19
DOI: 10.1530/ERC-17-0192 http://erc.endocrinology-journals.org © 2017 Society for Endocrinology Printed in Great Britain Published by Bioscientifica Ltd. Endocrine-Related Cancer 24:11 Review I M Goemann et al. Thyroid hormones and neoplasias Role of thyroid hormones in the neoplastic process: an overview Iuri Martin Goemann 1 , Mirian Romitti 1 , Erika L Souza Meyer 2 , Simone Magagnin Wajner 1 and Ana Luiza Maia 1 1 Thyroid Section, Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil 2 Department of Internal Medicine, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil Abstract Thyroid hormones (TH) are critical regulators of several physiological processes, which include development, differentiation and growth in virtually all tissues. In past decades, several studies have shown that changes in TH levels caused by thyroid dysfunction, disruption of deiodinases and/or thyroid hormone receptor (TR) expression in tumor cells, influence cell proliferation, differentiation, survival and invasion in a variety of neoplasms in a cell type-specific manner. The function of THs and TRs in neoplastic cell proliferation involves complex mechanisms that seem to be cell specific, exerting effects via genomic and nongenomic pathways, repressing or stimulating transcription factors, influencing angiogenesis and promoting invasiveness. Taken together, these observations indicate an important role of TH status in the pathogenesis and/or development of human neoplasia. Here, we aim to present an updated and comprehensive picture of the accumulated knowledge and the current understanding of the potential role of TH status on the different hallmarks of the neoplastic process. Introduction The association between thyroid hormone (TH) status and cancer was reported as early as 1896, when Beatson used thyroid extract as a potential treatment for breast cancer (Beatson 1896). Since then, an impressive expansion of knowledge has established THs as key regulators of several physiological processes, including the embryonic development, growth and metabolism of virtually all tissues (Yen 2001). Additionally, recent data have demonstrated critical roles of THs in cell proliferation, differentiation and survival (Dentice et al. 2007, Lin et al. 2009, Pascual & Aranda 2013, Romitti et al. 2013, Sterle et al. 2014, Miro et al. 2017). The human thyroid gland mainly secretes thyroxine (T 4 ), but the active hormone, triiodothyronine (T 3 ), mediates most of the hormonal actions. The main pathway for the production of the bioactive form in peripheral tissues occurs via outer ring deiodination of T 4 through the action of iodothyronine deiodinase types 1 and 2 (DIO1; D1 and DIO2; D2). In contrast, type 3 iodothyronine deiodinase (DIO3; D3) is mainly responsible for TH inactivation via inner-ring deiodination of both T 4 and T 3 (Maia et al. 2011). Intracellular T 3 bioavailability is controlled in a tissue-specific manner, depending mainly on its activation by D2 and inactivation by D3. Notably, proper deiodinase function depends on the availability of a yet unidentified thiol cofactor that acts as a reducing agent during the catalysis (Visser et al. 1976). Conditions that result in dysregulation of the intracellular redox state Endocrine-Related Cancer (2017) 24, R367–R385 R367–R385 Correspondence should be addressed to A L Maia Email [email protected] Key Words f thyroid hormones f thyroid hormone receptors f iodothyronine deiodinases f neoplasia f carcinogenesis Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AM via free access

Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

End

ocr

ine-

Rel

ated

Can

cer

24:11Review

I M Goemann et al. Thyroid hormones and neoplasias

10.1530/ERC-17-0192

Role of thyroid hormones in the neoplastic process: an overview

Iuri Martin Goemann1, Mirian Romitti1, Erika L Souza Meyer2, Simone Magagnin Wajner1 and Ana Luiza Maia1

1Thyroid Section, Endocrine Division, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil2Department of Internal Medicine, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil

Abstract

Thyroid hormones (TH) are critical regulators of several physiological processes, which

include development, differentiation and growth in virtually all tissues. In past decades,

several studies have shown that changes in TH levels caused by thyroid dysfunction,

disruption of deiodinases and/or thyroid hormone receptor (TR) expression in tumor

cells, influence cell proliferation, differentiation, survival and invasion in a variety of

neoplasms in a cell type-specific manner. The function of THs and TRs in neoplastic cell

proliferation involves complex mechanisms that seem to be cell specific, exerting effects

via genomic and nongenomic pathways, repressing or stimulating transcription factors,

influencing angiogenesis and promoting invasiveness. Taken together, these observations

indicate an important role of TH status in the pathogenesis and/or development of

human neoplasia. Here, we aim to present an updated and comprehensive picture of the

accumulated knowledge and the current understanding of the potential role of TH status

on the different hallmarks of the neoplastic process.

Introduction

The association between thyroid hormone (TH) status and cancer was reported as early as 1896, when Beatson used thyroid extract as a potential treatment for breast cancer (Beatson 1896). Since then, an impressive expansion of knowledge has established THs as key regulators of several physiological processes, including the embryonic development, growth and metabolism of virtually all tissues (Yen 2001). Additionally, recent data have demonstrated critical roles of THs in cell proliferation, differentiation and survival (Dentice et al. 2007, Lin et al. 2009, Pascual & Aranda 2013, Romitti  et  al. 2013, Sterle et al. 2014, Miro et al. 2017).

The human thyroid gland mainly secretes thyroxine (T4), but the active hormone, triiodothyronine (T3),

mediates most of the hormonal actions. The main pathway for the production of the bioactive form in peripheral tissues occurs via outer ring deiodination of T4 through the action of iodothyronine deiodinase types 1 and 2 (DIO1; D1 and DIO2; D2). In contrast, type 3 iodothyronine deiodinase (DIO3; D3) is mainly responsible for TH inactivation via inner-ring deiodination of both T4 and T3 (Maia et al. 2011). Intracellular T3 bioavailability is controlled in a tissue-specific manner, depending mainly on its activation by D2 and inactivation by D3. Notably, proper deiodinase function depends on the availability of a yet unidentified thiol cofactor that acts as a reducing agent during the catalysis (Visser et al. 1976). Conditions that result in dysregulation of the intracellular redox state

Endocrine-Related Cancer (2017) 24, R367–R385

2411

R367–R385

Correspondence should be addressed to A L Maia Email [email protected]

Key Words

f thyroid hormones

f thyroid hormone receptors

f iodothyronine deiodinases

f neoplasia

f carcinogenesis

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 2: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R368Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

possibly interfere with endogenous cofactor(s) levels, thereby impairing deiodinase activity (Wajner et al. 2015).

THs exert their effects through genomic (nuclear) and nongenomic (cytoplasmic or membrane TH receptor (TR)) pathways. The genomic mechanisms are mediated mostly by T3 through nuclear TRs. The TRα and TRβ genes encode the TH-binding TR isoforms TRα1 and TRβ1–β3 (Kim et al. 2012). T3 binds to nuclear TRs that activate the transcription of target genes by binding to TH response elements (TREs) located in the regulatory regions. Gene transcription is regulated by an exchange of corepressor (CoR) and coactivator (CoA) complexes. Negative TREs (nTREs) can mediate ligand-dependent transcriptional repression. However, in this case, the roles of CoAs and CoRs are not well defined (Yen 2001). The nature of the transcriptional response is determined by cell type and hormone status (Hulbert 2000, Aranda & Pascual 2001). On the other hand, the nongenomic effects are initiated by TH binding to integrin αVβ3 receptor, which leads to the activation of different signaling pathways, including mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K), signal transducers and activators of transcription (STAT) pathways. These cascades result in distinct cellular events, such as cell division, proliferation and angiogenesis (Bergh et al. 2005, Lin et al. 2007, Davis et al. 2008, 2009, Cheng et al. 2010).

In past decades, several clinical studies have indicated that an altered TH status might be a risk factor for the development of tumors, such as liver, breast, colon, prostate and thyroid malignancies (Boelaert  et  al. 2006, Reddy  et  al. 2007, Polyzos  et  al. 2008, Fiore  et  al. 2009, Hassan  et  al. 2009, Hellevik  et  al. 2009, Tosovic  et  al. 2010, 2014). However, other studies have described TH alterations as clinically favorable, such as hypothyroidism for high-grade glioblastomas (Hercbergs et al. 2003). Several in vitro and in vivo studies have demonstrated that THs influence a myriad of oncological events and control the balance between proliferation and differentiation, which is one of the most important hallmarks of TH action in cancer cells (Kress et al. 2009b, Dentice et al. 2013, Pascual & Aranda 2013). Changes in TH levels caused by thyroid dysfunction or the disruption of deiodinases and/or TR expression in tumor cells influence cell proliferation, differentiation, survival and invasion in a variety of neoplasms in a cell type-specific manner (Lin et al. 2008, Dentice et al. 2009, Pinto et al. 2011). The function of THs and TRs in neoplastic cell proliferation involves complex mechanisms that seem to be cell type specific, exerting effects via distinct pathways, repressing or stimulating

transcription factors, influencing angiogenesis and promoting invasiveness (Yen 2001, Kress et  al. 2009b). Here, we aim to present an updated picture of recent advances in the current understanding of the potential effects of TH status on the different hallmarks of the neoplastic process.

Overview of the neoplastic process

The hallmarks of the neoplastic process include sustained proliferation signaling, resistance to growth suppressors, evasion of programmed cell death, replicative immortality, sustained angiogenesis and promotion of invasion and metastasis (Hanahan & Weinberg 2000). In the past decade, two emerging characteristics have extended our understanding of this process: reprogramming energy metabolism and evasion from immune destruction, both contributing to a favorable tumor microenvironment (Kroemer & Pouyssegur 2008, de Souza  et  al. 2011, Hanahan & Weinberg 2011).

The acquisition of multiple cancer hallmarks depends on a succession of alterations in the cellular genome (Hanahan & Weinberg 2011). Alterations affecting the DNA maintenance machinery, such as defects in genes involved in the detection and repair of DNA damage, or tumor suppressor genes, have been associated with the progression of the neoplastic process (Kastan 2008, Jackson & Bartek 2009, Ciccia & Elledge 2010, Negrini et al. 2010).

Solid tumors can also recruit new blood vessels through the secretion of angiogenic factors. Vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF; FGF2) and platelet-derived growth factor (PDGF) are examples of molecules that promote the proliferation and migration of vascular endothelial cells and can severely constrain angiogenesis and tumor growth (Kim et al. 1993, Mousa et al. 2014).

Programmed cell death is a natural mechanism that is as important for healthy tissue growth as controlled cell proliferation. In order to grow indefinitely, cancer cells must overlap apoptosis mechanisms, disabling the cellular apoptosis-inducing circuitry. The intracellular apoptotic machinery depends on a family of proteolytic enzymes called caspases, which participate in a process that can be initiated by either extracellular or intracellular death signals. Caspase activation is tightly regulated by members of the B-cell lymphoma 2 (BCL2) and inhibitors of apoptosis proteins families, proteins that can either be pro- or anti-apoptotic (Evan & Vousden 2001, Lowe et al. 2004).

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 3: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R369Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

Another distinct attribute of cancer cells, which is functionally important for tumor development involves major reprogramming of the cellular energy metabolism to support continuous cell growth and proliferation, replacing the metabolic program that operates in most normal tissues (Hsu & Sabatini 2008). Neoplastic cells typically generate more reactive oxygen species (ROS) than normal cells, a mechanism that can be partially explained by oncogenic signaling and downregulated mitochondrial function (Lee  et  al. 1999, Gogvadze  et  al. 2008). ROS promote DNA damage and signaling mediation, and their presence may contribute to the transformation of cells (Dewhirst et al. 2008).

More recently, disruption of the mechanisms involved in cellular autophagy has emerged as a new hallmark of cancer (White 2015). Controlled autophagy prevents intracellular components, such as proteins, lipids and organelles, from accumulating, which can be harmful to cells (White 2012).

As the effects of THs on these processes are variable and complex, we comprehensively organized our review according to the cancer hallmarks described above (Fig. 1). The emerging effects of TH analogs on tumorigenesis and the disruption of signaling caused by TR mutations

have been discussed elsewhere (Cheng 2003, Gonzalez-Sancho et al. 2003, Aranda et al. 2009, Davis et al. 2014a,b, Mousa et al. 2014) and are not included in this review.

The roles of THs on the cellular hallmarks of cancer

TH effects on sustained proliferative signaling pathways

A vital capacity acquired by cancer cells involves their ability to sustain chronic proliferation through different pathways (Di Cristofano & Pandolfi 2000, Shields  et  al. 2000, Brazil & Hemmings 2001, Evan & Vousden 2001, Zhang & Liu 2002). THs influence cell growth, acting either as growth factors or as cell growth inhibitors through several proliferation pathways.

Davis and coworkers (Davis et al. 1999) demonstrated for the first time the nongenomic actions of THs in the induction of the MAPK pathway in HeLa and CV-1 cells (Lin  et  al. 1999a). T4 promotes the phosphorylation of MAPK and the co-immunoprecipitation of nuclear tyrosine phosphorylated MAPK with STAT-1a and STAT-3 (Lin et al. 1999b). This effect causes the MAPK-mediated serine phosphorylation of TRβ1, which dissociates the

Figure 1The effects of THs on the hallmarks of cancer involve several pathways and effectors. The THs (center) act via integrin αVβ3 or TRs (inner circle), modulating critical signaling pathways classically involved in carcinogenesis (middle circle). Note that for some nongenomically driven pathways, integrins have not been shown to be the membrane receptor mediators. Downstream targets of TH actions are represented in the outer circle.

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 4: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R370Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

TRβ1 and the co-repressor silencing mediator for retinoid receptors or TRs, thus affecting the nuclear receptor via a mechanism independent of the binding of T3 to TRβ1 (Davis et al. 2000). For this process to occur, a cell membrane T4 receptor is required. Later, the same group showed that a member of the plasma membrane heterodimeric integrin protein family, integrin αVβ3, binds T4 preferentially over T3 (Bergh et al. 2005). Presently, most of the nongenomic effects of THs are known to be mediated by activation of the integrin αVβ3 receptor, which sends several survival mechanism signals to the cell, including the stimulation of ERK- and AKT-dependent pathways (Cheng et al. 2010).

