal expression and somatic mutations of TRs in thyroid cancer

Similar to the expression levels reported for ER, PR, and RAR, an altered expression of TRs was detected in thyroid carcinomas. Comparison of the mRNA expression levels of TR isoforms in normal, hyperplastic, and neoplastic human thyroid tissues indicated that is significantly lower in papillary and follicular carcinomas than it is in normal thyroid. No differences, however, were found in the expression levels of and These findings suggest an association of the reduced expression of with the development of thyroid carcinomas.

(34, 35).

(5)

These studies, however, did not determine whether and were altered at the protein level.

In addition to the reduced expression of a lower expression of was found in 16 papillary thyroid carcinomas from Polish patients.

The and protein levels, however, were higher in cancerous tissues than in nearby healthy tissues, an indication of the complexity in the regulation of TR expression in these tumors (36). To understand the nature of TRs in these papillary thyroid carcinomas, cDNAs were cloned concurrently from both the tumor lesions and the healthy thyroids as controls. Sequence analyses indicated that 93.8% and 62.5%

of papillary thyroid carcinomas had mutations in and respectively. In contrast, no mutations were found in healthy thyroid controls, and only 11.1% and 22.2% of thyroid adenomas had mutations in and respectively. Func- tional analysis indicated that these mutated TRs lose their transactivation function and exhibit dominant negative activity (36).

The reduced expression of in papillary thyroid carcinomas was fur- ther confirmed in a more recent study of 16 Japanese patients (37). In contrast to the Polish patients, no amino acid-substitution-mutations were detected in the

cloned from these papillary thyroid carcinomas. The reasons for the different propensity in the mutations of the gene in these two groups of patients are not entirely clear. One possibility is that the Polish patients were from the post-Chernobyl population and that radiation exposure is a contributing factor to the high frequency of TR mutations. Indeed, five of the 16 Polish patients with mutated TRs were in their teens when the Chernobyl accident occurred. One of 16 patients received radi- ation treatment during her childhood because of another disease. The age of other patients ranged from 32–58 years old at the time of the Chernobyl accident (Monika Puzianowska-Kuznicka; personal communication). The validation of this hypothesis would require a cohort study with a larger number of patients and a detailed knowledge of irradiation dose received by the patients.

Another possibility is that the propensity of mutations of in papillary thyroid carcinomas could be affected by the patient’s ethnic origin. Genetic variation between different populations occurs frequently. For example, a wide variation in the frequency of RET/PTC rearrangements, a hallmark of papillary thyroid carcinoma, has been reported, ranging from a few percent in Japanese (38) and Saudi Arabian patients (39), to 18.8% in Italian patients (40), to 70% in New Caledonian and 85% in Australian patients (41). The frequency of polymorphisms associated with thyroid diseases also differs in Japanese and Caucasian populations (42). Clarification of the issue of whether genetic background affects the frequency of mutations in papillary thyroid carcinoma awaits additional analyses in patients with different ethnic origins.

Germline mutations of the gene in thyroid cancer: lessons learned from a unique mouse model of thyroid carcinogenesis

So far, the TR mutants identified in human cancers including thyroid carcinoma

are somatic mutations. A knock-in mouse that harbors a gerrnline mutation of the

gene has been created (43). The mutation was targeted to the gene locus

(6)

via homologous recombination and the Cre/loxP system. The mutation is called PV mouse) after a patient with the mutation who suffers from the disease known as resistance to thyroid hormone (RTH) (44, 45). RTH is a syndrome characterized by the elevated levels of circulating thyroid hormone that are associated with non- suppressible TSH. Some of the clinical features include attention-deficit hyperactivity disorder, mental retardation, short stature, decreased -weight, tachycardia, and hear- ing abnormalities (44, 45). PV has a unique mutation in exon 10, a C-insertion at codon 448, which produces a frame shift of the carboxyl-terminal 14 amino acids of In vitro studies revealed that PV has completely lost T3-binding activity, lacks transcriptional capacity, and exhibits potent dominant negative activity (46). Exten- sive characterization of the phenotype indicates that the mouse faithfully reproduces the human RTH (43). This mouse provides a valuable model for clarifying the role of germline mutations of the gene in carcinogenesis.

In addition to the phenotypes of RTH, homozygous mice exhibited the phenotype of age-dependent increased mortality. By the age of about 10 months, 50% had died, and by the age of 14–15 months, all mice were dead. In contrast, the heterozygous mice did not exhibit such abnormalities. Morphological examinations of the moribund mice indicate that as these mice aged, they spontaneously developed thyroid carcinoma (47). Histological evaluation of thyroids of 27 moribund mice showed capsular invasion (85%), vascular invasion (74%), anaplasia (37%), and metastasis to the lung and heart (26%) but not to lymph nodes (Table 1).

