Cancer Letters

Cancer Letters

Volume 192, Issue 2, 31 March 2003, Pages 121-132
Cancer Letters

Mini Review
Thyroid hormone receptors/THR genes in human cancer

https://doi.org/10.1016/S0304-3835(02)00614-6Get rights and content

Abstract

Thyroid hormone (triiodothyronine, T3) is a pleiotropic regulator of growth, differentiation and tissue homeostasis in higher organisms that acts through the control of target gene expression. Most, if not all, major T3 actions are mediated by specific high affinity nuclear receptors (TR) which are encoded by two genes, THRA and THRB. Several TRα and TRβ receptor isoforms are expressed. Abundant and contradictory literature exists on the relationship between circulating thyroid hormone levels, thyroid diseases and human cancer. In 1986, a connection between TR and cancer became evident when the chicken TRα1 was characterized as the c-erbA proto-oncogene, the cellular counterpart of the retroviral v-erbA oncogene. V-erbA causes erythroleukemias and sarcomas in birds, and hepatocellular carcinomas in transgenic mice. In recent years, many studies have analyzed the presence of quantitative (abnormal levels) or qualitative (mutations) alterations in the expression of THR genes in different types of human neoplasias. While their role in tumor generation or progression is currently unclear, both gross chromosomal and minor mutations (deletions, aberrant splicing, point mutations) and changes in the level of expression of THRA and THRB genes have been found. Together with other in vitro data indicating connections between TR and p53, Rb, cyclin D and other cell cycle regulators and oncogenes, these results suggest that THRA and THRB may be involved in human cancer.

Introduction

Thyroid hormone (triiodothyronine or T3) is an important regulator of growth, development and differentiation in vertebrates. T3 binds to specific high affinity receptors (thyroid receptors, TR) which belong to the superfamily of nuclear receptors [1]. They function as ligand-modulated transcription factors by binding to sequences known as thyroid hormone response elements (TRE) usually located in the promoter regions of target genes [2], [3]. Multiple studies have shown that gene regulation by T3 is modulated by the formation of heterodimers between TR and other members of the receptor superfamily, mainly the retinoic X receptors (RXR), and by subsequent interaction with transcriptional co-regulators (co-activators and co-repressors) and protein complexes [4], [5]. Both positive and negative effects of T3 on gene transcription have been described [6], [7]. In addition, ligand-activated TRs control gene transcription indirectly by interfering with the function of other transcription factors such as activating protein-1 (AP-1), formed by members of the c-Jun and c-Fos proto-oncogenic proteins [8], [9]. A third, less known mechanism of gene regulation by TR is the control of various post-transcriptional processes including splicing, mRNA stability and translation, and protein turn-over [10], [11], [12], probably by regulating gene transcription [13]. Finally, non-genomic actions of T3 through rapid mechanisms that remain uncharacterized have been also proposed [14].

The human TRs are encoded by the TRα (THRA) and TRβ (THRB) genes, located on human chromosomes 17 and 3, respectively. By alternative splicing and differential promoter usage, these two genes yield several polypeptides [2], [3] (Fig. 1). TRα1, TRβ1 and TRβ2 are well characterized and encode functional products. Another isoform named TRβ3 has been cloned from a rat osteosarcoma line and its function is still unknown [15]. Other isoforms such as TRα2 do not bind T3 or dimerize but they can still bind to TREs and inhibit functional receptors in transiently transfected cells [16], [17]. In vivo, however, this inhibitory action of TRα2 is not clear as cells expressing high TRα2 levels respond well to T3 [18] and effects of deleting TRα2 expression in mice have been observed [19]. The physiological roles of the non-binding polypeptides TRα2, TRΔβ3, and two additional truncated TRΔα1 and TRΔα2 polypeptides [20] remain elusive.

TRs affect gene transcription in four ways: (a) induction by hormone-bound TR or (b) inhibition by unliganded TR at positive TREs; and (c) inhibition by hormone-bound TR or (d) activation by unliganded TR at negative TREs. At positive TREs repression of basal transcription by unliganded TR (silencing) is mediated by interaction with co-repressors. TRβ2 seems to be the exception, activating transcription even in the absence of hormone [21]. Hormone binding induces a conformational change in TR leading to co-repressors release and subsequent co-activator binding that leads to modification of chromatin structure and transcriptional activation [5], [22], [23]. The mechanism of transcriptional inhibition at negative TREs is less well understood.