MAPK pathway The activation of MAPK (ERK1/2) by physiological levels of T4 influences tumor proliferation, as has been demonstrated in glioma (Davis  et al. 2006), follicular thyroid carcinoma (FTC) and papillary thyroid carcinoma (PTC) (Lin et  al. 2007), undifferentiated pheochromocytoma (Barbakadze et al. 2014) and myeloma (Cohen  et  al. 2015) (Fig.  2). In human breast cancer cells, T4 induces proliferation nongenomically, requiring ERK1/ERK2 and phosphorylating the estrogen receptor alpha (ERα). This observation highlights the crosstalk between THs and estrogen signaling pathways in certain cancer cells, culminating in specific intranuclear events

Figure 2Proposed mechanism of genomic and nongenomic actions of THs in the neoplastic process. The actions of THs occur at the plasma membrane, in the cytoplasm, and within the cell nucleus. To exert their genomic effects, T4 and T3 enter the cell through transporter proteins, such as monocarboxylate transporter (MCT) 8 and 10 or organic anion-transporting polypeptides. Inside the cells, D2 convert T4 to the active form T3, while D3 inactivates both THs, producing rT3 and T2 (1). T3 binds to nuclear TRs that activate transcription by binding TREs located in the regulatory regions of the target genes. Activity is regulated by an exchange of corepressor (CoR) and coactivator (CoA) complexes. Negative TREs (nTREs) can mediate ligand-dependent transcriptional repression; however, in this case, the roles of CoAs and CoRs are not well defined (2). THs can also regulate genes that do not contain a TRE by nongenomic effects. These ‘rapid effects’ are initiated by THs binding to integrin αVβ3 (3), leading to the activation of different signaling pathways and resulting in distinct cellular events, such as cell proliferation, migration, angiogenesis and apoptosis inhibition. One site of the integrin αVβ3 (4) binds T3 exclusively, activating PI3K via Src kinase (5), stimulating FAK, HIF-1α and mTOR, while also increasing the activity of the sodium pump (Na/K ATPase). The second site (4) binds T4 and T3, stimulating MAPK-dependent proliferation via phospholipase C (PLC) and protein kinase C (PKC), promoting the phosphorylation of several effectors (ERα, TRβ1, STAT1α, P52 and STAT-3, among others) (6). THs can induce the expression of matrix metalloproteinases (MMPs) nongenomically via MAPK and PI3K, thereby enhancing invasiveness (7). Another action THs initiate at the cell surface is modulation of the activity of the Na+/H+-exchanger and Na/K ATPase (8). Furthermore, T4 also interacts with a TRα variant in the cytoplasm to cause a modification of intracellular actin that contributes to cell migration (9). T3 negatively regulates UHRF1 through TRα1, leading to inhibition of cancer growth, by promoting stability of a cyclin-dependent kinase inhibitor (p21) (10). While T3 negatively or positively regulates Wnt/β-catenin expression, depending on the TR that is active, Wnt/β-catenin regulates the intracellular levels of T3 by modulating DIO2 and DIO3 expression. The D2 level is downregulated by β-catenin while D3 is induced, illustrating the complex crosstalk between THs and the Wnt/β-catenin pathway (11). Note that for some nongenomically driven pathways, integrin αVβ3 has not been demonstrated as the membrane receptor mediator.

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 5: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R371Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

(Tang et al. 2004). Another example of THs and estrogen crosstalk is the induction of proliferation in human lung cancer cells, which is initiated via the cell surface integrin αVβ3 (Meng et al. 2011).

T3 also activates MAPK nongenomically but only at supraphysiological levels (Davis et al. 2000, Kozawa et al. 2001). Studies in glioma cell lines have shown that T3 suppresses proliferation and induces redifferentiation in a mechanism independent of ERK 1/2 activation, suggesting a potential role of TRα1 (Liappas et al. 2011). In contrast, other studies have demonstrated that both T4 and T3 induce cell proliferation in glioblastoma and pheochromocytoma cells via ERK1/2 pathway activation (Lin et  al. 2009, Barbakadze et  al. 2014). In ovarian tumor cells, physiological concentrations of T3 and T4 induce MAPK-dependent cell proliferation and support cell survival in a process that requires an intact TH–integrin interaction for ERK activation (Shinderman-Maman et al. 2016).

The interaction between THs and the RAS signaling pathway also deserves attention due to its important role in carcinogenesis. RAS proteins act as key membrane signaling mediators by transferring information from this cellular compartment to the nucleus. RAS activates several pathways to regulate cell growth, survival, differentiation and angiogenesis; MAPK is a key downstream target of these pathways (Lowy & Willumsen 1993). Activating mutations in RAS genes and the consequent aberrations in the expression of the RAS–MAPK complex are implicated in several human cancers (Downward 2003, Rajalingam et al. 2007). Cyclin D1, which is critical for cell cycle progression, is one of the main elements mediating the proliferative effects of RAS oncogenes (Filmus  et  al. 1994). T3, acting through TRα1 and TRβ1, not only blocks the RAS-mediated proliferation of neuroblastoma cells via the regulation of cyclic AMP response elements but also represses their transcriptional activity, thus reducing the cyclin D1 levels and consequently the cell proliferation (Garcia-Silva & Aranda 2004). Studies performed using hepatocarcinoma (HCC) cells and breast cancer cells originally lacking TRs have shown that the reexpression of TRβ1 abolishes tumor growth and migration (Martinez-Iglesias  et  al. 2009) while preventing tumor formation by RAS-transformed cells in nude mice, even under hypothyroid conditions (Aranda et  al. 2009). In neuroblastoma (Neuro-2a) cells overexpressing TRβ1, T3 treatment blocks cell proliferation through an arrest of cells in G0/G1 and induces morphological and functional cell differentiation through acetylcholinesterase activity (Lebel et al. 1994). Taken together, these data indicate that

a loss of the expression and/or function of TRs could result in a selective advantage for malignant transformation in RAS-dependent tumors.

PI3K/protein kinase B pathway The PI3K/protein kinase B (AKT) pathway also plays a pivotal role in the regulation of cell growth and proliferation and its deregulation contributes to cellular transformation in a variety of neoplasms (Furuya  et  al. 2006, Franke 2008). Several nongenomic and genomic TH actions in tumors occur via the PI3K pathway. Incubation of endothelial cells with T3 increases the association of TRα1 with the p85α subunit of PI3K by non-transcriptional mechanisms, leading to the phosphorylation and activation of AKT (Hiroi et al. 2006). Notably, in a mouse model of FTC, a TRβ mutant can activate the PI3K regulatory subunit p85α, affecting signaling in both the nuclear and extranuclear compartments (Furuya et  al. 2006). Experimental data obtained using PTC and neuroblastoma cell lines show that T3 promotes the activation of ERK, AKT and Src. T3 can also induce AKT phosphorylation nongenomically through TRβ1 (Cao et al. 2009, Perri et al. 2014). In insulinoma cell lines (rRINm5F and hCM) that express TR isoforms TRα1, TRα2 and TRβ1, T3 induces cell proliferation and is also able to promote survival due to a regulation of different cellular apoptotic proteins, specifically activating the PI3K pathway (Verga Falzacappa  et  al. 2006). In non-tumoral β-cells, T3 action in the AKT pathway is also mediated by TRβ1, which contributes to the stimulation of proliferation and survival both in a rapid and long-term manner (Verga Falzacappa  et al. 2009). Interestingly, in contrast, T3 treatment enhances PI3K activity in glioblastoma cells but leads to nonproliferative downstream functions (Lin et al. 2009). Taken together, these observations show the critical role of T3 nongenomic effects on the rapid PI3K-AKT/PKB-mTOR activation in normal and neoplastic cells (Cao  et  al. 2005, Kenessey & Ojamaa 2006, Storey  et  al. 2006, Verga Falzacappa et al. 2009, Perri et al. 2014).

Unlike T3, T4 is unable to activate PI3K nongenomically, supporting the concept that the integrin αVβ3 receptor contains two specific sites in the hormone-binding domain. One site binds T3 exclusively and activates PI3K via Src kinase. The second site binds both T4 and T3, which in turn, activates ERK1/2-dependent tumor cell proliferation (Fig. 2) (Lin et al. 2009).

Recently, alternative mechanisms for T3- and T4-dependent AKT activation have been proposed. In human umbilical vein endothelial cells (HUVECs), neither T4- nor T3-induced AKT phosphorylation was attenuated by the addition of tetrac (which blocks T4 from binding to the

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 6: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R372Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

integrin αVβ3 receptor) suggesting that integrin αVβ3 is not involved in the nongenomic actions of THs in these cells and raising the question whether membrane-localized TRs are involved in such rapid actions of THs. Of interest, the blockade of D2 activity abolished AKT phosphorylation, indicating that the conversion of D2-catalyzed T4 to T3 is required for TRα1/PI3K-mediated nongenomic actions of T4 in HUVECs (Aoki et al. 2015).

Wnt/β-catenin pathway The Wnt signaling pathway has a critical role in the embryonic development and regeneration of tissues. Mutations and/or deregulated expression of the Wnt pathway can induce cancer (Polakis 1999, Klaus & Birchmeier 2008). β-Catenin, a central mediator in the Wnt pathway, interacts with E-cadherin to control the cellular functions (Gottardi & Gumbiner 2001). The relationship between T3 and the Wnt pathway was demonstrated by an elegant study performed by Miller and coworkers (Miller  et  al. 2001), which showed that T3-induced cell proliferation is associated with the immediate silencing of Wnt signaling in rat pituitary cells. Later, studies in colon cancer cells demonstrated that T3/TRβ1 suppress the transcription of cyclin D1 by wild-type β-catenin (Natsume  et  al. 2003). Therefore, T3/TR signaling can negatively regulate the Wnt pathway by inhibiting transactivation by β-catenin/Tcf on the cyclin D1 promoter. The physical interaction of β-catenin and TRβ was also demonstrated in a mouse model of thyroid cancer. T3 binding to TRβ weakened the β-catenin/TRβ interaction, increasing the amount of β-catenin available to be degraded via the proteasomal pathway (Guigon et al. 2008).

β-Catenin also interacts with TRα1, but causes different effects when compared to β-catenin/TRβ interaction. TRα1 is primarily responsible for cell cycle regulation and proliferation in the normal intestinal epithelium (Kress et al. 2009a). In these cells, T3-activated-TRα1 receptor directly controls the transcription of the β-catenin in vitro, promoting cell proliferation (Plateroti  et al. 2006). TRα1 overexpression also enhances the intestinal tumorigenic process in a predisposed genetic background. In human CaCo2 cells, TRα1 interacts with the β-catenin/Tcf4 complex, leading to a reduced TRα1 functionality. In this model, TRα1 is recruited to interact with Wnt-responsive element regions in pre-cancerous and cancerous intestinal lesions and stabilizes Wnt effectors on their target genes (Kress  et  al. 2010, Sirakov  et  al. 2012). Remarkably, the Wnt/β-catenin pathway modulates the colonic epithelium T3 concentration through the coordinated

effects of D3 and D2 enzymes (Fig. 2). D3 is a downstream target upregulated by Wnt/β-catenin, while unknown mechanisms downregulate D2. In colon cancer cells, D3 depletion causes intracellular T3 levels to rise, promoting differentiation and reducing proliferation (Dentice  et al. 2012). These observations demonstrate the complexity of the interactions among THs, deiodinases and the Wnt pathway in the balance of cell proliferation and differentiation. Notably, the effects of THs on colorectal cancer stem cells (CSCs) enhance the chemotherapy sensitivity and might be clinically important in the colon cancer therapy (Catalano et al. 2016).

TH and Wnt/β-catenin interactions are also involved in the hepatocellular physiopathology by regulating the cell cycle during development and regeneration in the liver (Francavilla  et al. 1994, Bockhorn  et al. 2007, Lade & Monga 2011). T3 enhances the activation of β-catenin in hepatocytes by increasing its phosphorylation through the activation of protein kinase A (PKA), indicating that T3-PKA-β-catenin crosstalk is essential for normal hepatocyte proliferation (Fanti  et al. 2014). Wnt-β-catenin signaling is constitutively activated in HCC (Ihara et al. 1996) but a contributing role of THs in liver tumor proliferation through this pathway remains to be demonstrated.

Sonic hedgehog (SHH) pathway SHH signaling promotes cell differentiation and organ formation during embryogenesis (McMahon  et  al. 2003). SHH remains active in some organs through adulthood, and the deregulation of this pathway can result in uncontrolled cell proliferation (Pasca di Magliano & Hebrok 2003). Notably, SHH signaling is required not only for cancer initiation but also for growth and survival of several types of cancer (Fan  et  al. 1997, Oro  et  al. 1997, Ruiz i Altaba  et  al. 2002, Pasca di Magliano & Hebrok 2003, Dentice et al. 2007).

Basal cell carcinoma (BCC), the most prevalent cancer in light-skinned individuals, is associated with increased levels of D3, the main TH-inactivating enzyme. SHH, through Gli family zinc finger 2 (Gli2), directly induces D3 expression, which in turn reduces intracellular T3 levels and increases cell proliferation, indicating that D3 overexpression is a major player in BCC progression. Indeed, D3 depletion (or T3 treatment) significantly reduces proliferation and cyclin D1 levels in malignant keratinocytes (Dentice et  al. 2007). T3 treatment or D3 depletion also downregulates miR21, a key miRNA involved in oncogenesis. In an opposite manner, miR21 positively regulates D3 expression in BCC through grainyhead-like

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 7: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R373Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

transcription factor 3 (GRHL3) (Di Girolamo et al. 2016). The crosstalk between the SHH and MAPK pathways for D3 upregulation has also been demonstrated in human PTC cell lines (Romitti  et  al. 2012, 2016). Similarly, D3 depletion reduces cell proliferation and decreases cyclin D1 levels (Romitti et al. 2016). Taken together, these data support the link between D3 overexpression and SHH/Gli2 pathway reactivation, suggesting that decreased intracellular levels of THs may be a critical factor for tumor growth, at least in some types of cancer.

Other less characterized TH effects in neoplastic process TH effects on other signaling pathways have also been described. In T-cell lymphomas (TCL), T3 activates αvβ3 integrin signaling inducing cell proliferation and angiogenesis, in part, via the upregulation of VEGF (Sterle et  al. 2014, Cayrol  et  al. 2015). Interestingly, a paradoxical effect was found in mouse models inoculated with TCLs, in which high circulating levels of THs favored T lymphoma growth, while hypothyroidism promoted tumor dissemination (Sterle et al. 2016). Moreover, in vitro short-term TCL exposure to THs led to proliferation, while a longer treatment increased tumor cell apoptosis (Mihara  et  al. 1999, Sterle et  al. 2016). In embryonic carcinoma cells, T3 treatment decreased the growth rate via the rapid downregulation of E2F1, a key regulator of proliferation. This effect is dependent on the presence of active TRs (Nygard et al. 2003).

Recently, an interaction was demonstrated between TRβ and nuclear corepressor 1 (NCoR), a coregulatory protein that mediates transcriptional repression via certain nuclear receptors. TRβ increases NCoR levels, thus suppressing the transcription of prometastatic genes, whereas decreased NCoR leads to increased tumor growth, invasion and metastasis, suggesting that NCoR is a critical mediator of the suppressive actions of TRβ in tumor growth and metastasis (Martinez-Iglesias et al. 2016a).