Representative examples of the pathological features of capsular invasion (Panel A), vascular invasion (Panel B), anaplasia (Panel C), and metastasis to the lung (Panel D) are shown in Figure 1. The histological features and the metastatic patterns indicate that the thyroid carcinoma developed in mice is follicular. Thus

mice provide the first animal model for studying the molecular genetics underlying follicular thyroid carcinogenesis.

Using microarrays consisting of 20,000 mouse cDNAs, Ying et al. recently pro- filed the global alterations in gene expression in the thyroids of mice at 6 months of age, at which time metastasis had begun (48). They found that 185 genes were up-regulated (2- to 17-fold) and 92 were down-regulated (2- to 20-fold).

The majority (~60%) of these altered genes are unnamed. Functional clustering of

named genes with reported functions (100 genes) indicated that ~39% were tumor-,

metastasis/invasion-, and cell cycle-related. Importantly, several tumor-related genes,

such as cyclin D1, pituitary tumor transforming gene-1, cathespin D, and transform-

ing growth factor that have been reported to be over-expressed in human thyroid

(7)

Figure 1. Pathological features in thyroid glands and metastasis in the lung of Histologic sections from tissues of showed evidence of capsular invasion in thyroid (A) (arrows), vascular invsion in thyroid (B)(arrows), anaplasia in thyroid (C) and metastatic thyroid carcinoma lesions in lung (arrow).

cancers were found to be activated in the arrays (49–53). Analyses of the gene pro- files suggested that the signaling pathways mediated by TSH, peptide growth factors, transforming growth tumor necrosis and nuclear factor were activated, whereas pathways mediated by peroxisome proliferator-activated receptor

were repressed (48). These findings suggest that the expression of the mutant directly and indirectly alters multiple signaling pathways that could contribute to the development of thyroid cancer and that thyroid carcinogenesis is mediated by multiple genetic events.

The frequent occurrence of the somatic mutations in several human cancers (30, 32, 36, 54, 55) and the development of follicular thyroid carcinoma in

(47) raise the question of whether PV could function to initiate carcinogenesis. On the

basis of observations that but not develop follicular thyroid

carcinoma, it is unlikely that PV could act alone to initiate thyroid carcinogenesis. One

of the significant differences in phenotypes between and mice is

that the circulating serum TSH concentration in is ~275-fold higher

than that in (43). TSH is the main regulator of thyrocyte differentiation

and proliferation, and the possibility that it is an initiator of thyroid carcinogenesis has

been intensively studied (56, 57). Recent clinical and biochemical studies, however, do

not support the role of TSH as an initiator of follicular carcinoma (58, 57). Additional

genetic changes need to occur for the transformation of the hyperproliferative thyroid

cells to cancer cells. On the basis of these considerations, it is reasonable to propose

(8)

that mutation of the two alleles of the gene could be one of the genetic changes leading to the transformation of the hyperproliferative thyroid cells to cancer cells.

This hypothesis needs to be tested in future studies.

ABNORMALITIES OF PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR IN THYROID CANCER

is a nuclear receptor that is involved in a wide range of cellular processes including adipogenesis, inflammation, atherosclerosis, cell cycle control, apoptosis, and carcinogenesis (59, 60). is abundantly expressed in adipose tissue, large intestine, and hematopoietic cells, and it is moderately expressed in kidney, liver, and small intestine (61). It was recently found also to express in the thyroid (48).

inhibits cell growth, and one of the mechanisms in inhibition of cell proliferation is by reducing E2F/DP DNA-binding and transcriptional activity (62). Consistently, activation of signaling by its ligands has been shown to block cell proliferation of various malignant cells and, in some cases, to induce differentiation and apoptosis (63–68). Ohta et al. reported that is expressed in human papillary thyroid carcinoma cell lines (69). Significant, but variable expression of

was detected in four of the six cell lines studied. Consistent with findings in other cancer cell lines (63–68), cell proliferation was inhibited and apoptosis was induced by treatment with troglitazone. Ohta et al. also found that troglitazone significantly reduced tumor growth and prevented distant metastasis of BHP18–21 tumors in nude mice in vivo (69). In a more recent study, Martelli et al. also evaluated whether is involved in the growth regulation of normal and tumor thyroid cells (70). No mutations were detected in exons 3 and 5 in human thyroid carcinoma cell lines and tissues. The growth of thyroid carcinoma cells was inhibited by treatment with agonists, but no growth inhibitory effect was observed in NPA cells by agonists that did not express Growth inhibition induced by agonists or by overexpression of the gene in thyroid carcinoma cells was associated with increased p27 protein levels and apoptotic cell death (70).

provide an unprecedented opportunity to study the role of in thyroid carcinogenesis in vivo. Using quantitative real-time PCR and Northern blotting, Yin et al. found that the expression of was repressed 50%–

60% in the thyroids of at the ages of 4, 6, and 12 months (71).