TR are modular proteins (Fig. 2). Their most characteristic domains are the highly conserved DNA-binding domain (DBD) which is also involved in dimerization, and the carboxy-terminal ligand-binding domain (LBD) responsible for T3 binding, dimerization and interaction with co-regulators [1]. Interaction with the cell basal transcription apparatus depends on the amino-terminal region (activating function 1, AF-1), while hormone-dependent transcription regulation (co-repressor and co-activator binding) relies on the extreme carboxy-terminal helix 12 sequence (activating function-2, AF-2). However, some isoforms such as rat TRβ3 [15] or the chicken TRβ0 [24] and TRαp40 (translated from an internal AUG codon; [25]) lack the AF-1 region.

In 1986, TRα1 was found to be the cellular counterpart of the retroviral v-erbA oncogene that contributes to the appearance of erythroleukemia and sarcomas in birds [26], [27]. v-erbA encodes a highly mutated chicken TRα1 protein that does not bind T3 and acts as a constitutive repressor of T3-regulated genes [28]. V-erbA has weak oncogenic potential itself but potentiates the activity of oncoproteins derived from tyrosine kinases and downstream signal transducers (see reviews [29], [30]). It enhances the transformed phenotype of cultured erythroblasts and fibroblasts by arresting cellular differentiation and favoring proliferation (see reviews [29], [30]). Importantly, v-erbA transgenic mice develop hepatocellular carcinomas [31].

In recent years, increasing evidence has suggested that aberrant expression and mutations in THR genes could be associated with carcinogenesis in humans. The focus of this review will be to provide an overview of what we know about the status and expression of THR genes in human cancers.

Section snippets

Thyroid hormone and cancer

Many studies in vitro and in vivo have related thyroid hormones and human cancer since Beatson [32] described the use of thyroid extracts for breast cancer treatment more than a century ago. Abundant data indicate that thyroid status and disease affect tumor formation, growth and metastasis in experimental animals and humans [33], [34]. While these studies demonstrated modulation by thyroid hormone of the neoplastic process, no consistent pattern of effects was seen, which suggests confounding

Thyroid hormone receptors in human tumors: cytogenetic alterations

Since the characterization in 1986 of the c-erbA proto-oncogene as the chicken TRα1 gene, a number of studies have analyzed the status of TR isoforms in a variety of human neoplasias. Several reports have shown TR alterations and suggested that they could be involved in the development of human cancers.

High frequencies of somatic deletions have been observed in chromosome 3p where THRB lies, in many malignancies including small cell lung, breast, head and neck, renal cell, uterine cervical,

Thyroid hormone receptors in human cancer: expression and integrity

Besides large chromosomal defects, altered expression and small mutations on THR genes have also been reported.

Concluding remarks

The role of thyroid hormone in human cancers has yet to be elucidated. From a series of studies reviewed here, alterations in the level of expression and integrity of THR genes occur in different neoplasias (Table 1). The biological relevance for the pathogenesis and progression of these processes is, however, unknown.

The evidence of alterations that affect THR genes, their expression and the activity of their protein products in human cancers makes the analysis of THR genes in human cancer an

Acknowledgements

We thank Professor B. Vennström and Dr D. Forrest for critical reading and discussion of this review, and Robin Rycroft for his valuable assistance in the preparation of the English manuscript. The work of the authors is supported by Grants 08.1/0069/2000 from the Comunidad Autónoma de Madrid and SAF2001-2291 from Ministerio de Ciencia y Tecnologı́a of Spain.

References (104)

  • C.J. Wang et al.

    Expression of thyroid hormone receptors in human pituitary tumor cells

    Cancer Lett.

    (1995)
  • M. Puzianowska-Kuznicka et al.

    Expression of thyroid hormone receptors is disturbed in human renal clear cell carcinoma

    Cancer Lett.

    (2000)
  • J.W. Lee et al.