Evading growth suppressors

TH and TRs can act as tumor suppressors in specific types of tumors. These TH-mediated effects have been studied mostly in hepatic neoplastic and non-neoplastic cells, where T3 was shown to inhibit cell proliferation and to induce differentiation. T3 has a suppressive effect on the growth of specific liver tumors such as hepatoma, where the proliferative inhibitory effect of T3 is mediated by TGF-β upregulation (Yen  et  al. 2006). T3/TR signaling

mediates Dickkopf 4 (DKK4) expression that inhibits the proliferation and migration of hepatoma cells via blockade of the Wnt signaling pathway (Liao et al. 2012). Similarly, THs inhibit cell proliferation by promoting p21 stability through endoglin upregulation (Lin et al. 2013a). Moreover, in TRα1-overexpressing hepatoma cells, T3/TR signaling promotes inhibition of liver cancer cell growth via downregulation of the ubiquitin-like with PHD and ring finger domains 1 (UHRF1) (Wu et al. 2015).

Interestingly, the treatment of preneoplastic hepatocytes with T3 or GC-1 (a TRβ antagonist) leads to a loss of markers associated with neoplastic processes, such as glutathione S-transferase and gamma glutamyl transpeptidase. Meanwhile, T3 promotes the reacquisition of the activity of glucose 6-phosphatase and adenosine triphosphatase, two enzymes expressed in normal hepatocytes. Notably, the reduction in the number of preneoplastic lesions occurs despite an increase in cell proliferation, indicating that active TRs negatively influence the carcinogenic process through the redifferentiation of preneoplastic hepatocytes (Perra  et  al. 2009). In a similar manner, T3 reduced the tumor development and metastasis rate in rats exposed to cycles of TH therapy. These data suggest that T3 could act as an anticarcinogenic molecule, most likely leading to hepatocyte redifferentiation (Ledda-Columbano et al. 2000).

Similarly, studies evaluating the effect of THs on glioma cell lines demonstrated T3-dependent cell redifferentiation at nearly physiological concentrations of the hormone. Remarkably, more aggressive tumors were more sensitive to the T3 inhibitory effects over cell proliferation, an effect that was mediated, at least in part, by TRα1 overexpression (Liappas et al. 2011). Consistent with these observations, it has been shown that several genes related to neuroblastoma cell differentiation are responsive to THs (Bedo et al. 2011).

On the other hand, TR action on tumor proliferation and metastasis might occur independently of the presence of T3 (Martinez-Iglesias et al. 2016b). Nevertheless, these effects have become increasingly difficult to study, in part due to the heterogeneous expression of TRs among different cancer types (and even within the same tumor type), the presence of TR mutations deregulating downstream pathways, and, as mentioned previously, due to parallel nongenomic effects of T3/T4 on the cellular metabolism (Cheng 2003, Chan & Privalsky 2006). Indeed, TRs, particularly the TRβ isoform, can act

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 8: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R374Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

as tumor suppressors, with a functional loss of TRα1/β promoting tumor development and metastasis (Garcia-Silva & Aranda 2004, Martinez-Iglesias et al. 2009, 2016b, Zhu et al. 2010).

Evading cell death and enabling replicative immortality

TH actions have also been demonstrated in the evasion of programmed cell death, an important feature of neoplastic transformation (Shih et al. 2001, Lin et al. 2007, 2008). In brief, apoptosis can be divided into two major circuits: the extrinsic and intrinsic apoptotic programs. The extrinsic apoptosis pathway involves the interaction of ligands, such as tumor necrosis factor (TNF)-α and Fas ligand, with specific receptors on the cell surface. THs decrease TNF-α, Fas receptor and Fas ligand expression and the activity of caspase-3, thus suppressing apoptosis in non-tumoral models (Laoag-Fernandez  et al. 2004). An anti-apoptotic role of THs is also supported by the effect of T3 on apoptosis regulators. T3 decreases the cellular abundance of caspases and the proapoptotic Bcl-2-associated X protein (BAX) and increases the expression of the anti-apoptotic X-linked inhibitor of apoptosis protein (XIAP) (Zhang et al. 2011, Sterle et al. 2014). When considering the intrinsic apoptosis pathway, there is evidence that T3 administration protects hypothyroid rat liver cells from apoptosis induced by oxidative stress in a non-tumoral model (Mukherjee et al. 2014). TH also regulates proteins involved in the intrinsic apoptosis pathway. For example, T3 induces the expression of myeloid cell leukemia 1 (MCL-1), a Bcl-2-related protein located in the outer mitochondrial membrane (Pietrzak & Puzianowska-Kuznicka 2008), while T4 downregulates expression of the BAX gene, the gene product of which is proapoptotic mitochondria. These anti-apoptotic effects of THs are in accordance with the evidence that molecules inhibiting T4 action (tetrac/nanotetrac) have proapoptotic effects on tumor growth (Yalcin et al. 2010a).

The nongenomic effects of T4 in the apoptotic pathway occur, at least in part, via induction of the MAPK pathway, initiated through the integrin αVβ3 receptor (Lin et al. 2007). The T4-induced MAPK activation results in the serine phosphorylation of the oncogene suppressor p53, STAT1α, STAT-3 and TRβ1, leading to proliferative and anti-apoptotic effects (Lin et  al. 1999b, 2003, Davis et al. 2000, Shih et al. 2001). The T4 anti-apoptotic effect was demonstrated in human PTC and FTC cell lines incubated with resveratrol (RV), an apoptosis inducer that also initiates signaling via the plasma membrane integrin

αVβ3 (Lin et al. 2007, 2008). In glioma cells, RV increases the nuclear content of cyclooxygenase-2 (COX2) via MAPK induction, while the incubation of RV-treated cells with T4 decreases the levels of the cytosolic proapoptotic protein B-cell lymphoma extra-large (Bcl-xL) and the formation of nuclear complexes between pERK and COX2. These effects lead to a blockage of p53 phosphorylation, thus inhibiting apoptosis (Lin et  al. 2007). However, others have demonstrated that high concentrations of T3 induce breast cancer cell apoptosis via the TRβ-dependent downregulation of the anti-apoptotic senescence marker protein-30 gene (SMP30) (Sar et al. 2011). The involvement of TRβ in apoptotic pathways is further supported by studies showing that TRβ can act as a tumor suppressor, interfering with the recruitment of retinoblastoma protein and p53 via the SV40Tag oncoprotein through a protein–protein interaction (Kim et  al. 2012). TNF-related apoptosis-inducing ligand (TRAIL/Apo2L) is a potent effector of tumorigenesis that not only promotes apoptosis but also triggers non-apoptotic pathways (Johnstone et al. 2008). T3 upregulates TRAIL expression at the transcriptional level in TR-overexpressing hepatoma cells, which in turn promotes cell migration and invasion (Chi et al. 2012).

Compilation of data supports the anti-apoptotic activity of THs in several tumor cells. TH action occurs mainly through physiological levels of T4 via genomic and nongenomic signaling modulating multiple components of the extrinsic and intrinsic apoptosis pathways.

The maintenance of telomere integrity and telomerase protect cells from apoptosis. Telomerase inhibition elicits an apoptotic response in cancer cells, while restoration of telomerase activity in somatic cells promotes resistance to apoptosis (Mondello & Scovassi 2004). Thus far, no studies on the effect of THs on telomerase activity in cancer models have been reported. However, hypothyroidism leads to decreased telomerase activity in stem cells (Simsek et al. 2014), an observation that should be further explored.

Tissue invasion and metastasis

The spread of cells from the primary lesion to distant organs is the most worrisome aspect of cancer. Alterations in cell shape and in their attachment to both other cells and the extracellular matrix (ECM) are essential for this process (Fidler 2003). Tumor cells must invade the basement membrane and migrate through the ECM surrounding the tumor epithelium to spread, which occurs

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 9: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R375Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

mainly via interactions between integrin receptors and ECM components. Matrix metalloproteinase-9 (MMP-9) is a pivotal matrix metalloproteinase that contributes to ECM degradation, thereby enhancing invasiveness (Egeblad & Werb 2002). THs contribute to the regulation of cell adhesion and migration in several tumor models (Dietrich  et al. 2000, Liao  et al. 2010) For instance, THs induce MMP-9 via the αVβ3-MAPK pathway, promoting increased adhesion to fibronectin and enhancing cell migration in myeloma cells (Cohen et al. 2014).

THs status might influence the spread of liver cell cancer. However, as already mentioned, the effects of THs on liver tumorigenesis are complex and depend on TR expression status, cancer stage and other co-effectors present in the tumor microenvironment (Wu  et  al. 2013). Acting mostly through TRs, actions of TH on HCC development may lead to the suppression or promotion of prometastatic mechanisms. T3 enhances HCC cell invasion in vitro and in vivo (Wu et  al. 2013). T3 treatment increases the invasive capacity of HepG2 cells expressing TRs, possibly due to the upregulation of furin, a calcium-dependent serine endoprotease, which increases the processing of MMP-2 and MMP-9. Moreover, T3 administration to mice inoculated with HepG2-TRα1 cells caused furin overexpression. Notably, these animals displayed greater tumor sizes and metastasis rates than euthyroid animals, supporting the metastasis-promoting effect of T3 in HCC (Chen et al. 2008). Several members of the MMP family, including MMP-2, MMP-9 and MMP-7, are upregulated upon r-TRAIL stimulation in hepatoma cells, an effect confirmed by increased invasiveness in both in vitro and in vivo models (Chi et al. 2012). Cathepsin H, a protease involved in the degradation of ECM components, leading to cancer cell migration and metastasis, is induced by T3 in HCC cells, enhancing the invasion potential of hepatoma cells in vitro and in vivo (Wu  et  al. 2011). Likewise, T3 treatment in HCC cells also enhanced tumor cell migration and invasion by stimulating the overexpression of brain-specific serine protease 4 protein levels, which was associated with ERK1/2-C/EBPβ-VEGF cascade activation (Chen  et  al. 2014). Inversely, other studies have demonstrated that T3 treatment of the same cells leads to spondin 2 overexpression, which inhibits cell invasion and migration (Liao et  al. 2010). T3 treatment also upregulates the expression of DKK4 protein, an antagonist of Wnt, in HepG2 TR-expressing cells (Chi et al. 2013), suggesting that the T3 upregulation of the TR/DKK4/Wnt/β-catenin cascade inhibits the metastasis of hepatoma cells (Liao et al. 2012).

T3-induced cell migration in HCC is mediated in part to a reduction in miR-17 and miR-130b expression (Lin et al. 2013b, 2015) and the overexpression of miR-21 (Huang  et  al. 2013). The overexpression of miR-17 markedly inhibits HCC cell migration and invasion in vitro and in vivo via the suppression of MMP-3 (Lin et al. 2013b), whereas the effect of miR-130b involves the regulation of genes critical for metastasis, such as MMP-9, mTOR, ERK1/2, AKT and STAT-3 (Lin et al. 2015).

Breast cancer cell migration is also influenced by the nongenomic action of T3. The focal adhesion kinase (FAK) protein is an essential regulator of the actin cytoskeleton, thus modulating the steps involved in cell migration and invasion. T3, acting through integrin αVβ3, promotes the phosphorylation of FAK by activating the Src/FAK/PI3K pathway, thereby modulating cell adhesion and migration (Flamini et al. 2017).

Induction of angiogenesis

Tumor growth, invasion and metastasis are strongly dependent on angiogenesis (Folkman 1995). The initiation and maintenance of a vascular supply involve the local release of angiogenic molecules, such as VEGF, FGF2, PDGF, TGFs and angiopoietins (Angs) (Risau 1997). The concept of TH-induced neovascularization was first described a decade ago in the chick chorioallantoic membrane assay of angiogenesis (Davis et al. 2004, Bergh et al. 2005). TH pro-angiogenic effects seem to be mainly promoted by T4 binding to integrin αVβ3, followed by MAPK signal transduction. The TH-αvβ3 complex causes the transcription of several factors, such as TRβ1, ERs, TP53 and STATs, leading to the increased expression of angiogenic modulators, such as FGF2, VEGF and Ang-2 (Mousa  et  al. 2005, 2014, Bockhorn et  al. 2007, Davis et al. 2009). The addition of T3 to cultures of HCC, lung and kidney carcinoma cells leads to HIF-1α induction and increases in VEGF levels (Otto & Fandrey 2008). T3 upregulates HIF-1α through the PI3K pathway, which in turn stimulates the secretion of HIF-responsive genes, such as VEGF, FGF2, interleukin-6, stromal cell-derived factor-1 and TGF-β1 (Davis  et  al. 2011). T3 and T4 also regulate the differentiation and migration of mesenchymal stem cells (MSCs) via integrin αVβ3. This regulation affects not only indicators of tissue remodeling and invasion, such as tenascin-C (TN-C) and thrombospondin-1 (TSP1), but also proteins associated with angiogenesis, such as α-smooth muscle actin (α-SMA), desmin and VEGF, thus contributing to tumor stroma dysregulation (Schmohl et al. 2015).

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 10: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R376Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

Of note, tetrac reduces VEGF-A mRNA levels while increases the transcripts of the THBS1 gene, an adhesive glycoprotein that mediates cell-to-cell and cell-to-matrix interactions, blocking the T4 proangiogenic effects (Glinskii  et  al. 2009, Yalcin  et  al. 2010b). Indeed, tetrac administration to nude mice inoculated with FTC or medullary thyroid carcinoma (MTC) cells reduces the vascularization and growth of grafted tumors (Yalcin et  al. 2010a,b). The tetrac-associated inhibition of angiogenesis has been observed in a variety of tumor xenografts, indicating a therapeutic potential that merits exploration in clinical settings (Yalcin  et al. 2009, 2013, Mousa et al. 2012).

Genomic instability and cellular senescence

Genomic instability is a hallmark of most cancer cells. Failure in maintaining DNA integrity impairs cell proliferation and survival, resulting in senescence, a phenomenon in which normal cells cease to divide. Cells can be induced to senesce via DNA damage due to increased ROS levels (Campisi 2013). T3, mediated by TRβ, induces senescence in mouse embryonic fibroblasts, promoting DNA damage secondary to oxidative stress. The effect is dependent on the activation of ataxia telangiectasia mutated (ATM)/adenosine monophosphate-activated protein kinase (PRKAA), proteins that play pivotal roles in detecting genomic damage (Zambrano et al. 2014). Of note, TRβ1 and TRβ2 are highly expressed in retinoblastoma cells, and participate in maintaining genomic stability (Pappas et al. 2017).