Immunohistologic analysis demonstrated that the expression of protein in the primary lesions of was less than that in the thyroids of wild-type mice and was not detectable in the metastasis in the lung (unpublished results), an indication that the expression of protein remained low during thyroid carcinogenesis.

Moreover, PV was found to abolish ligand (troglitazone)-dependent transcriptional activity of in primary cultured thyroid cells from wild-type mice (71). The PV-induced transcriptional repression could be due to PV’s competition with

for binding to the peroxisome proliferator-activated receptor response element (PPRE)

present in the downstream target genes. Indeed, gel shift assay showed that

the in vitro translated PV protein could bind to PPRE. This notion is supported by the

(9)

Figure 2. Camparison of the expression of and lipoprotein (LpL) mRNA in the thyroids of and wild-type mice at different ages by quantitative real-time PCR. Relative expression levels of and LpL(B) mRNA in the thyroid glands were determined using age matched wild-type and mutant mice at the ages of 4 and 12 months as marked. The data are expressed as

finding that the lipoprotein lipase (LpL) gene, a known downstream target gene (72), was repressed ~5-fold, as shown by cDNA microarrays (48). Subsequent analyses by quantitative real-time PCR further demonstrated that the expression of the LpL gene was down-regulated (Panel B; Figure 2) concurrently with

(Panel A; Figure 2) in the thyroid glands of mice at the ages of 6 and 12 months, thus confirming the repression of signal pathways during thyroid carcinogenesis (71). These results indicate that reduced expression of

and repression of its transcriptional activity are associated with thyroid carcinogenesis

mean

(10)

and raise the possibility that can be tested as a potential molecular target for prevention and treatment of follicular thyroid carcinoma.

That the attenuation of the signaling pathways is associated with the devel- opment and progression of follicular thyroid carcinoma is also supported by the findings that the rearrangement occurs frequently in human follicular thyroid carcinomas, less frequently in adenomas, but not at all in papillary thyroid carcinomas (73–76). Even though the molecular actions of the rearrangement, particularly in its relation to the thyroid follicular carcinoma, has yet to be clarified, it is known that the fusion of PAX8, a thyroid transcription factor, to the amino ter- minus of results in the loss of the transcriptional activity of (73).

Moreover, protein acts to inhibit thiazolidinedione-induced transacti- vation by in a dominant negative manner (73). Taken together, these studies suggest that suppression of signaling is closely linked to the development and progression of follicular thyroid carcinoma.

CONCLUDING REMARKS

Studies in the past few decades have clearly established that nuclear receptors play significant roles in the development and progression of several endocrine tumors, such as breast and prostate cancers. Progress in understanding the role of nuclear receptors in thyroid carcinoma lags behind that in breast and prostate cancers. Studies so far indicate that altered expression of ER, PR, RAR, TR, or is associated with thyroid carcinomas. More studies are warranted to clarify the functional consequences of altered expressed receptors and to elucidate their signaling pathways in relation to carcinogenesis of the thyroid. These efforts will not only advance our understanding of the molecular genetics of thyroid cancer, but also provide opportunities to develop novel strategies for prevention and treatment.

The discovery that mice spontaneously develop follicular thyroid carci- noma indicates that, in addition to altered expression of nuclear receptors, mutation of nuclear receptors is another abnormality that could contribute to thyroid carcino- genesis. It is currently unknown whether, in addition to the gene, mutations of other nuclear receptors could also contribute to the development and progression of thyroid cancer. The finding that the mouse can be used as an animal model of follicular thyroid carcinoma opens the door to further study of the molecular genetic events underlying carcinogenesis, to identifying signature genes during differ- ent stages of tumor progression for clinical diagnosis, and to the testing of drugs and other treatment modalities.

ACKNOWLEDGMENTS

The author thanks all of her colleagues and collaborators who have contributed to the work described in this review.

REFERENCES

1. Cheng, S.-y. Multiple mechanisms for regulation of the transcriptional activity of thyroid hormone

receptors. Rev Endocr Metab Disord 1: 9–18, 2000.

(11)

2.