    Thyroid hormone receptor α1 (c-erbAα1) suppressed transforming phenotype of nasopharyngeal carcinoma cell line

    Cancer Lett.

    (2002)
  • H.-M. Lin et al.

    Cyclin D1 is a ligand-independent co-repressor for thyroid hormone receptors

    J. Biol. Chem.

    (2002)
  • Y.M. Tokumoto et al.

    Posttranscriptional regulation of p18 and p27 Cdk inhibitor proteins and the timing of oligodendrocyte differentiation

    Dev. Biol.

    (2002)
  • G. Pérez-Juste et al.

    The cyclin-dependent kinase inhibitor p27kip1 is involved in thyroid hormone-mediated neuronal differentiation

    J. Biol. Chem.

    (1999)
  • A. Muñoz et al.

    Biological activities of thyroid hormone receptors

    Eur. J. Endocrinol.

    (1997)
  • J. Bernal et al.

    Thyroid hormone and the development of the brain

    Curr. Opin. Endocr. Diab.

    (1998)
  • N.J. McKenna et al.

    Nuclear receptor coregulators. Cellular and molecular biology

    Endocr. Rev.

    (1999)
  • H.-J. Lee et al.

    Recent advances in understanding thyroid hormone receptor coregulators

    J. Biomed. Sci.

    (1999)
  • J. Bernal

    Action of thyroid hormone in brain

    J. Endocrinol. Invest.

    (2002)
  • X.K. Zhang et al.

    Novel pathway for thyroid hormone receptor action through interaction with jun and fos oncogene activities

    Mol. Cell. Biol.

    (1991)
  • C. Caelles et al.

    Nuclear hormone receptor antagonism with AP-1 by inhibition of the JNK pathway

    Genes Dev.

    (1997)
  • J.E. Silva et al.

    Effects of congenital hypothyroidism on microtubule-associated protein-2 expression in the cerebellum of the rat

    Endocrinology

    (1990)
  • F. Aniello et al.

    Splicing of juvenile and adult tau mRNA variants is regulated by thyroid hormone

    Proc. Natl Acad. Sci. USA

    (1991)
  • P.J. Davis et al.

    Nongenomic actions of thyroid hormone

    Thyroid

    (1999)
  • G.R. Williams

    Cloning and characterization of two novel thyroid hormone receptor β isoforms

    Mol. Cell. Biol.

    (2000)
  • M.A. Lazar et al.

    Human carboxyl-terminal variant of α-type c-erbA inhibits trans-activation by thyroid hormone receptors without binding thyroid hormone

    Proc. Natl Acad. Sci. USA

    (1989)
  • R.J. Koenig et al.

    Inhibition of thyroid hormone action by a non-hormone binding c-erbA protein generated by alternative mRNA splicing

    Nature

    (1989)
  • C. Salto et al.

    Ablation of TRα2 and a concomitant overexpression of a1 yields a mixed hypo- and hyperthyroid phenotype in mice

    Mol. Endocrinol.

    (2001)
  • O. Chassande et al.

    Identification of transcripts initiated from an internal promoter in the c-erbA α locus that encode inhibitors of retinoic acid receptor-α and triiodothyronine receptor activities

    Mol. Endocrinol.

    (1997)
  • M. Sjörberg et al.

    Ligand-dependent and -independent transactivation by thyroid hormone receptor β2 is determined by the structure of the hormone response element

    Mol. Cell. Biol.

    (1995)
  • D. Forrest et al.

    Contrasting developmental and tissue-specific expression of α and β thyroid hormone receptor genes

    EMBO J.

    (1990)
  • J. Bigler et al.

    c-erbA encodes multiple proteins in chicken erythroid cells

    Mol. Cell. Biol.

    (1988)
  • J. Sap et al.

    The c-erbA protein is a high affinity receptor for thyroid hormone

    Nature

    (1986)
  • C. Weinberger et al.

    The c-erbA encodes a thyroid hormone receptor

    Nature

    (1986)
  • J. Sap et al.

    Repression of transcription mediated at a thyroid response element by the v-erbA oncogene product

    Nature

    (1989)
  • A. Rascle et al.