Dysregulation of cell bioenergetics/energy metabolism

The sustenance of cancer cells also depends on metabolic adaptations. Tumor cells are characterized by increased aerobic glycolysis and lactic acid production in normoxic conditions. This phenomenon, which has been a biochemical hallmark of cancer for decades, is known as the Warburg effect (Koppenol  et  al. 2011, Warburg 1956). Lately, some studies have established a connection between the mitochondrial and TH metabolisms in the context of modulating the Warburg phenomenon in breast cancer (Suhane & Ramanujan 2011, Silvestri et al. 2014). The authors evaluated the effects of T3 in modulating the bioenergetics profiles by monitoring glucose uptake, lactate generation and the mitochondrial oxygen consumption rate. Interestingly, they showed that T3 directly increases the mitochondrial metabolism in

aggressive breast cancer cells and directly regulates one of the isoforms of pyruvate kinase that is vital for sustaining the Warburg effect (Suhane & Ramanujan 2011).

Oxidative stress is known to disrupt the function of deiodinases (Wajner  et  al. 2011), key enzymes for the regulation of the intracellular levels of active THs (St  Germain  et  al. 2009, Maia et  al. 2011). Neoplastic cells are known to be hypoxic, a condition that has been shown to upregulate D3 expression through HIF-1 in non-tumoral models (Simonides et al. 2008, Ciavardelli et al. 2014). D3 reactivation in the neoplastic cells of solid tumors increases TH inactivation and reduces the metabolic rate, which may favor cell proliferation. This phenomenon has been associated with a poor therapeutic response and an increased risk of recurrence (Keith & Simon 2007). In a non-tumoral model of rat brain, D3 participates in the hypoxia-induced reduction in TH signaling. Moreover, ischemia/hypoxia induces a heat-shock protein 40 (HSP40)-mediated translocation of D3 to the nucleus, facilitating TH inactivation proximal to the TH receptors (Jo et al. 2012, Huang et al. 2014). THs can directly protect or damage cells by modulating oxidative stress (Mancini  et  al. 2016). Thus, it is reasonable to consider that intracellular TH levels contribute to the disruption of tumoral bioenergetics. The effects of THs on glycolytic fueling require further exploration since common pathways appear to be activated in several tumors (DeBerardinis et al. 2008).

Intracellular microenvironment: deiodinase control over TH status

The intracellular TH status is highly dependent on the activation or inactivation of THs by deiodinases. Particularly, alterations in the balance between TH-activating and TH-inactivating deiodinases can be critical in modulating the balance between cell proliferation and differentiation (Kress et  al. 2009b, Dentice et  al. 2013). Indeed, changes in the expression levels of deiodinases are present in several malignant human neoplasias. DIO1 downregulation occurs in renal, lung, hepatic and prostate cancer tissues (Dutkiewicz et al. 1995, Sabatino  et  al. 2000, Pachucki  et  al. 2001, Wawrzynska et al. 2003). Studies performed using human PTC samples found a consistent decrease in DIO1 levels compared with the surrounding thyroid tissue, suggesting that diminished DIO1 expression might be an early event in thyroid cell dedifferentiation. In contrast, DIO1 and D1 activity levels are increased in follicular adenoma and FTC

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 11: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R377Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

samples (de Souza Meyer  et al. 2005). In renal clear cell cancer, miR-224 expression correlates negatively with the DIO1 mRNA level and T3 concentration, suggesting that miR-224 induces intracellular hypothyroidism via reduced D1 function (Boguslawska  et al. 2011). Interestingly, D1 activity does not differ significantly between benign and malignant tumors as compared with healthy liver parenchyma cells (Kornasiewicz et al. 2014). In contrast, D1 activity in non-cancerous breast tissues is very low or non-measurable, whereas it is increased in breast cancer, indicating a tissue-specific regulation of D1 expression (Debski et al. 2007).

Changes in DIO2 expression have also been demonstrated in several human neoplasias. DIO2 expression is induced in most brain tumors, including those derived from glial cells (Mori  et  al. 1993, Murakami  et  al. 2000, Nauman  et  al. 2004), FTC cells and MTC cells (Kim  et  al. 2003, Meyer  et  al. 2008). In contrast, DIO2 mRNA and activity are decreased in PTC cells as compared with normal follicular thyroid cells (Arnaldi et al. 2005, de Souza Meyer et al. 2005).

Increased DIO3 expression is observed in several human tumor types, including astrocytoma, oligodendroglioma, glioblastoma multiforme and BCC (Gereben  et  al. 2008). Tumoral D3 activity is markedly elevated in vascular tumors, including infantile hemangioma and hemangioendothelioma in adults (Huang et al. 2000, 2002, Luongo et al. 2013), even to the extent of inducing clinical hypothyroidism (consumptive hypothyroidism). Opposing regulation of DIO3 or DIO1/DIO2 expression has been reported in various human neoplasias, such as PTC, TSH tumors, BCC and colon cancer (de Souza Meyer et  al. 2005, Dentice et  al. 2007, 2012, Luongo et  al. 2013, Romitti et  al. 2016). Studies performed using 105 pituitary tumors demonstrated that D2 and D3 mRNA levels were significantly augmented in pituitary tumors compared with normal pituitary tissue. In the rare TSH-secreting pituitary tumor subtype, increased D3 expression and D2 mRNA downregulation were observed, which may explain the ‘resistance’ of these tumors to TH feedback (Tannahill et al. 2002). In human BCC samples, upregulated DIO3 expression correlated with the functional status of the SHH pathway described above, which is a critical oncogenic pathway (Dentice et  al. 2007). Interestingly, co-expression of D3 and D2 was found in BCC, and manipulation of the expression of each enzyme, with consequent alteration of intracellular TH levels, dramatically modifies the proliferative potential of BCC (Miro et  al. 2017). This illustrates the critical regulatory role of THs on proliferation of certain tumors.

The induction of DIO3 expression was also recently demonstrated in human PTC samples. Remarkably, D3 levels were positively associated with increased tumor size and increased rates of local and distant metastasis at diagnosis (Romitti et  al. 2012). Most interesting, D3 upregulation in PTC samples is modulated by crosstalk between the MAPK and SHH pathways and varies according to the genetic alterations in this tumor type (Romitti et al. 2016). Increased DIO3 expression was also observed in FTC but not in medullary or anaplastic thyroid carcinoma samples (Romitti et al. 2012). Higher levels of D3 were also detected in human intestinal adenoma and carcinoma compared with healthy intestinal tissue. However, DIO3 expression was reduced in lesions with higher histological grades (Dentice et al. 2012).

Tumor microenvironment

Increasing evidence indicates that what is occurring inside tumor cells depends on exogenous stimuli originating around the tumor cells (Joyce & Pollard 2009, Goubran  et  al. 2014). Specifically, surrounding tumor stroma and immune cells can be ‘activated,’ thus influencing tumor behavior.

Evading immune destruction and promoting inflammation

The immune system antagonizes and enhances tumor development and progression. The tumor-associated inflammatory response has the paradoxical effects of promoting tumorigenesis and helping neoplastic cells acquire hallmark capabilities (Colotta  et  al. 2009, Grivennikov  et  al. 2010). The endocrine and immune systems are complexly interconnected, and THs affect immune cells, modulating their responses (De Vito et al. 2011).

THs seem to enhance the antiviral action of interferon-γ via the MAPK pathway (Lin  et  al. 1996). Moreover, T3 activates PI3K/AKT signaling, thus activating myeloid cell leukemia-1 (MCL1) (Pietrzak & Puzianowska-Kuznicka 2008) and the HIF1A gene (Lin et  al. 2009), which are critical molecules that elicit the immune response.

In vitro models have shown that T3 promotes tumor growth through the modulation of soluble factors released by surrounding microglial cells (Perrotta  et al. 2015). In contrast, the T3–TRβ complex influences the antitumor responses of dendritic cells (DCs), the main antigen-presenting cells during tumor growth when activated by T cells (Alamino et al. 2015). This TH effect seems to depend on AKT activation (Mascanfroni  et  al. 2010), while AKT

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 12: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R378Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

phosphorylation enhances DC survival (Park et al. 2006). In addition to the complex effects of THs on T lymphoma cell proliferation and death, Sterle’s group has investigated thyroid status in the tumor microenvironment (Sterle et al. 2016). They found that THs have a substantial effect on the distribution of different immune cell populations and on lymphocyte infiltration, particularly on the prevalence of cytotoxic T cells. Together, these results highlight the importance of THs in modulating the immune response and related signaling in the tumor milieu through different pathways.

Cancer stem cells (CSCs)

CSCs may be involved in tumor initiation and may drive tumor progression. They carry oncogenic and tumor suppressor mutations that genetically define the disease. Both T3 and T4 increase the migration of MSCs toward tumor signals and increase the invasion of MSCs into tumor cell spheroids, thus impacting crucial steps of tumor stroma formation (Schmohl et al. 2015). In a model of HCC CSCs, T4 was a potent promoter of CSC self-renewal. TH signaling in HCC occurs through the nuclear receptor TRα with the cooperation of NF-κB, inducing the expression of stem cell genes, such as CD44, BMI1, NOTCH1 and HIF1A, thus enhancing the self-renewal of HCC CSCs (Wang  et  al. 2016). However, evidence of impact of TH on CSCs remains scarce.

Conclusion and future directions

In conclusion, an extensive set of data has indicated that the status of THs plays a significant role in the carcinogenesis process. Changes in TH levels seem to occur due to a disruption in TR and/or deiodinase expression and via nongenomic signaling pathways that broadly contribute to the acquisition of steps necessary for cancer development. TH status alterations are known to contribute to cancer development and/or progression via direct effects on virtually all the hallmarks of cancer. Therefore, adjuvant therapies targeting TH actions might be considered alternative treatments for cancer cell proliferation, metastasis and angiogenesis.

The genomic and nongenomic actions of THs overlap in the regulation of pro- and anti-tumoral cascades that lead to cancer growth. THs have a wide effect on tumoral progression, contributing to the acquisition of all hallmarks of cancer by predisposed cells. Moreover, intracellular TH changes due to a disruption in deiodinase status seem to be critical for modulating cell proliferation

and differentiation. Accordingly, experimental and observational studies indicate TH status imbalance as a risk factor for several neoplasias. Furthermore, clinical trials have demonstrated that induced hypothyroidism leads to extended survival in different types of cancer (Hercbergs et al. 2003, 2015). Targeting cancer pathways to control tumor dissemination has been studied through integrin αVβ3 blockade, in an effort to inhibit angiogenesis. Pharmacologically targeting the membrane receptor with tetrac and other derivatives inhibits the trophic effects of the hormone in some cancer cells (Rebbaa et al. 2008, Yalcin et  al. 2010b). Moreover, targeting the SHH pathway in BCC inhibited proliferation in clinical settings (Sekulic et al. 2012, Tang et al. 2012), although the direct effect on D3 activity was not analyzed. Theoretically, the pharmacological modulation of intracellular TH levels in a cell-specific manner could contribute to cancer treatments. In the same way, blocking pathways abnormally activated by THs, without interfering with the systemic balance of the TH metabolism, could lead to proapoptotic and anti-proliferative actions to control tumor growth or enhance the effectiveness of existing chemotherapeutic cancer drugs.

Declaration of interestThe authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of this review.

FundingThis work was supported by the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) (457547/2013-8); Fundação de Amparo a Pesquisa do Rio Grande do Sul (FAPERGS) (10/0051-9) and Fundo de Incentivo a Pesquisa do Hospital de Clínicas de Porto Alegre (FIPE) (16-0246), Brasil.

References

Alamino VA, Mascanfroni ID, Montesinos MM, Gigena N, Donadio AC, Blidner AG, Milotich SI, Cheng SY, Masini-Repiso AM, Rabinovich GA, et al. 2015 Antitumor responses stimulated by dendritic cells are improved by triiodothyronine binding to the thyroid hormone receptor beta. Cancer Research 75 1265–1274. (doi:10.1158/0008-5472.CAN-14-1875)

Aoki T, Tsunekawa K, Araki O, Ogiwara T, Nara M, Sumino H, Kimura T & Murakami M 2015 Type 2 iodothyronine deiodinase activity is required for rapid stimulation of PI3K by thyroxine in human umbilical vein endothelial cells. Endocrinology 156 4312–4324. (doi:10.1210/en.2014-1988)

Aranda A, Martinez-Iglesias O, Ruiz-Llorente L, Garcia-Carpizo V & Zambrano A 2009 Thyroid receptor: roles in cancer. Trends in Endocrinology and Metabolism 20 318–324. (doi:10.1016/j.tem.2009.03.011)

Aranda A & Pascual A 2001 Nuclear hormone receptors and gene expression. Physiological Reviews 81 1269–1304.

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 13: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R379Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

Arnaldi LA, Borra RC, Maciel RM & Cerutti JM 2005 Gene expression profiles reveal that DCN, DIO1, and DIO2 are underexpressed in benign and malignant thyroid tumors. Thyroid 15 210–221. (doi:10.1089/thy.2005.15.210)

Barbakadze T, Natsvlishvili N & Mikeladze D 2014 Thyroid hormones differentially regulate phosphorylation of ERK and Akt via integrin alphavbeta3 receptor in undifferentiated and differentiated PC-12 cells. Cell Biochemistry and Function 32 282–286. (doi:10.1002/cbf.3013)

Beatson GT 1896 On the treatment of inoperable cases of carcinoma of the mamma: suggestions for a new method of treatment, with illustrative cases. Lancet 148 162–165. (doi:10.1016/S0140-6736(01)72384-7)

Bedo G, Pascual A & Aranda A 2011 Early thyroid hormone-induced gene expression changes in N2a-beta neuroblastoma cells. Journal of Molecular Neuroscience 45 76–86. (doi:10.1007/s12031-010-9389-y)

Bergh JJ, Lin HY, Lansing L, Mohamed SN, Davis FB, Mousa S & Davis PJ 2005 Integrin alphaVbeta3 contains a cell surface receptor site for thyroid hormone that is linked to activation of mitogen-activated protein kinase and induction of angiogenesis. Endocrinology 146 2864–2871. (doi:10.1210/en.2005-0102)

Bockhorn M, Frilling A, Benko T, Best J, Sheu SY, Trippler M, Schlaak JF & Broelsch CE 2007 Tri-iodothyronine as a stimulator of liver regeneration after partial and subtotal hepatectomy. European Surgical Research 39 58–63. (doi:10.1159/000098443)

Boelaert K, Horacek J, Holder RL, Watkinson JC, Sheppard MC & Franklyn JA 2006 Serum thyrotropin concentration as a novel predictor of malignancy in thyroid nodules investigated by fine-needle aspiration. Journal of Clinical Endocrinology and Metabolism 91 4295–4301. (doi:10.1210/jc.2006-0527)

Boguslawska J, Wojcicka A, Piekielko-Witkowska A, Master A & Nauman A 2011 MiR-224 targets the 3′UTR of type 1 5′-iodothyronine deiodinase possibly contributing to tissue hypothyroidism in renal cancer. PLoS ONE 6 e24541. (doi:10.1371/journal.pone.0024541)