3.

4.

5.

6.

7.

8.

9.

10.

11.

12.

13.

14.

15.

16.

17.

18.

19.

20.

21.

22.

23.

24.

25.

26.

Yen, P. M. Physiological and molecular basis of thyroid hormone action. Physiol Rev 81: 1097–1142, 2001.

McKenna, N. J., Lanz, R. B., and O’Malley, B. W. Nuclear receptor coregulators: cellular and molecular biology. Endocrinol Rev, 20: 321–344, 1999.

Hermanson, O., Glass, C. K., and Rosenfeld, M. G. Nuclear receptor coregulators: multiple modes of modification. Trends Endocrinol Metab, 13: 55–60, 2002.

Levi, F, Franceschi, S., Gulie, C., Negri, E., and La Vecchia, C. Female thyroid cancer: the role of reproductive and hormonal factors in Switzerland. Oncology, 50: 309–315, 1993.

Jemal, A., Murray, T., Samuels, A., Ghafoor, A., Ward, E., and Thun, M.J. Cancer statistics. CA Cancer J Clin, 53: 5–26, 2003.

Miki, H., Oshimo, K., Inoue, H., Morimoto, T., and Monden, Y. Sex hormone receptors in human thyroid tissues. Cancer, 66: 1759–1762, 1990.

Mizukami, Y, Michigishi, T., Nonomura, A., Hashimoto, T., Noguchi, M., and Matsubara, F. Estrogen and estrogen receptors in thyroid carcinomas. J Surg Oncol, 47: 165–169, 1991.

Yane, K., Kitahori, Y, Konishi, N., Okaichi, K., Ohnishi, T., Miyahara, H., Matsunaga, T., Lin, J. C., and Hiasa,Y. Expression of the estrogen receptor in human thyroid neoplasms. Cancer Lett, 84: 59–66, 1994.

Lewy-Trenda, I. Estrogen and progesterone receptors in neoplastic and non-neoplastic thyroid lesions.

Pol J Pathol, 53: 67–72, 2002.

Yang, K., Pearson, C. E., and Samaan, N. A. Estrogen receptor and hormone responsiveness of medullary thyroid carcinoma cells in continuous culture. Cancer Res 48: 2760–2763, 1988.

Banu, K.S., Govindarajulu, P., and Aruldhas, M. M. Testosterone and estradiol have specific differential modulatory effect on the proliferation of human thyroid papillary and follicular carcinoma cell lines independent of TSH action. Endocr Pathol, 12: 315–327, 2001.

Manole, D., Schildknecht, B., Gosnell, B., Adams, E., and Derwahl, M. Estrogen promotes growth of human thyroid tumor cells by different molecular mechanisms. J Clin Endocrinol Metab, 86: 1072–

1077, 2001.

Bur, M., Shiraki, W., and Masood, S. Estrogen and progesterone receptor detection in neoplastic and non-neoplastic thyroid tissues. Mod Pathol, 6: 469–472, 1993.

van Hoeven, K. H., Menendez-Botet, C. J., Strong, E. W, and Huvos, A. G. Estrogen and progesterone receptor content in human thyroid disease. Am J Clin Pathol, 99: 175–181, 1993.

Colomer, A., Martinez-Mas, J. V., Matias-Guiu, X., Llorens, A., Cabezas, R., Prat, J., and Garcia- Ameijeiras, A. Sex-steroid hormone receptors in human medullary thyroid carcinoma. Mod Pathol, 9:

68–72, 1996.

Bonacci, R., Pinchera, A., Fierabracci, P., Gigliotti, A., Grasso, L., and Giani, C. Relevance of estrogen and progesterone receptors enzyme immunoassay in malignant, benign and surrounding normal thyroid tissue. J Endocrinol Invest, 19: 159–164, 1996.

Metaye, T., Millet, C., Kraimps, J. L., Aubouin, B., Barbier, J., and Begon, F. Estrogen receptors and cathepsin D in human thyroid tissue. Cancer, 72: 1991–1996, 1993.

Jaklic, B. R., Rushin, J., and Ghosh, B. C. Estrogen and progesterone receptors in thyroid lesions. Ann Surg Oncol, 2: 429–434, 1995.

Verma, A. K. Retinoids in chemoprevention of cancer. J Biol Regul Homeost Agents, 17: 92–97, 2003.