    The v-erbA oncogene

  • D. Thormeyer et al.

    The v-erbA oncogene (review)

    Int. J. Mol. Med.

    (1999)
  • C. Barlow et al.

    Thyroid abnormalities and hepatocellular carcinoma in mice transgenic for v-erbA

    EMBO J.

    (1994)
  • D.L. Guernsey

    Thyroid hormone action

    Cancer J.

    (1993)
  • P.P.A. Smyth

    The thyroid and breast cancer: a significant association?

    Ann. Med.

    (1997)
  • F. Leduc et al.

    Loss of heterozygosity in a gene coding for a thyroid hormone receptor in lung cancers

    Am. J. Hum. Genet.

    (1989)
  • K. Sisley et al.

    Loss of heterozygosity of the thyroid hormone receptor β in posterior uveal melanoma

    Melanoma Res.

    (1993)
  • L.C. Chen et al.

    Detection of two separate regions on chromosome 3p in breast cancers

    Cancer Res.

    (1994)
  • I.U. Ali et al.

    Presence of two members of c-erbA receptor gene family (c-erbAβ and c-erbA2) in smallest region of somatic homozygosity on chromosome 3p21-3p25 in human breast carcinoma

    J. Natl Cancer Inst.

    (1989)
  • A. Dobrovic et al.

    erbA-related genes coding for DNA-binding hormone receptors localized to chromosome 3p21-3p25 and deleted in small cell lung carcinoma

    Cancer Res.

    (1988)
  • H. Drabkin et al.

    Localization of human ERBA2 to the 3p22-3p24.1 region of chromosome 3 and variable deletion in small cell lung cancer

    Proc. Natl Acad. Sci. USA

    (1988)
  • T. Huber-Gieseke et al.

    Lack of loss of heterozygosity at the c-erbA β locus in gastrointestinal tumors

    Oncology

    (1997)
  • P.A. Futreal et al.

    Detection of frequent allelic loss on proximal chromosome 17q in sporadic breast carcinoma using microsatellite length polymorphisms

    Cancer Res.

    (1992)
  • Cited by (92)

    • The antigenic link between thyroid autoimmunity and breast cancer

      2020, Seminars in Cancer Biology
      Citation Excerpt :

      In particular, the receptors of thyroid hormones are expressed in both normal [32,33] and neoplastic [34,35] mammary epithelial cells. The thyroid hormones, especially T3, exert a permissive role in breast development and lactation since inducing its differentiation and lobular growth in an oestrogen-like manner [32,33,35–38]. Similarly, in rodent models of tumour transplant the tumour growth and metastases appeared to be stimulated by thyroid hormones, whereas treatment-induced hypothyroidism had opposite effects, determining a favorable outcome not only for BC, but also other cancer types [39].

    • Thyroid hormone in the regulation of hepatocellular carcinoma and its microenvironment

      2018, Cancer Letters
      Citation Excerpt :

      No such relationship, however, was found in men (see Table 1) [22,23,34–36,46–48]. The role of TH and TRs in HCC is further supported by studies describing the association between somatic TR mutation and human neoplasia (reviewed in Ref. [49]). In an earlier study, it was shown that naturally occurring TRα mutations (V390A) (E350K/P398S) from HCCs of two patients abrogate the functions of TRs via a dominant negative effect.

    • Contribution of mammalian selenocysteine-containing proteins to carcinogenesis

      2017, Journal of Trace Elements in Medicine and Biology
      Citation Excerpt :

      Additionally, a decrease in the activity of this enzyme and the expression of d1 was registered in liver hemangioma [108], renal clear cell cancer [109], lung cancer, pituitary adenomas [110,111]. At the same time, human breast cancer tissues exhibited an increase of D1 activity, as compared to healthy tissue [112,113], assuming that the metabolism of thyroid hormones in neoplastic tissues is changed [114,115]. It was hypothesized that proinflammatory cytokines promote a reduction of the activity of D1, though analysis of enzymatic activity of D1 in rat hepatic cells phi1 under the action of TNF-α, IL-1β and IL-G carried out by Davies and co-authors provided no confirmation for this assumption [116].

    View all citing articles on Scopus
    View full text