Brazil DP & Hemmings BA 2001 Ten years of protein kinase B signalling: a hard Akt to follow. Trends in Biochemical Sciences 26 657–664. (doi:10.1016/S0968-0004(01)01958-2)

Campisi J 2013 Aging, cellular senescence, and cancer. Annual Review of Physiology 75 685–705. (doi:10.1146/annurev-physiol-030212-183653)

Cao X, Kambe F, Moeller LC, Refetoff S & Seo H 2005 Thyroid hormone induces rapid activation of Akt/protein kinase B-mammalian target of rapamycin-p70S6K cascade through phosphatidylinositol 3-kinase in human fibroblasts. Molecular Endocrinology 19 102–112. (doi:10.1210/me.2004-0093)

Cao X, Kambe F, Yamauchi M & Seo H 2009 Thyroid-hormone-dependent activation of the phosphoinositide 3-kinase/Akt cascade requires Src and enhances neuronal survival. Biochemical Journal 424 201–209. (doi:10.1042/BJ20090643)

Catalano V, Dentice M, Ambrosio R, Luongo C, Carollo R, Benfante A, Todaro M, Stassi G & Salvatore D 2016 Activated thyroid hormone promotes differentiation and chemotherapeutic sensitization of colorectal cancer stem cells by regulating Wnt and BMP4 signaling. Cancer Research 76 1237–1244. (doi:10.1158/0008-5472. CAN-15-1542)

Cayrol F, Diaz Flaque MC, Fernando T, Yang SN, Sterle HA, Bolontrade M, Amoros M, Isse B, Farias RN, Ahn H, et al. 2015 Integrin alphavbeta3 acting as membrane receptor for thyroid hormones mediates angiogenesis in malignant T cells. Blood 125 841–851. (doi:10.1182/blood-2014-07-587337)

Chan IH & Privalsky ML 2006 Thyroid hormone receptors mutated in liver cancer function as distorted antimorphs. Oncogene 25 3576–3588. (doi:10.1038/sj.onc.1209389)

Chen RN, Huang YH, Lin YC, Yeh CT, Liang Y, Chen SL & Lin KH 2008 Thyroid hormone promotes cell invasion through activation of furin expression in human hepatoma cell lines. Endocrinology 149 3817–3831. (doi:10.1210/en.2007-0989)

Chen CY, Chung IH, Tsai MM, Tseng YH, Chi HC, Tsai CY, Lin YH, Wang YC, Chen CP, Wu TI, et al. 2014 Thyroid hormone enhanced human hepatoma cell motility involves brain-specific serine protease 4 activation via ERK signaling. Molecular Cancer 13 162. (doi:10.1186/1476-4598-13-162)

Cheng SY 2003 Thyroid hormone receptor mutations in cancer. Molecular and Cellular Endocrinology 213 23–30. (doi:10.1016/j.mce.2003.10.051)

Cheng SY, Leonard JL & Davis PJ 2010 Molecular aspects of thyroid hormone actions. Endocrine Reviews 31 139–170. (doi:10.1210/er.2009-0007)

Chi HC, Chen SL, Liao CJ, Liao CH, Tsai MM, Lin YH, Huang YH, Yeh CT, Wu SM, Tseng YH, et al. 2012 Thyroid hormone receptors promote metastasis of human hepatoma cells via regulation of TRAIL. Cell Death and Differentiation 19 1802–1814. (doi:10.1038/cdd.2012.58)

Chi HC, Liao CH, Huang YH, Wu SM, Tsai CY, Liao CJ, Tseng YH, Lin YH, Chen CY, Chung IH, et al. 2013 Thyroid hormone receptor inhibits hepatoma cell migration through transcriptional activation of Dickkopf 4. Biochemical and Biophysical Research Communications 439 60–65. (doi:10.1016/j.bbrc.2013.08.028)

Ciavardelli D, Bellomo M, Crescimanno C & Vella V 2014 Type 3 deiodinase: role in cancer growth, stemness, and metabolism. Frontiers in Endocrinology 5 215. (doi:10.3389/fendo.2014.00215)

Ciccia A & Elledge SJ 2010 The DNA damage response: making it safe to play with knives. Molecular Cell 40 179–204. (doi:10.1016/j.molcel.2010.09.019)

Cohen K, Flint N, Shalev S, Erez D, Baharal T, Davis PJ, Hercbergs A, Ellis M & Ashur-Fabian O 2014 Thyroid hormone regulates adhesion, migration and matrix metalloproteinase 9 activity via alphavbeta3 integrin in myeloma cells. Oncotarget 5 6312–6322. (doi:10.18632/oncotarget.2205)

Cohen K, Ellis M, Shinderman E, Khoury S, Davis PJ, Hercbergs A & Ashur-Fabian O 2015 Relevance of the thyroid hormones-alphavbeta3 pathway in primary myeloma bone marrow cells and to bortezomib action. Leukemia and Lymphoma 56 1107–1114. (doi:10.3109/10428194.2014.947612)

Colotta F, Allavena P, Sica A, Garlanda C & Mantovani A 2009 Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis 30 1073–1081. (doi:10.1093/carcin/bgp127)

Davis PJ, Shih A, Lin HY, Martino LJ & Davis FB 2000 Thyroxine promotes association of mitogen-activated protein kinase and nuclear thyroid hormone receptor (TR) and causes serine phosphorylation of TR. Journal of Biological Chemistry 275 38032–38039. (doi:10.1074/jbc.M002560200)

Davis FB, Mousa SA, O’Connor L, Mohamed S, Lin HY, Cao HJ & Davis PJ 2004 Proangiogenic action of thyroid hormone is fibroblast growth factor-dependent and is initiated at the cell surface. Circulation Research 94 1500–1506. (doi:10.1161/01.RES.0000130784.90237.4a)

Davis FB, Tang HY, Shih A, Keating T, Lansing L, Hercbergs A, Fenstermaker RA, Mousa A, Mousa SA, Davis PJ, et al. 2006 Acting via a cell surface receptor, thyroid hormone is a growth factor for glioma cells. Cancer Research 66 7270–7275. (doi:10.1158/0008-5472.CAN-05-4365)

Davis PJ, Leonard JL & Davis FB 2008 Mechanisms of nongenomic actions of thyroid hormone. Frontiers in Neuroendocrinology 29 211–218. (doi:10.1016/j.yfrne.2007.09.003)

Davis PJ, Davis FB & Mousa SA 2009 Thyroid hormone-induced angiogenesis. Current Cardiology Reviews 5 12–16. (doi:10.2174/ 157340309787048158)

Davis PJ, Davis FB, Mousa SA, Luidens MK & Lin HY 2011 Membrane receptor for thyroid hormone: physiologic and pharmacologic implications. Annual Review of Pharmacology and Toxicology 51 99–115. (doi:10.1146/annurev-pharmtox-010510-100512)

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 14: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R380Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

Davis PJ, Glinsky GV, Lin HY, Leith JT, Hercbergs A, Tang HY, Ashur-Fabian O, Incerpi S & Mousa SA 2014a Cancer cell gene expression modulated from plasma membrane integrin alphavbeta3 by thyroid hormone and nanoparticulate tetrac. Frontiers in Endocrinology 5 240. (doi:10.3389/fendo.2014.00240)

Davis PJ, Lin HY, Sudha T, Yalcin M, Tang HY, Hercbergs A, Leith JT, Luidens MK, Ashur-Fabian O, Incerpi S, et al. 2014b Nanotetrac targets integrin alphavbeta3 on tumor cells to disorder cell defense pathways and block angiogenesis. OncoTargets and Therapy 7 1619–1624. (doi:10.2147/ott.s67393)

de Souza AC, Justo GZ, de Araujo DR & Cavagis AD 2011 Defining the molecular basis of tumor metabolism: a continuing challenge since Warburg’s discovery. Cellular Physiology and Biochemistry 28 771–792. (doi:10.1159/000335792)

de Souza Meyer EL, Dora JM, Wagner MS & Maia AL 2005 Decreased type 1 iodothyronine deiodinase expression might be an early and discrete event in thyroid cell dedifferentation towards papillary carcinoma. Clinical Endocrinology 62 672–678. (doi:10.1111/j.1365-2265.2005.02277.x)

De Vito P, Incerpi S, Pedersen JZ, Luly P, Davis FB & Davis PJ 2011 Thyroid hormones as modulators of immune activities at the cellular level. Thyroid 21 879–890. (doi:10.1089/thy.2010.0429)

DeBerardinis RJ, Lum JJ, Hatzivassiliou G & Thompson CB 2008 The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metabolism 7 11–20. (doi:10.1016/j.cmet.2007.10.002)

Debski MG, Pachucki J, Ambroziak M, Olszewski W & Bar-Andziak E 2007 Human breast cancer tissue expresses high level of type 1 5′-deiodinase. Thyroid 17 3–10. (doi:10.1089/thy.2006.0012)

Dentice M, Luongo C, Huang S, Ambrosio R, Elefante A, Mirebeau-Prunier D, Zavacki AM, Fenzi G, Grachtchouk M, Hutchin M, et al. 2007 Sonic hedgehog-induced type 3 deiodinase blocks thyroid hormone action enhancing proliferation of normal and malignant keratinocytes. PNAS 104 14466–14471. (doi:10.1073/pnas.0706754104)

Dentice M, Ambrosio R & Salvatore D 2009 Role of type 3 deiodinase in cancer. Expert Opinion on Therapeutic Targets 13 1363–1373. (doi:10.1517/14728220903339122)

Dentice M, Luongo C, Ambrosio R, Sibilio A, Casillo A, Iaccarino A, Troncone G, Fenzi G, Larsen PR & Salvatore D 2012 beta-Catenin regulates deiodinase levels and thyroid hormone signaling in colon cancer cells. Gastroenterology 143 1037–1047. (doi:10.1053/j.gastro.2012.06.042)

Dentice M, Marsili A, Zavacki A, Larsen PR & Salvatore D 2013 The deiodinases and the control of intracellular thyroid hormone signaling during cellular differentiation. Biochimica et Biophysica Acta 1830 3937–3945. (doi:10.1016/j.bbagen.2012.05.007)

Dewhirst MW, Cao Y & Moeller B 2008 Cycling hypoxia and free radicals regulate angiogenesis and radiotherapy response. Nature Reviews Cancer 8 425–437. (doi:10.1038/nrc2397)

Di Cristofano A & Pandolfi PP 2000 The multiple roles of PTEN in tumor suppression. Cell 100 387–390. (doi:10.1016/S0092-8674(00)80674-1)

Di Girolamo D, Ambrosio R, De Stefano MA, Mancino G, Porcelli T, Luongo C, Di Cicco E, Scalia G, Vecchio LD, Colao A, et al. 2016 Reciprocal interplay between thyroid hormone and microRNA-21 regulates hedgehog pathway-driven skin tumorigenesis. Journal of Clinical Investigation 126 2308–2320. (doi:10.1172/JCI84465)

Dietrich JB, Zaepfel M & Kuchler-Bopp S 2000 Expression of intercellular adhesion molecule-1 in C6 glioma cells is up-regulated by thyroid hormone. Neuroreport 11 2855–2860. (doi:10.1097/00001756-200009110-00006)

Downward J 2003 Targeting RAS signalling pathways in cancer therapy. Nature Reviews Cancer 3 11–22. (doi:10.1038/nrc969)

Dutkiewicz S, Witeska A & Nauman A 1995 The deiodination of thyroxine to triiodothyronine in the testes of patients with prostate

cancer. International Urology and Nephrology 27 81–85. (doi:10.1007/BF02575224)

Egeblad M & Werb Z 2002 New functions for the matrix metalloproteinases in cancer progression. Nature Reviews Cancer 2 161–174. (doi:10.1038/nrc745)

Evan GI & Vousden KH 2001 Proliferation, cell cycle and apoptosis in cancer. Nature 411 342–348. (doi:10.1038/35077213)

Fan H, Oro AE, Scott MP & Khavari PA 1997 Induction of basal cell carcinoma features in transgenic human skin expressing Sonic Hedgehog. Nature Medicine 3 788–792. (doi:10.1038/nm0797-788)

Fanti M, Singh S, Ledda-Columbano GM, Columbano A & Monga SP 2014 Tri-iodothyronine induces hepatocyte proliferation by protein kinase A-dependent beta-catenin activation in rodents. Hepatology 59 2309–2320. (doi:10.1002/hep.26775)

Fidler IJ 2003 The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nature Reviews Cancer 3 453–458. (doi:10.1038/nrc1098)

Filmus J, Robles AI, Shi W, Wong MJ, Colombo LL & Conti CJ 1994 Induction of cyclin D1 overexpression by activated ras. Oncogene 9 3627–3633.