Ralhan, R., and Kaur, J. Retinoids as chemopreventive agents. J Biol Regul Homeost Agents, 17:

66–91, 2003.

del Senno, L., Rossi, R., Gandini, D., Piva, R., Franceschetti, P., and degli Uberti, E. C. Retinoic acid-induced decrease of DNA synthesis and peroxidase mRNA levels in human thyroid cells expressing retinoic acid receptor alpha mRNA. Life Sci, 53: 1039–1048, 1993.

del Senno, L., Rossi, R., Franceschetti, P., delgi Uberti, E. C. Expression of all-trans-retinoic acid receptor RNA in human thyroid cells. Biochem Mol Biol Int, 33: 1107–1115, 1994.

Rochaix, P., Monteil-Onteniente, S., Rochette-Egly, C., Caratero, C., Voigt, J. J., and Jozan, S. Reduced expression of retinoic acid receptor beta protein (RAR beta) in human papillary thyroid carcinoma:

immunohistochemical and western blot study. Histopathology, 33: 337–343, 1998.

Schmutzler, C., Brtko, J., Winzer, R., Jakobs, T. C., Meissner-Weigl, J., Simon, D., Goretzki, P. E., and Kohrle, J. Functional retinoid and thyroid hormone receptors in human thyroid-carcinoma cell lines and tissues. Int J Cancer, 76: 368–376, 1998.

Simon, D., Korber, C., Krausch, M., Segering,J., Groth, P., Gorges, R., Grunwald, F., Muller-Gartner,

H. W., Schmutzler, C., Kohrle, J., Roher, H. D., and Reiners, C. Clinical impact of retinoids in

(12)

redifferentiation therapy of advanced thyroid cancer: final results of a pilot study. Eur J Nucl Med Mol Imaging, 29: 775–782, 2002.

Sap, J., Munoz, A., Damm, K., Goldberg, Y., Ghysdael, J., Leutz, A., Beug, H., and Vennstrom, B. The c-erb-A protein is a high-affinity receptor for thyroid hormone. Nature, 324: 635–640, 1986.

Thormeyer, D. and Baniahmad, A. The v-erbA oncogene. Int J Mol Med, 4: 351–358, 1999.

Barlow, C., Meister, B., Lardelli, M., Lendahl, U., and Vennstrom, B. Thyroid abnormalities and hepatocellular carcinoma in mice transgenic for v-erbA. EMBO J, 13: 4241–4250, 1994.

Lin, K. H., Shieh, H. Y., Chen, S. L., and Hsu, H. C. Expression of mutant thyroid hormone nuclear receptors in human hepatocellular carcinoma cells. Mol Carcinog, 26: 53–61, 1999.

Puzianowska-Kuznicka, M., Nauman, A., Madej, A., Tanski, Z., Cheng, S., Nauman, J. Expression of thyroid hormone receptors is disturbed in human renal clear cell carcinoma. Cancer Lett, 155: 145–152, 2000.

Kamiya, Y., Puzianowska-Kuznicka, M., McPhie, P., Nauman, J., Cheng, S.Y., and Nauman, A. Expres- sion of mutant thyroid hormone nuclear receptors is associated with human renal clear cell carcinoma.

Carcinogenesis, 23: 25–33, 2002.

Lin, K. H., Zhu, X. G., Shieh, H. Y, Hsu, H. C., Chen, S. T, McPhie, P., and Cheng, S. Y. Identification of naturally occurring dominant negative mutants of thyroid hormone alpha 1 and beta 1 receptors in a human hepatocellular carcinoma cell line. Endocrinology 137: 4073–4081, 1996.

Wallin, G., Bronnegard, M., Grimelius, L., McGuire, J., and Torring, O. Expression of the thyroid hormone receptor, the oncogenes c-myc and H-ras, and the 90 kD heat shock protein in normal, hyperplastic, and neoplastic human thyroid tissue. Thyroid, 2: 307–313, 1992.

Bronnegard, M., Torring, O., Boos, J., Sylven, C., Marcus, C., and Wallin, G., 1994. Expression of thyrotropin receptor and thyroid hormone receptor messenger ribonucleic acid in normal, hyperplastic, and neoplastic human thyroid tissue. J. Clin Endocrinol Metab, 79: 384–389, 1994.

Puzianowska-Kuznick, M., Krystyniak, A., Madej, A., Cheng, S.-y., and Nauman, J. Contribution of functionally impaired thyroid hormone receptor mutants to the tumorigenesis of thyroid papillary cancer. J Clin Endocrinol Metab, 87: 1120–1128, 2002.

Takano, T., Miyauchi, A., Yoshida, H., Nakata, Y., Kuma, K., Amino, N. Expression of TRbeta 1 mRNAs with functionally impaired mutations is rare in thyroid papillary carcinoma. J Clin Endocrinol Metab 88: 3447–3449, 2003.

Namba, H., Yamashita, S., Pei, H. C., Ishikawa, N., Villadolid, M. C., Tominaga, T, Kimura, H., Tsuruta, M., Yokoyama, N., and Izumi, M. Lack of PTC gene (ret proto-oncogene rearrangement) in human thyroid tumors. Endocrinol Jpn, 38: 627–632, 1991.

Zou, M., Shi, Y., and Farid, N. R. Low rate of ret proto-oncogene activation (PTC/retTPC) in papillary Cinti, R., Yin, L.,Ilc, K., Berger, N., Basolo, F., Cuccato, S., Giannini, R., Torre, G., Miccoli, P., Amati, P., Romeo, G., and Corvi, R. RET rearrangements in papillary thyroid carcinomas and adenomas detected by interphase FISH. Cytogenet Cell Genet, 88: 56–61, 2000.

Chua, E. L., Wu, W. M., Tran, K. T, McCarthy, S. W., Lauer, C. S., Dubourdieu, D., Packham, N., O’Brien, C. J., Turtle,J. R., and Dong, Q. Prevalence and distribution of ret/ptc 1, 2, and 3 in papillary thyroid carcinoma in New Caledonia and Australia, J Clin Endocrinol Metab, 85: 2733–2739, 2000.

Bednarczuk, T., Hiromatsu, Y., Fukutani, T., Jazdzewski, K., Miskiewicz, P., Osikowska, M., Nauman, J. Association of cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) gene polymorphism and non- genetic factors with Graves’ ophthalmopathy in European and Japanese populations. Eur J Endocrinol,

148: 13–18, 2003.

Kaneshige, M., Kaneshige, K., Zhu, X. G., Dace, A., Garrett, L., Carter, T. A., Kazlauskaite, R., Pankratz, D. G., Wynshaw-Boris, A., Refetoff, S., Weintraub, B., Willingham, M. C., Barlow, C., and Cheng, S.-y. Mice with a targeted mutation in the thyroid hormone receptor gene exhibit impaired growth and resistance to thyroid hormone. Proc Natl Acad Sci USA, 97: 13209–13214, 2000.

Yen, P. M. Molecular basis of resistance to thyroid hormone. Trends Endocrinol Metab, 14: 327–333, 2003.

Weiss, R. E. and Refetoff, S. Resistance to thyroid hormone. Rev Endocrinol Metab Disord, 1: 97–108, 2000.

Meier, C. A., Dickstein, B. M., Ashizawa, K., McClaskey,J. H., Muchmore, P., Ransom, S. C., Merke, J. B., Hao, E. U., Usala, S. J., Bercu, B. B., Cheng, S.-y., and Weintraub, B. D. Variable transcriptional activity and ligand binding of mutant 1 3,3’,5-triiodo-L-thyronine receptors from four families with generalized resistance to thyroid hormone. Mol Endocrinol, 6: 248–258, 1992.

27.

28.

29.

30.

31.

32.

33.

34.

35.

36.

37.

38.

39.

40.

41.

42.

43.

44.

45.

46.

thyroid carcinomas from Saudi Arabia. Cancer, 73: 176–180, 1994.

(13)

Suzuki, H., Willingham, M. C., and Cheng, S. Y. Mice with a mutation in the thyroid receptor beta gene spontaneously develop thyroid carcinoma: a mouse model of thyroid carcinogenesis. Thyroid, 12:

963–969, 2002.

Ying, H., Suzuki, H., Furumoto, H., Walker, R., Meltzer, P., Billingham, M. C., and Cheng, S.

Y. Alterations in genomic profiles during tumor progression in a mouse model of follicular thyroid carcinoma. Carcinogenesis, 24: 1467–1479, 2003.

Khoo, M. L., Beasley, N. J., Ezzat, S., Freeman, J. L., Asa, S. L. Overexpression of cyclin D1 and underexpression of p27 predict lymph node metastases in papillary thyroid carcinoma. J Clin Endocrinol Metab, 87: 1814–1818, 2002.

Bieche, I., Franc, B., Vidaud, D., Vidaud, M., and Lidereau, R. Analyses of MYC, ERBB2, and CCND1 genes in benign and malignant thyroid follicular cell tumors by real-time polymerase chain reaction. Thyroid, 11: 147–152, 2001.

Heaney, A. P., Nelson, V., Fernando, M., and Horwitz, G., 2001. Transforming events in thyroid tumorigenesis and their association with follicular lesions. J Clin Endocrinol Metab, 86: 5025–5032, 2001.