Fiore E, Rago T, Provenzale MA, Scutari M, Ugolini C, Basolo F, Di Coscio G, Berti P, Grasso L, Elisei R, et al. 2009 Lower levels of TSH are associated with a lower risk of papillary thyroid cancer in patients with thyroid nodular disease: thyroid autonomy may play a protective role. Endocrine-Related Cancer 16 1251–1260. (doi:10.1677/ERC-09-0036)

Flamini MI, Uzair ID, Pennacchio GE, Neira FJ, Mondaca JM, Cuello-Carrion FD, Jahn GA, Simoncini T & Sanchez AM 2017 Thyroid hormone controls breast cancer cell movement via integrin alphaV/beta3/SRC/FAK/PI3-kinases. Hormones and Cancer [in press]. (doi:10.1007/s12672-016-0280-3)

Folkman J 1995 Angiogenesis in cancer, vascular, rheumatoid and other disease. Nature Medicine 1 27–31. (doi:10.1038/nm0195-27)

Francavilla A, Carr BI, Azzarone A, Polimeno L, Wang Z, Van Thiel DH, Subbotin V, Prelich JG & Starzl TE 1994 Hepatocyte proliferation and gene expression induced by triiodothyronine in vivo and in vitro. Hepatology 20 1237–1241. (doi:10.1016/0270-9139(94)90763-3)

Franke TF 2008 PI3K/Akt: getting it right matters. Oncogene 27 6473–6488. (doi:10.1038/onc.2008.313)

Furuya F, Hanover JA & Cheng SY 2006 Activation of phosphatidylinositol 3-kinase signaling by a mutant thyroid hormone beta receptor. PNAS 103 1780–1785. (doi:10.1073/pnas.0510849103)

Garcia-Silva S & Aranda A 2004 The thyroid hormone receptor is a suppressor of ras-mediated transcription, proliferation, and transformation. Molecular and Cellular Biology 24 7514–7523. (doi:10.1128/MCB.24.17.7514-7523.2004)

Gereben B, Zeold A, Dentice M, Salvatore D & Bianco AC 2008 Activation and inactivation of thyroid hormone by deiodinases: local action with general consequences. Cellular and Molecular Life Sciences 65 570–590. (doi:10.1007/s00018-007-7396-0)

Glinskii AB, Glinsky GV, Lin HY, Tang HY, Sun M, Davis FB, Luidens MK, Mousa SA, Hercbergs AH & Davis PJ 2009 Modification of survival pathway gene expression in human breast cancer cells by tetraiodothyroacetic acid (tetrac). Cell Cycle 8 3562–3570. (doi:10.4161/cc.8.21.9963)

Gogvadze V, Orrenius S & Zhivotovsky B 2008 Mitochondria in cancer cells: what is so special about them? Trends in Cell Biology 18 165–173. (doi:10.1016/j.tcb.2008.01.006)

Gonzalez-Sancho JM, Garcia V, Bonilla F & Munoz A 2003 Thyroid hormone receptors/THR genes in human cancer. Cancer Letters 192 121–132. (doi:10.1016/S0304-3835(02)00614-6)

Gottardi CJ & Gumbiner BM 2001 Adhesion signaling: how beta-catenin interacts with its partners. Current Biology 11 R792–794. (doi:10.1016/S0960-9822(01)00473-0)

Goubran HA, Kotb RR, Stakiw J, Emara ME & Burnouf T 2014 Regulation of tumor growth and metastasis: the role of tumor

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 15: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R381Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

microenvironment. Cancer Growth and Metastasis 7 9–18. (doi:10.4137/CGM.S11285)

Grivennikov SI, Greten FR & Karin M 2010 Immunity, inflammation, and cancer. Cell 140 883–899. (doi:10.1016/j.cell.2010.01.025)

Guigon CJ, Zhao L, Lu C, Willingham MC & Cheng SY 2008 Regulation of beta-catenin by a novel nongenomic action of thyroid hormone beta receptor. Molecular and Cellular Biology 28 4598–4608. (doi:10.1128/MCB.02192-07)

Hanahan D & Weinberg RA 2000 The hallmarks of cancer. Cell 100 57–70. (doi:10.1016/S0092-8674(00)81683-9)

Hanahan D & Weinberg RA 2011 Hallmarks of cancer: the next generation. Cell 144 646–674. (doi:10.1016/j.cell.2011.02.013)

Hassan MM, Kaseb A, Li D, Patt YZ, Vauthey JN, Thomas MB, Curley SA, Spitz MR, Sherman SI, Abdalla EK, et al. 2009 Association between hypothyroidism and hepatocellular carcinoma: a case-control study in the United States. Hepatology 49 1563–1570. (doi:10.1002/hep.22793)

Hellevik AI, Asvold BO, Bjoro T, Romundstad PR, Nilsen TI & Vatten LJ 2009 Thyroid function and cancer risk: a prospective population study. Cancer Epidemiology, Biomarkers and Prevention 18 570–574. (doi:10.1158/1055-9965.EPI-08-0911)

Hercbergs A, Johnson RE, Ashur-Fabian O, Garfield DH & Davis PJ 2015 Medically induced euthyroid hypothyroxinemia may extend survival in compassionate need cancer patients: an observational study. Oncologist 20 72–76. (doi:10.1634/theoncologist.2014-0308)

Hercbergs AA, Goyal LK, Suh JH, Lee S, Reddy CA, Cohen BH, Stevens GH, Reddy SK, Peereboom DM, Elson PJ, et al. 2003 Propylthiouracil-induced chemical hypothyroidism with high-dose tamoxifen prolongs survival in recurrent high grade glioma: a phase I/II study. Anticancer Research 23 617–626.

Hiroi Y, Kim HH, Ying H, Furuya F, Huang Z, Simoncini T, Noma K, Ueki K, Nguyen NH, Scanlan TS, et al. 2006 Rapid nongenomic actions of thyroid hormone. PNAS 103 14104–14109. (doi:10.1073/pnas.0601600103)

Hsu PP & Sabatini DM 2008 Cancer cell metabolism: Warburg and beyond. Cell 134 703–707. (doi:10.1016/j.cell.2008.08.021)

Huang SA, Tu HM, Harney JW, Venihaki M, Butte AJ, Kozakewich HP, Fishman SJ & Larsen PR 2000 Severe hypothyroidism caused by type 3 iodothyronine deiodinase in infantile hemangiomas. New England Journal of Medicine 343 185–189. (doi:10.1056/NEJM200007203430305)

Huang SA, Fish SA, Dorfman DM, Salvatore D, Kozakewich HP, Mandel SJ & Larsen PR 2002 A 21-year-old woman with consumptive hypothyroidism due to a vascular tumor expressing type 3 iodothyronine deiodinase. Journal of Clinical Endocrinology and Metabolism 87 4457–4461. (doi:10.1210/jc.2002-020627)

Huang YH, Lin YH, Chi HC, Liao CH, Liao CJ, Wu SM, Chen CY, Tseng YH, Tsai CY, Lin SY, et al. 2013 Thyroid hormone regulation of miR-21 enhances migration and invasion of hepatoma. Cancer Research 73 2505–2517. (doi:10.1158/0008-5472.CAN-12-2218)

Huang L, Yu Z, Zhang T, Zhao X & Huang G 2014 HSP40 interacts with pyruvate kinase M2 and regulates glycolysis and cell proliferation in tumor cells. PLoS ONE 9 e92949. (doi:10.1371/journal.pone.0092949)

Hulbert AJ 2000 Thyroid hormones and their effects: a new perspective. Biological Reviews of the Cambridge Philosophical Society 75 519–631. (doi:10.1017/S146479310000556X)

Ihara A, Koizumi H, Hashizume R & Uchikoshi T 1996 Expression of epithelial cadherin and alpha- and beta-catenins in nontumoral livers and hepatocellular carcinomas. Hepatology 23 1441–1447. (doi:10.1053/jhep.1996.v23.pm0008675162)

Jackson SP & Bartek J 2009 The DNA-damage response in human biology and disease. Nature 461 1071–1078. (doi:10.1038/nature08467)

Jo S, Kallo I, Bardoczi Z, Arrojo e Drigo R, Zeold A, Liposits Z, Oliva A, Lemmon VP, Bixby JL, Gereben B, et al. 2012 Neuronal hypoxia induces Hsp40-mediated nuclear import of type 3 deiodinase as an

adaptive mechanism to reduce cellular metabolism. Journal of Neuroscience 32 8491–8500. (doi:10.1523/jneurosci.6514-11.2012)

Johnstone RW, Frew AJ & Smyth MJ 2008 The TRAIL apoptotic pathway in cancer onset, progression and therapy. Nature Reviews Cancer 8 782–798. (doi:10.1038/nrc2465)

Joyce JA & Pollard JW 2009 Microenvironmental regulation of metastasis. Nature Reviews Cancer 9 239–252. (doi:10.1038/nrc2618)

Kastan MB 2008 DNA damage responses: mechanisms and roles in human disease: 2007 G.H.A. Clowes Memorial Award Lecture. Molecular Cancer Research 6 517–524. (doi:10.1158/1541-7786.MCR-08-0020)

Keith B & Simon MC 2007 Hypoxia-inducible factors, stem cells, and cancer. Cell 129 465–472. (doi:10.1016/j.cell.2007.04.019)

Kenessey A & Ojamaa K 2006 Thyroid hormone stimulates protein synthesis in the cardiomyocyte by activating the Akt-mTOR and p70S6K pathways. Journal of Biological Chemistry 281 20666–20672. (doi:10.1074/jbc.M512671200)

Kim KJ, Li B, Winer J, Armanini M, Gillett N, Phillips HS & Ferrara N 1993 Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature 362 841–844. (doi:10.1038/362841a0)

Kim BW, Daniels GH, Harrison BJ, Price A, Harney JW, Larsen PR & Weetman AP 2003 Overexpression of type 2 iodothyronine deiodinase in follicular carcinoma as a cause of low circulating free thyroxine levels. Journal of Clinical Endocrinology and Metabolism 88 594–598. (doi:10.1210/jc.2002-020921)

Kim DW, Zhao L, Hanover J, Willingham M & Cheng SY 2012 Thyroid hormone receptor beta suppresses SV40-mediated tumorigenesis via novel nongenomic actions. American Journal of Cancer Research 2 606–619. (doi:10.1158/1538-7445.AM2012-606)

Klaus A & Birchmeier W 2008 Wnt signalling and its impact on development and cancer. Nature Reviews Cancer 8 387–398. (doi:10.1038/nrc2389)

Koppenol WH, Bounds PL & Dang CV 2011 Otto Warburg’s contributions to current concepts of cancer metabolism. Nature Reviews Cancer 11 325–337. (doi:10.1038/nrc3038)

Kornasiewicz O, Debski M, Grat M, Lenartowicz B, Stepnowska M, Szalas A, Bar-Andziak E & Krawczyk M 2014 Enzymatic activity of type 1 iodothyronine deiodinase in selected liver tumors. Archives of Medical Science 10 801–805. (doi:10.5114/aoms.2013.34323)

Kozawa O, Hatakeyama D, Yoshida M, Kamiya Y, Kondo C, Matsuno H & Uematsu T 2001 Activation of p44/p42 mitogen-activated protein kinase limits triiodothyronine-stimulated alkaline phosphatase activity in osteoblasts. Biochemical and Biophysical Research Communications 286 1140–1143. (doi:10.1006/bbrc.2001.5515)

Kress E, Rezza A, Nadjar J, Samarut J & Plateroti M 2009a The frizzled-related sFRP2 gene is a target of thyroid hormone receptor alpha1 and activates beta-catenin signaling in mouse intestine. Journal of Biological Chemistry 284 1234–1241. (doi:10.1074/jbc.M806548200)

Kress E, Samarut J & Plateroti M 2009b Thyroid hormones and the control of cell proliferation or cell differentiation: paradox or duality? Molecular and Cellular Endocrinology 313 36–49. (doi:10.1016/j.mce.2009.08.028)

Kress E, Skah S, Sirakov M, Nadjar J, Gadot N, Scoazec JY, Samarut J & Plateroti M 2010 Cooperation between the thyroid hormone receptor TRalpha1 and the WNT pathway in the induction of intestinal tumorigenesis. Gastroenterology 138 1863–1874. (doi:10.1053/j.gastro.2010.01.041)

Kroemer G & Pouyssegur J 2008 Tumor cell metabolism: cancer’s Achilles’ heel. Cancer Cell 13 472–482. (doi:10.1016/j.ccr.2008.05.005)

Lade AG & Monga SP 2011 Beta-catenin signaling in hepatic development and progenitors: which way does the WNT blow? Developmental Dynamics 240 486–500. (doi:10.1002/dvdy.22522)

Laoag-Fernandez JB, Matsuo H, Murakoshi H, Hamada AL, Tsang BK & Maruo T 2004 3,5,3′-Triiodothyronine down-regulates Fas and Fas

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 16: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R382Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

ligand expression and suppresses caspase-3 and poly (adenosine 5′-diphosphate-ribose) polymerase cleavage and apoptosis in early placental extravillous trophoblasts in vitro. Journal of Clinical Endocrinology and Metabolism 89 4069–4077. (doi:10.1210/jc.2003-032208)

Lebel JM, Dussault JH & Puymirat J 1994 Overexpression of the beta 1 thyroid receptor induces differentiation in neuro-2a cells. PNAS 91 2644–2648. (doi:10.1073/pnas.91.7.2644)

Ledda-Columbano GM, Perra A, Loi R, Shinozuka H & Columbano A 2000 Cell proliferation induced by triiodothyronine in rat liver is associated with nodule regression and reduction of hepatocellular carcinomas. Cancer Research 60 603–609.

Lee AC, Fenster BE, Ito H, Takeda K, Bae NS, Hirai T, Yu ZX, Ferrans VJ, Howard BH & Finkel T 1999 Ras proteins induce senescence by altering the intracellular levels of reactive oxygen species. Journal of Biological Chemistry 274 7936–7940. (doi:10.1074/jbc.274.12.7936)

Liao CH, Yeh SC, Huang YH, Chen RN, Tsai MM, Chen WJ, Chi HC, Tai PJ, Liao CJ, Wu SM, et al. 2010 Positive regulation of spondin 2 by thyroid hormone is associated with cell migration and invasion. Endocrine-Related Cancer 17 99–111. (doi:10.1677/ERC-09-0050)

Liao CH, Yeh CT, Huang YH, Wu SM, Chi HC, Tsai MM, Tsai CY, Liao CJ, Tseng YH, Lin YH, et al. 2012 Dickkopf 4 positively regulated by the thyroid hormone receptor suppresses cell invasion in human hepatoma cells. Hepatology 55 910–920. (doi:10.1002/hep.24740)

Liappas A, Mourouzis I, Zisakis A, Economou K, Lea RW & Pantos C 2011 Cell-type-dependent thyroid hormone effects on glioma tumor cell lines. Journal of Thyroid Research 2011 856050. (doi:10.4061/2011/856050)

Lin HY, Thacorf HR, Davis FB & Davis PJ 1996 Potentiation by thyroxine of interferon-gamma-induced antiviral state requires PKA and PKC activities. American Journal of Physiology 271 C1256–C1261.

Lin HY, Davis FB, Gordinier JK, Martino LJ & Davis PJ 1999a Thyroid hormone induces activation of mitogen-activated protein kinase in cultured cells. American Journal of Physiology 276 C1014–C1024.