Ishigaki, K., Namba, H., Nakashima, M., Nakayama, T., Mitsutake, N., Hayashi, T., Maeda, S., Ichinose, M., Kanematsu, T., Yamashita, S. Aberrant localization of beta-catenin correlates with overexpression of its target gene in human papillary thyroid cancer. J Clin Endocrinol Metab, 87: 433–440, 2002.

Holting, T., Goretzki, P. E., and Duh,Q. Y. Follicular thyroid cancer cells: a model of metastatic tumor in vitro. Oncol Rep, 8: 3–8, 2001.

Yen, P. M. and Cheng, S-y. Germline and somatic thyroid hormone receptor mutations in man. J Endocrinolo Invest, 26: (pages not known), in press, 2003.

Lin, K. H., Zhu, X. G., Hsu, H. C., Chen, S. L., Shieh, H. Y., Chen, S. T., McPhie, P., and Cheng, S. Y. Dominant negative activity of mutant thyroid hormone alpha 1 receptors from patients with hepatocellular carcinoma. Endocrinology, 138: 5308–5315, 1997.

Kimura, T., Van Keymeulen, A., Golstein, J., Fusco, A., Dumont, J. E., and Roger, P. P. Regulation of thyroid cell proliferation by TSH and other factors: a critical evaluation of in vitro models. Endocr Rev, 22: 631–656, 2001.

Rivas, M. and Santisteban, P. TSH-activated signaling pathways in thyroid carcinogenesis. Molecular Cellular Endo Review (in press), 2003.

Fagin, J.A. Branded from the start-distinct oncogenic initiating events may determine tumor fate in the thyroid (minireview). Mol Endocrinol, 16: 903–911, 2002.

Desvergne, B. and Wahli, W. Peroxisome proliferator-activated receptors: nuclear control of metabolism.

Endocr Rev, 20: 649–688, 1999.

Fajas, L., Webril, M. B., Auwerx, J. Peroxisome proliferator-activated receptor-gamma: from adipoge- nesis to carcinogenesis. J Mol Endocrinol, 27: 1–9, 2001.

Fajas, L., Auboeuf, D., Raspe, E., Schoonjans, K., Lefebvre, A. M., Saladin, R., Najib, J., Laville, M., Fruchart.J. C., Deeb, S., Vidal-Puig, A., Flier, J., Briggs, M. R., Staels, B., Vidal, H., and Auwerx, J.

The organization, promoter analysis, and expression of the human PPARgamma gene. J Biol Chem, 272: 18779–18789, 1997.

Altiok, S., Xu, M., and Spiegelman, B. M. PPARgamma induces cell cycle withdrawal: inhibition of E2F/DP DNA-binding activity via down-regulation of PP2A. Genes Dev, 11: 1987–1998, 1997.

Tontonoz, P., Singer, S., Forman, B. M., Sarraf, P., Fletcher,J. A., Fletcher, C. D., Brun, R. P., Mueller, E., Altiok, S., Oppenheim, H., Evans, R. M., and Spiegelman, B. M. Terminal differentiation of human liposarcoma cells induced by ligands for peroxisome proliferator-activated receptor gamma and the retinoid X receptor. Proc Natl Acad Sci USA, 94: 237–241, 1997.

Elstner, E., Muller, C., Koshizuka, K., Williamson, E. A., Park, D., Asou, H., Shintaku, P., Said, J. W., Heber, D., and Koeffler, H. P. Ligands for peroxisome proliferator-activated receptor and retinoic acid receptor inhibit growth and induce apoptosis of human breast cancer cells in vitro and in BNX mice.

Proc Natl Acad Sci USA, 95: 8806–8811, 1998.

Mueller, E., Sarraf, P., Tontonoz, P., Evans, R. M., Martin, K. J., Zhang, M., Fletcher, C., Singer, S., Spiegelman, B. M. Terminal differentiation of human breast cancer through PPAR. Mol Cell, 1:

465–470, 1998.

Brockman, J. A., Gupta, R. A., and DuBois, R. N. Activation of PPAR leads to inhibition of anchorage- independent growth of human colorectal cancer cells. Gastroenterology, 115: 1049–1055, 1998.

Kuboto, T., Koshizuka, K., Williamson, I. A., Asou, H., Said, J. W., Holden, S., Miyoshi, I., Koeffler, H. P. Ligand for peroxisome proliferator activated receptor (troglitazone) has potent anti-tumor effects against human prostate cancer both in vitro and in vivo. Cancer Res, 58: 3344–3352, 1998.

47.

48.

49.

50.

51.

52.

53.

54.

55.

56.

57.

58.

59.

60.

61.

62.

63.

64.

65.

66.

67.

(14)

68.

69.

70.

71.

72.

73.

74.

75.

76.

Sarraf, P., Mueller, E., Jones, D., King, F.J., De Angelo, D.J., Partridge, J. B., Holden, S. A., Chen, L.

B., Singer, S., Fletcher, C., and Spiegelman, B. M. Differentiation and reversal of malignant changes in colon cancer through PPAR gamma. Nat Med, 4: 1046–1052, 1998.

Ohta, K., Endo, T., Haraguchi, K., Hershman, J. M., Onaya, T. Ligands for peroxisome proliferator- activated receptor gamma inhibit growth and induce apoptosis of human papillary thyroid carcinoma cells. J Clin Endocrinol Metab 86: 2170–2177, 2001.

Martelli, M. L., Iuliano, R., Le Pera, I., Sama’, I., Monaco, C., Cammarota, S., Kroll, T., Chiariotti, L., Santoro, M., Fusco, A. Inhibitory effects of peroxisome poliferator-activated receptor gamma on thyroid carcinoma cell growth. J Clin Endocrinol Metab, 87: 4728–4735, 2002.

Ying, H., Suzuki, H., Zhao, L., Willingham, M. C., Meltzer, P., Cheng, S. Y. Mutant thyroid hormone receptor beta represses the expression and transcriptional activity of peroxisome proliferator-activated receptor gamma during thyroid carcinogenesis. Cancer Res, 63: 5274–5280, 2003.

Schoonjans, K., Peinado-Onsurbe, J., Lefebvre, A. M., Heyman, R. A., Briggs, M., Deeb, S., Staels, B., and Auwerx, J. PPARalpha and PPARgamma activators direct a distinct tissue-specific transcriptional response via a PPRE in the lipoprotein lipase gene. EMBO J., 15: 5336–5348, 1996.

Kroll, T. G., Sarraf, P., Pecciarini, L., Chen, C. J., Mueller, E., Spiegelman, B. M., and Fletcher, J. A.

PAX8-PPARgamma1 fusion oncogene in human thyroid carcinoma. Science, 289: 1357–1360, 2000.

Nikiforova, M. N., Biddinger, P. W., Caudill, C. M., Kroll, T. G., and Nikiforov, Y. E. PAX8- PPARgamma rearrangement in thyroid tumors: RT-PCR and immunohistochemical analyses. Am J Surg Pathol, 26: 1016–1023, 2002.

Marques, A. R., Espadinha, C., Catarino, A. L., Moniz, S., Pereira, T., Sobrinho, L. G., Leite, V. Expres- sion of PAX8-PPAR gamma 1 rearrangements in both follicular thyroid carcinomas and adenomas. J Clin Endocrinol Metab, 87: 3947–3952, 2002.

French, C. A., Alexander, E. K., Cibas, E. S., Nose, V., Laguette, J., Faquin, W., Garber, J., Moore, F.

Jr., Fletcher,J. A., Larsen, P. R., Kroll, T. G. Genetic and biological subgroups of low-stage follicular

thyroid cancers Am J Pathol, 162: 1053–1060, 2003.

Riferimenti

Documenti correlati

Even though the tested compounds show, on average, poor activities on this isozyme, the availability of resolved structures in complex with structurally related activators [ 19 ]

The significant role of both ADH and PDC for hypoxia survival has been demonstrated in many plant species, which differ in their degree of low oxygen tolerance, such as maize,

This is due to the correla- tion between the acceptance curve and the response function, which are simultaneously extracted in the Michel spectrum fit, and to the dependence of

Euroasiatic: species mainly diffused in the temperate regions of Europe and Asia (Euras), or limited to western Europe and Asia (EurWAs) or southern Europe and

Thyroid hormone levels have been reported to be normal, increased, and decreased in obese patients (4); this discrepancy among studies probably reflects the fact that patients

Phenolic compounds, ascorbic acid and carotenoids, derived from different plant species, are able to protect the skin by preventing UV penetration, reducing inflammation and

delle disposizioni del titolo I-bis del t.u.f., ma legittima la stessa Commissione a compiere tutti gli atti necessari all’accertamento delle violazioni, utilizzando

+zdjhv, e| d odujh dprxqw zkhq wkh| fdq uhvhw wkhp1 Lq wkh deryh prg0 hov/ wkh| zrxog eh zloolqj wr gr vr rqo| iru rqh uhdvrq= wr suhvhuyh ghpdqg1 Wkh| uhfrjqlvh wkdw/ iroorzlqj