Lin HY, Shih A, Davis FB & Davis PJ 1999b Thyroid hormone promotes the phosphorylation of STAT3 and potentiates the action of epidermal growth factor in cultured cells. Biochemical Journal 338 427–432. (doi:10.1042/bj3380427)

Lin HY, Zhang S, West BL, Tang HY, Passaretti T, Davis FB & Davis PJ 2003 Identification of the putative MAP kinase docking site in the thyroid hormone receptor-beta1 DNA-binding domain: functional consequences of mutations at the docking site. Biochemistry 42 7571–7579. (doi:10.1021/bi0273967)

Lin HY, Tang HY, Shih A, Keating T, Cao G, Davis PJ & Davis FB 2007 Thyroid hormone is a MAPK-dependent growth factor for thyroid cancer cells and is anti-apoptotic. Steroids 72 180–187. (doi:10.1016/j.steroids.2006.11.014)

Lin HY, Tang HY, Keating T, Wu YH, Shih A, Hammond D, Sun M, Hercbergs A, Davis FB & Davis PJ 2008 Resveratrol is pro-apoptotic and thyroid hormone is anti-apoptotic in glioma cells: both actions are integrin and ERK mediated. Carcinogenesis 29 62–69. (doi:10.1093/carcin/bgm239)

Lin HY, Sun M, Tang HY, Lin C, Luidens MK, Mousa SA, Incerpi S, Drusano GL, Davis FB & Davis PJ 2009 L-Thyroxine vs. 3,5,3′-triiodo-L-thyronine and cell proliferation: activation of mitogen-activated protein kinase and phosphatidylinositol 3-kinase. American Journal of Physiology: Cell Physiology 296 C980–C991. (doi:10.1152/ajpcell.00305.2008)

Lin YH, Huang YH, Wu MH, Wu SM, Chi HC, Liao CJ, Chen CY, Tseng YH, Tsai CY, Tsai MM, et al. 2013a Thyroid hormone suppresses cell proliferation through endoglin-mediated promotion of p21 stability. Oncogene 32 3904–3914. (doi:10.1038/onc.2013.5)

Lin YH, Liao CJ, Huang YH, Wu MH, Chi HC, Wu SM, Chen CY, Tseng YH, Tsai CY, Chung IH, et al. 2013b Thyroid hormone receptor represses miR-17 expression to enhance tumor metastasis in human hepatoma cells. Oncogene 32 4509–4518. (doi:10.1038/onc.2013.309)

Lin YH, Wu MH, Liao CJ, Huang YH, Chi HC, Wu SM, Chen CY, Tseng YH, Tsai CY, Chung IH, et al. 2015 Repression of microRNA-130b by thyroid hormone enhances cell motility. Journal of Hepatology 62 1328–1340. (doi:10.1016/j.jhep.2014.12.035)

Lowe SW, Cepero E & Evan G 2004 Intrinsic tumour suppression. Nature 432 307–315. (doi:10.1038/nature03098)

Lowy DR & Willumsen BM 1993 Function and regulation of ras. Annual Review of Biochemistry 62 851–891. (doi:10.1146/annurev.bi.62.070193.004223)

Luongo C, Trivisano L, Alfano F & Salvatore D 2013 Type 3 deiodinase and consumptive hypothyroidism: a common mechanism for a rare disease. Frontiers in Endocrinology 4 115. (doi:10.3389/fendo.2013.00115)

Maia AL, Goemann IM, Meyer EL & Wajner SM 2011 Deiodinases: the balance of thyroid hormone: type 1 iodothyronine deiodinase in human physiology and disease. Journal of Endocrinology 209 283–297. (doi:10.1530/JOE-10-0481)

Mancini A, Di Segni C, Raimondo S, Olivieri G, Silvestrini A, Meucci E & Curro D 2016 Thyroid hormones, oxidative stress, and inflammation. Mediators of Inflammation 2016 6757154. (doi:10.1155/2016/6757154)

Martinez-Iglesias O, Garcia-Silva S, Tenbaum SP, Regadera J, Larcher F, Paramio JM, Vennstrom B & Aranda A 2009 Thyroid hormone receptor beta1 acts as a potent suppressor of tumor invasiveness and metastasis. Cancer Research 69 501–509. (doi:10.1158/0008-5472.CAN-08-2198)

Martinez-Iglesias O, Alonso-Merino E & Aranda A 2016a Tumor suppressive actions of the nuclear receptor corepressor 1. Pharmacological Research 108 75–79. (doi:10.1016/j.phrs.2016.04.027)

Martinez-Iglesias OA, Alonso-Merino E, Gomez-Rey S, Velasco-Martin JP, Martin Orozco R, Luengo E, Garcia Martin R, Ibanez de Caceres I, Fernandez AF, Fraga MF, et al. 2016b Autoregulatory loop of nuclear corepressor 1 expression controls invasion, tumor growth, and metastasis. PNAS 113 E328–E337. (doi:10.1073/pnas.1520469113)

Mascanfroni ID, Montesinos Mdel M, Alamino VA, Susperreguy S, Nicola JP, Ilarregui JM, Masini-Repiso AM, Rabinovich GA & Pellizas CG 2010 Nuclear factor (NF)-kappaB-dependent thyroid hormone receptor beta1 expression controls dendritic cell function via Akt signaling. Journal of Biological Chemistry 285 9569–9582. (doi:10.1074/jbc.M109.071241)

McMahon AP, Ingham PW & Tabin CJ 2003 Developmental roles and clinical significance of hedgehog signaling. Current Topics in Developmental Biology 53 1–114. (doi:10.1016/S0070-2153(03)53002-2)

Meng R, Tang HY, Westfall J, London D, Cao JH, Mousa SA, Luidens M, Hercbergs A, Davis FB, Davis PJ, et al. 2011 Crosstalk between integrin alphavbeta3 and estrogen receptor-alpha is involved in thyroid hormone-induced proliferation in human lung carcinoma cells. PLoS ONE 6 e27547. (doi:10.1371/journal.pone.0027547)

Meyer EL, Goemann IM, Dora JM, Wagner MS & Maia AL 2008 Type 2 iodothyronine deiodinase is highly expressed in medullary thyroid carcinoma. Molecular and Cellular Endocrinology 289 16–22. (doi:10.1016/j.mce.2008.04.009)

Mihara S, Suzuki N, Wakisaka S, Suzuki S, Sekita N, Yamamoto S, Saito N, Hoshino T & Sakane T 1999 Effects of thyroid hormones on apoptotic cell death of human lymphocytes. Journal of Clinical Endocrinology and Metabolism 84 1378–1385. (doi:10.1210/jc.84.4.1378)

Miller LD, Park KS, Guo QM, Alkharouf NW, Malek RL, Lee NH, Liu ET & Cheng SY 2001 Silencing of Wnt signaling and activation of multiple metabolic pathways in response to thyroid hormone-stimulated cell proliferation. Molecular and Cellular Biology 21 6626–6639. (doi:10.1128/MCB.21.19.6626-6639.2001)

Miro C, Ambrosio R, De Stefano MA, Di Girolamo D, Di Cicco E, Cicatiello AG, Mancino G, Porcelli T, Raia M, Del Vecchio L, et al. 2017 The concerted action of type 2 and type 3 deiodinases regulates the cell cycle and survival of basal cell carcinoma cells. Thyroid 27 567–576. (doi:10.1089/thy.2016.0532)

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 17: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R383Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

Mondello C & Scovassi AI 2004 Telomeres, telomerase, and apoptosis. Biochemistry and Cell Biology 82 498–507. (doi:10.1139/o04-048)

Mori K, Yoshida K, Kayama T, Kaise N, Fukazawa H, Kiso Y, Kikuchi K, Aizawa Y & Abe K 1993 Thyroxine 5-deiodinase in human brain tumors. Journal of Clinical Endocrinology and Metabolism 77 1198–1202. (doi:10.1210/jc.77.5.1198)

Mousa SA, O’Connor LJ, Bergh JJ, Davis FB, Scanlan TS & Davis PJ 2005 The proangiogenic action of thyroid hormone analogue GC-1 is initiated at an integrin. Journal of Cardiovascular Pharmacology 46 356–360. (doi:10.1097/01.fjc.0000175438.94906.a0)

Mousa SA, Yalcin M, Bharali DJ, Meng R, Tang HY, Lin HY, Davis FB & Davis PJ 2012 Tetraiodothyroacetic acid and its nanoformulation inhibit thyroid hormone stimulation of non-small cell lung cancer cells in vitro and its growth in xenografts. Lung Cancer 76 39–45. (doi:10.1016/j.lungcan.2011.10.003)

Mousa SA, Lin HY, Tang HY, Hercbergs A, Luidens MK & Davis PJ 2014 Modulation of angiogenesis by thyroid hormone and hormone analogues: implications for cancer management. Angiogenesis 17 463–469. (doi:10.1007/s10456-014-9418-5)

Mukherjee S, Samanta L, Roy A, Bhanja S & Chainy GB 2014 Supplementation of T3 recovers hypothyroid rat liver cells from oxidatively damaged inner mitochondrial membrane leading to apoptosis. BioMed Research International 2014 590897. (doi:10.1155/2014/590897)

Murakami M, Araki O, Morimura T, Hosoi Y, Mizuma H, Yamada M, Kurihara H, Ishiuchi S, Tamura M, Sasaki T, et al. 2000 Expression of type II iodothyronine deiodinase in brain tumors. Journal of Clinical Endocrinology and Metabolism 85 4403–4406. (doi:10.1210/jc.85.11.4403)

Natsume H, Sasaki S, Kitagawa M, Kashiwabara Y, Matsushita A, Nakano K, Nishiyama K, Nagayama K, Misawa H, Masuda H, et al. 2003 Beta-catenin/Tcf-1-mediated transactivation of cyclin D1 promoter is negatively regulated by thyroid hormone. Biochemical and Biophysical Research Communications 309 408–413. (doi:10.1016/j.bbrc.2003.08.019)

Nauman P, Bonicki W, Michalik R, Warzecha A & Czernicki Z 2004 The concentration of thyroid hormones and activities of iodothyronine deiodinases are altered in human brain gliomas. Folia Neuropathologica 42 67–73.

Negrini S, Gorgoulis VG & Halazonetis TD 2010 Genomic instability – an evolving hallmark of cancer. Nature Reviews Molecular Cell Biology 11 220–228. (doi:10.1038/nrm2858)

Nygard M, Wahlstrom GM, Gustafsson MV, Tokumoto YM & Bondesson M 2003 Hormone-dependent repression of the E2F-1 gene by thyroid hormone receptors. Molecular Endocrinology 17 79–92. (doi:10.1210/me.2002-0107)

Oro AE, Higgins KM, Hu Z, Bonifas JM, Epstein EH Jr & Scott MP 1997 Basal cell carcinomas in mice overexpressing sonic hedgehog. Science 276 817–821. (doi:10.1126/science.276.5313.817)

Otto T & Fandrey J 2008 Thyroid hormone induces hypoxia-inducible factor 1alpha gene expression through thyroid hormone receptor beta/retinoid x receptor alpha-dependent activation of hepatic leukemia factor. Endocrinology 149 2241–2250. (doi:10.1210/en.2007-1238)

Pachucki J, Ambroziak M, Tanski Z, Luczak J, Nauman J & Nauman A 2001 Type I 5′-iodothyronine deiodinase activity and mRNA are remarkably reduced in renal clear cell carcinoma. Journal of Endocrinological Investigation 24 253–261. (doi:10.1007/BF03343855)

Pappas L, Xu XL, Abramson DH & Jhanwar SC 2017 Genomic instability and proliferation/survival pathways in RB1-deficient malignancies. Advances in Biological Regulation 64 20–32. (doi:10.1016/j.jbior.2017.01.002)

Park D, Lapteva N, Seethammagari M, Slawin KM & Spencer DM 2006 An essential role for Akt1 in dendritic cell function and tumor immunotherapy. Nature Biotechnology 24 1581–1590. (doi:10.1038/nbt1262)

Pasca di Magliano M & Hebrok M 2003 Hedgehog signalling in cancer formation and maintenance. Nature Reviews Cancer 3 903–911. (doi:10.1038/nrc1229)

Pascual A & Aranda A 2013 Thyroid hormone receptors, cell growth and differentiation. Biochimica et Biophysica Acta 1830 3908–3916. (doi:10.1016/j.bbagen.2012.03.012)

Perra A, Kowalik MA, Pibiri M, Ledda-Columbano GM & Columbano A 2009 Thyroid hormone receptor ligands induce regression of rat preneoplastic liver lesions causing their reversion to a differentiated phenotype. Hepatology 49 1287–1296. (doi:10.1002/hep.22750)

Perri A, Catalano S, Bonofiglio D, Vizza D, Rovito D, Qi H, Aquila S, Panza S, Rizza P, Lanzino M, et al. 2014 T3 enhances thyroid cancer cell proliferation through TRbeta1/Oct-1-mediated cyclin D1 activation. Molecular and Cellular Endocrinology 382 205–217. (doi:10.1016/j.mce.2013.10.001)

Perrotta C, De Palma C, Clementi E & Cervia D 2015 Hormones and immunity in cancer: are thyroid hormones endocrine players in the microglia/glioma cross-talk? Frontiers in Cellular Neuroscience 9 236. (doi:10.3389/fncel.2015.00236)

Pietrzak M & Puzianowska-Kuznicka M 2008 Triiodothyronine utilizes phosphatidylinositol 3-kinase pathway to activate anti-apoptotic myeloid cell leukemia-1. Journal of Molecular Endocrinology 41 177–186. (doi:10.1677/JME-08-0010)

Pinto M, Soares P & Ribatti D 2011 Thyroid hormone as a regulator of tumor induced angiogenesis. Cancer Letters 301 119–126. (doi:10.1016/j.canlet.2010.11.011)

Plateroti M, Kress E, Mori JI & Samarut J 2006 Thyroid hormone receptor alpha1 directly controls transcription of the beta-catenin gene in intestinal epithelial cells. Molecular and Cellular Biology 26 3204–3214. (doi:10.1128/MCB.26.8.3204-3214.2006)

Polakis P 1999 The oncogenic activation of beta-catenin. Current Opinion in Genetics and Development 9 15–21. (doi:10.1016/S0959-437X(99)80003-3)

Polyzos SA, Kita M, Efstathiadou Z, Poulakos P, Slavakis A, Sofianou D, Flaris N, Leontsini M, Kourtis A & Avramidis A 2008 Serum thyrotropin concentration as a biochemical predictor of thyroid malignancy in patients presenting with thyroid nodules. Journal of Cancer Research and Clinical Oncology 134 953–960. (doi:10.1007/s00432-008-0373-7)

Rajalingam K, Schreck R, Rapp UR & Albert S 2007 Ras oncogenes and their downstream targets. Biochimica et Biophysica Acta 1773 1177–1195. (doi:10.1016/j.bbamcr.2007.01.012)

Rebbaa A, Chu F, Davis FB, Davis PJ & Mousa SA 2008 Novel function of the thyroid hormone analog tetraiodothyroacetic acid: a cancer chemosensitizing and anti-cancer agent. Angiogenesis 11 269–276. (doi:10.1007/s10456-008-9110-8)

Reddy A, Dash C, Leerapun A, Mettler TA, Stadheim LM, Lazaridis KN, Roberts RO & Roberts LR 2007 Hypothyroidism: a possible risk factor for liver cancer in patients with no known underlying cause of liver disease. Clinical Gastroenterology and Hepatology 5 118–123. (doi:10.1016/j.cgh.2006.07.011)

Risau W 1997 Mechanisms of angiogenesis. Nature 386 671–674. (doi:10.1038/386671a0)

Romitti M, Wajner SM, Zennig N, Goemann IM, Bueno AL, Meyer EL & Maia AL 2012 Increased type 3 deiodinase expression in papillary thyroid carcinoma. Thyroid 22 897–904. (doi:10.1089/thy.2012.0031)

Romitti M, Ceolin L, Siqueira DR, Ferreira CV, Wajner SM & Maia AL 2013 Signaling pathways in follicular cell-derived thyroid carcinomas (review). International Journal of Oncology 42 19–28. (doi:10.3892/ijo.2012.1681)

Romitti M, Wajner SM, Ceolin L, Ferreira CV, Ribeiro RV, Rohenkohl HC, Weber Sde S, Lopez PL, Fuziwara CS, Kimura ET, et al. 2016 MAPK and SHH pathways modulate type 3 deiodinase expression in papillary thyroid carcinoma. Endocrine-Related Cancer 23 135–146. (doi:10.1530/ERC-15-0162)

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 18: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R384Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

Ruiz i Altaba A, Sanchez P & Dahmane N 2002 Gli and hedgehog in cancer: tumours, embryos and stem cells. Nature Reviews Cancer 2 361–372. (doi:10.1038/nrc796)

Sabatino L, Iervasi G, Ferrazzi P, Francesconi D & Chopra IJ 2000 A study of iodothyronine 5′-monodeiodinase activities in normal and pathological tissues in man and their comparison with activities in rat tissues. Life Science 68 191–202. (doi:10.1016/S0024-3205(00)00929-2)

Sar P, Peter R, Rath B, Das Mohapatra A & Mishra SK 2011 3,3′5 Triiodo L thyronine induces apoptosis in human breast cancer MCF-7 cells, repressing SMP30 expression through negative thyroid response elements. PLoS ONE 6 e20861. (doi:10.1371/journal.pone.0020861)

Schmohl KA, Muller AM, Wechselberger A, Ruhland S, Salb N, Schwenk N, Heuer H, Carlsen J, Goke B, Nelson PJ, et al. 2015 Thyroid hormones and tetrac: new regulators of tumour stroma formation via integrin alphavbeta3. Endocrine-Related Cancer 22 941–952. (doi:10.1530/ERC-15-0245)

Sekulic A, Migden MR, Oro AE, Dirix L, Lewis KD, Hainsworth JD, Solomon JA, Yoo S, Arron ST, Friedlander PA, et al. 2012 Efficacy and safety of vismodegib in advanced basal-cell carcinoma. New England Journal of Medicine 366 2171–2179. (doi:10.1056/NEJMoa1113713)

Shields JM, Pruitt K, McFall A, Shaub A & Der CJ 2000 Understanding Ras: ‘it ain’t over ‘til it’s over’. Trends in Cell Biology 10 147–154. (doi:10.1016/S0962-8924(00)01740-2)

Shih A, Lin HY, Davis FB & Davis PJ 2001 Thyroid hormone promotes serine phosphorylation of p53 by mitogen-activated protein kinase. Biochemistry 40 2870–2878. (doi:10.1021/bi001978b)

Shinderman-Maman E, Cohen K, Weingarten C, Nabriski D, Twito O, Baraf L, Hercbergs A, Davis PJ, Werner H, Ellis M, et al. 2016 The thyroid hormone-alphavbeta3 integrin axis in ovarian cancer: regulation of gene transcription and MAPK-dependent proliferation. Oncogene 35 1977–1987. (doi:10.1038/onc.2015.262)

Silvestri E, Coppola M, Cioffi F & Goglia F 2014 Proteomic approaches for the study of tissue specific effects of 3,5,3′-triiodo-L-thyronine and 3,5-diiodo-L-thyronine in conditions of altered energy metabolism. Frontiers in Physiology 5 491. (doi:10.3389/fphys.2014.00491)

Simonides WS, Mulcahey MA, Redout EM, Muller A, Zuidwijk MJ, Visser TJ, Wassen FW, Crescenzi A, da-Silva WS, Harney J, et al. 2008 Hypoxia-inducible factor induces local thyroid hormone inactivation during hypoxic-ischemic disease in rats. Journal of Clinical Investigation 118 975–983. (doi:10.1172/JCI32824)

Simsek T, Duruksu G, Okcu A, Aksoy A, Erman G, Utkan Z, Canturk Z & Karaoz E 2014 Effect of hypothyroidism in the thyroidectomized rats on immunophenotypic characteristics and differentiation capacity of adipose tissue derived stem cells. European Review for Medical and Pharmacological Sciences 18 617–629.

Sirakov M, Skah S, Lone IN, Nadjar J, Angelov D & Plateroti M 2012 Multi-level interactions between the nuclear receptor TRalpha1 and the WNT effectors beta-catenin/Tcf4 in the intestinal epithelium. PLoS ONE 7 e34162. (doi:10.1371/journal.pone.0034162)

St Germain DL, Galton VA & Hernandez A 2009 Minireview: defining the roles of the iodothyronine deiodinases: current concepts and challenges. Endocrinology 150 1097–1107. (doi:10.1210/ en.2008-1588)

Sterle HA, Valli E, Cayrol F, Paulazo MA, Martinel Lamas DJ, Diaz Flaque MC, Klecha AJ, Colombo L, Medina VA, Cremaschi GA, et al. 2014 Thyroid status modulates T lymphoma growth via cell cycle regulatory proteins and angiogenesis. Journal of Endocrinology 222 243–255. (doi:10.1530/JOE-14-0159)

Sterle HA, Barreiro Arcos ML, Valli E, Paulazo MA, Mendez Huergo SP, Blidner AG, Cayrol F, Diaz Flaque MC, Klecha AJ, Medina VA, et al. 2016 The thyroid status reprograms T cell lymphoma growth and modulates immune cell frequencies. Journal of Molecular Medicine 94 417–429. (doi:10.1007/s00109-015-1363-2)

Storey NM, Gentile S, Ullah H, Russo A, Muessel M, Erxleben C & Armstrong DL 2006 Rapid signaling at the plasma membrane by a nuclear receptor for thyroid hormone. PNAS 103 5197–5201. (doi:10.1073/pnas.0600089103)

Suhane S & Ramanujan VK 2011 Thyroid hormone differentially modulates Warburg phenotype in breast cancer cells. Biochemical and Biophysical Research Communications 414 73–78. (doi:10.1016/j.bbrc.2011.09.024)

Tang HY, Lin HY, Zhang S, Davis FB & Davis PJ 2004 Thyroid hormone causes mitogen-activated protein kinase-dependent phosphorylation of the nuclear estrogen receptor. Endocrinology 145 3265–3272. (doi:10.1210/en.2004-0308)

Tang JY, Mackay-Wiggan JM, Aszterbaum M, Yauch RL, Lindgren J, Chang K, Coppola C, Chanana AM, Marji J, Bickers DR, et al. 2012 Inhibiting the hedgehog pathway in patients with the basal-cell nevus syndrome. New England Journal of Medicine 366 2180–2188. (doi:10.1056/NEJMoa1113538)

Tannahill LA, Visser TJ, McCabe CJ, Kachilele S, Boelaert K, Sheppard MC, Franklyn JA & Gittoes NJ 2002 Dysregulation of iodothyronine deiodinase enzyme expression and function in human pituitary tumours. Clinical Endocrinology 56 735–743. (doi:10.1046/j.1365-2265.2002.01541.x)

Tosovic A, Bondeson AG, Bondeson L, Ericsson UB, Malm J & Manjer J 2010 Prospectively measured triiodothyronine levels are positively associated with breast cancer risk in postmenopausal women. Breast Cancer Research 12 R33. (doi:10.1186/bcr2587)

Tosovic A, Bondeson AG, Bondeson L, Ericsson UB & Manjer J 2014 T3 levels in relation to prognostic factors in breast cancer: a population-based prospective cohort study. BMC Cancer 14 536. (doi:10.1186/1471-2407-14-536)

Verga Falzacappa C, Panacchia L, Bucci B, Stigliano A, Cavallo MG, Brunetti E, Toscano V & Misiti S 2006 3,5,3′-Triiodothyronine (T3) is a survival factor for pancreatic beta-cells undergoing apoptosis. Journal of Cellular Physiology 206 309–321. (doi:10.1002/jcp.20460)

Verga Falzacappa C, Patriarca V, Bucci B, Mangialardo C, Michienzi S, Moriggi G, Stigliano A, Brunetti E, Toscano V & Misiti S 2009 The TRbeta1 is essential in mediating T3 action on Akt pathway in human pancreatic insulinoma cells. Journal of Cellular Biochemistry 106 835–848. (doi:10.1002/jcb.22045)

Visser TJ, Does-Tobe I, Docter R & Hennemann G 1976 Subcellular localization of a rat liver enzyme converting thyroxine into tri-iodothyronine and possible involvement of essential thiol groups. Biochemical Journal 157 479–482. (doi:10.1042/bj1570479)

Wajner SM, Goemann IM, Bueno AL, Larsen PR & Maia AL 2011 IL-6 promotes nonthyroidal illness syndrome by blocking thyroxine activation while promoting thyroid hormone inactivation in human cells. Journal of Clinical Investigation 121 1834–1845. (doi:10.1172/jci44678)

Wajner SM, Rohenkohl HC, Serrano T & Maia AL 2015 Sodium selenite supplementation does not fully restore oxidative stress-induced deiodinase dysfunction: Implications for the nonthyroidal illness syndrome. Redox Biology 6 436–445. (doi:10.1016/j.redox.2015.09.002)

Wang T, Xia L, Ma S, Qi X, Li Q, Xia Y, Tang X, Cui D, Wang Z, Chi J, et al. 2016 Hepatocellular carcinoma: thyroid hormone promotes tumorigenicity through inducing cancer stem-like cell self-renewal. Scientific Reports 6 25183. (doi:10.1038/srep25183)

Warburg O 1956 On the origin of cancer cells. Science 123 309–314. (doi:10.1126/science.123.3191.309)

Wawrzynska L, Sakowicz A, Rudzinski P, Langfort R & Kurzyna M 2003 The conversion of thyroxine to triiodothyronine in the lung: comparison of activity of type I iodothyronine 5′ deiodinase in lung cancer with peripheral lung tissues. Monaldi Archives for Chest Disease 59 140–145.

White E 2012 Deconvoluting the context-dependent role for autophagy in cancer. Nature Reviews Cancer 12 401–410. (doi:10.1038/nrc3262)

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access

Page 19: Role of thyroid hormones in the neoplastic process: an ... · Review I M Goemann et al. Thyroid hormones and neoplasias 24:11 10.1530/ERC-17-0192 Role of thyroid hormones in the neoplastic

R385Review I M Goemann et al. Thyroid hormones and neoplasiasEn

do

crin

e-R

elat

ed C

ance

r

DOI: 10.1530/ERC-17-0192http://erc.endocrinology-journals.org © 2017 Society for Endocrinology

Printed in Great BritainPublished by Bioscientifica Ltd.

24:11

White E 2015 The role for autophagy in cancer. Journal of Clinical Investigation 125 42–46. (doi:10.1172/JCI73941)

Wu SM, Huang YH, Yeh CT, Tsai MM, Liao CH, Cheng WL, Chen WJ & Lin KH 2011 Cathepsin H regulated by the thyroid hormone receptors associate with tumor invasion in human hepatoma cells. Oncogene 30 2057–2069. (doi:10.1038/onc.2010.585)

Wu SM, Cheng WL, Lin CD & Lin KH 2013 Thyroid hormone actions in liver cancer. Cellular and Molecular Life Sciences 70 1915–1936. (doi:10.1007/s00018-012-1146-7)

Wu SM, Cheng WL, Liao CJ, Chi HC, Lin YH, Tseng YH, Tsai CY, Chen CY, Lin SL, Chen WJ, et al. 2015 Negative modulation of the epigenetic regulator, UHRF1, by thyroid hormone receptors suppresses liver cancer cell growth. International Journal of Cancer 137 37–49. (doi:10.1002/ijc.29368)

Yalcin M, Bharali DJ, Lansing L, Dyskin E, Mousa SS, Hercbergs A, Davis FB, Davis PJ & Mousa SA 2009 Tetraidothyroacetic acid (tetrac) and tetrac nanoparticles inhibit growth of human renal cell carcinoma xenografts. Anticancer Research 29 3825–3831.

Yalcin M, Bharali DJ, Dyskin E, Dier E, Lansing L, Mousa SS, Davis FB, Davis PJ & Mousa SA 2010a Tetraiodothyroacetic acid and tetraiodothyroacetic acid nanoparticle effectively inhibit the growth of human follicular thyroid cell carcinoma. Thyroid 20 281–286. (doi:10.1089/thy.2009.0249)

Yalcin M, Dyskin E, Lansing L, Bharali DJ, Mousa SS, Bridoux A, Hercbergs AH, Lin HY, Davis FB, Glinsky GV, et al. 2010b Tetraiodothyroacetic acid (tetrac) and nanoparticulate tetrac arrest growth of medullary carcinoma of the thyroid. Journal of Clinical

Endocrinology and Metabolism 95 1972–1980. (doi:10.1210/jc.2009-1926)

Yalcin M, Lin HY, Sudha T, Bharali DJ, Meng R, Tang HY, Davis FB, Stain SC, Davis PJ & Mousa SA 2013 Response of human pancreatic cancer cell xenografts to tetraiodothyroacetic acid nanoparticles. Hormones and Cancer 4 176–185. (doi:10.1007/s12672-013-0137-y)

Yen PM 2001 Physiological and molecular basis of thyroid hormone action. Physiological Reviews 81 1097–1142.

Yen CC, Huang YH, Liao CY, Liao CJ, Cheng WL, Chen WJ & Lin KH 2006 Mediation of the inhibitory effect of thyroid hormone on proliferation of hepatoma cells by transforming growth factor-beta. Journal of Molecular Endocrinology 36 9–21. (doi:10.1677/jme.1.01911)

Zambrano A, Garcia-Carpizo V, Gallardo ME, Villamuera R, Gomez-Ferreria MA, Pascual A, Buisine N, Sachs LM, Garesse R & Aranda A 2014 The thyroid hormone receptor beta induces DNA damage and premature senescence. Journal of Cell Biology 204 129–146. (doi:10.1083/jcb.201305084)

Zhang W & Liu HT 2002 MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell Research 12 9–18. (doi:10.1038/sj.cr.7290105)

Zhang C, Xia G & Tsang BK 2011 Interactions of thyroid hormone and FSH in the regulation of rat granulosa cell apoptosis. Frontiers in Bioscience 3 1401–1413. (doi:10.2741/e342)

Zhu XG, Zhao L, Willingham MC & Cheng SY 2010 Thyroid hormone receptors are tumor suppressors in a mouse model of metastatic follicular thyroid carcinoma. Oncogene 29 1909–1919. (doi:10.1038/onc.2009.476)

Received in final form 19 July 2017Accepted 24 August 2017

Downloaded from Bioscientifica.com at 04/26/2021 05:16:11AMvia free access