Next Article in Journal
Male and Female Animals Respond Differently to High-Fat Diet and Regular Exercise Training in a Mouse Model of Hyperlipidemia
Previous Article in Journal
Combinations of Piperine with Hydroxypropyl-β-Cyclodextrin as a Multifunctional System
Previous Article in Special Issue
The Archaeal Small Heat Shock Protein Hsp17.6 Protects Proteins from Oxidative Inactivation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Molecular Chaperones and Thyroid Cancer

by
Letizia Paladino
1,2,
Alessandra Maria Vitale
1,2,
Radha Santonocito
1,
Alessandro Pitruzzella
1,2,3,
Calogero Cipolla
4,
Giuseppa Graceffa
4,
Fabio Bucchieri
1,
Everly Conway de Macario
5,
Alberto J. L. Macario
2,5 and
Francesca Rappa
1,*
1
Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy
2
Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy
3
Consorzio Universitario Caltanissetta, University of Palermo, 93100 Caltanissetta, Italy
4
Department of Surgical Oncology and Oral Sciences, University of Palermo, 90127 Palermo, Italy
5
Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(8), 4196; https://doi.org/10.3390/ijms22084196
Submission received: 29 March 2021 / Revised: 15 April 2021 / Accepted: 16 April 2021 / Published: 18 April 2021
(This article belongs to the Special Issue Molecular Chaperones 3.0)

Abstract

:
Thyroid cancers are the most common of the endocrine system malignancies and progress must be made in the areas of differential diagnosis and treatment to improve patient management. Advances in the understanding of carcinogenic mechanisms have occurred in various fronts, including studies of the chaperone system (CS). Components of the CS are found to be quantitatively increased or decreased, and some correlations have been established between the quantitative changes and tumor type, prognosis, and response to treatment. These correlations provide the basis for identifying distinctive patterns useful in differential diagnosis and for planning experiments aiming at elucidating the role of the CS in tumorigenesis. Here, we discuss studies of the CS components in various thyroid cancers (TC). The chaperones belonging to the families of the small heat-shock proteins Hsp70 and Hsp90 and the chaperonin of Group I, Hsp60, have been quantified mostly by immunohistochemistry and Western blot in tumor and normal control tissues and in extracellular vesicles. Distinctive differences were revealed between the various thyroid tumor types. The most frequent finding was an increase in the chaperones, which can be attributed to the augmented need for chaperones the tumor cells have because of their accelerated metabolism, growth, and division rate. Thus, chaperones help the tumor cell rather than protect the patient, exemplifying chaperonopathies by mistake or collaborationism. This highlights the need for research on chaperonotherapy, namely the development of means to eliminate/inhibit pathogenic chaperones.

1. Introduction

1.1. Thyroid Tumors

Thyroid is an important component of the endocrine system involved in the regulation of basal metabolism, heart rate, blood pressure, and temperature. Thyroid pathologies can cause serious diseases, for example, hyperthyroidism and hypothyroidism. The thyroid disease goiter is characterized by an increase in the gland volume, which can be diffuse or nodular. Although in most cases, the thyroid nodules are benign, they are malignant in a small fraction of patients, representing cases of thyroid cancer (TC).
TC is the most common endocrine cancer. Its incidence has been growing in recent years, especially in females, suggesting that female hormones may be involved in pathogenesis [1]. There are various types of TC, which are classified considering the cell of origin and histological characteristics into two main groups: a) well-differentiated, including papillary thyroid carcinoma (PTC), follicular thyroid carcinoma (FTC), and Hürthle cell carcinoma; and b) poorly differentiated (PDTCs), such as anaplastic/undifferentiated thyroid carcinoma (ATC) and medullary thyroid cancer (MTC). PTC, FTC, and ATC originate from follicular cells, while MTC stems from parafollicular C-cells and constitutes a minor fraction of TC [2,3].
Fine-needle aspiration (FNA) biopsy is the most common diagnostic test in the initial evaluation of patients with a thyroid nodule, yielding a diagnostic accuracy ranging from 70% to 97%. Despite the benefits of FNA cytology for diagnosing papillary, medullary, and anaplastic TC, it is not helpful in determining whether follicular or Hürthle cell thyroid growths are benign or malignant [4]. Therefore, approximately 10% of FNA results lead to misdiagnosis [5]. For this reason, specific genetic alterations, such as proto-oncogene serine/threonine protein kinase (RAF) mutations, have been identified as a reliable marker for improving diagnosis [6]. Other promising biomarkers are members of the chaperone system, whose role in thyroid carcinogenesis is still poorly understood.

1.2. The Chaperone System

The main components of the chaperone (also called chaperoning) system are the molecular chaperones, some of which are called heat shock proteins or Hsps [7,8]. They have canonical functions pertinent to maintenance of protein homeostasis that include interactions with the ubiquitin–proteasome system and chaperone-mediated autophagy mechanisms [9,10] and other functions (noncanonical) involving interaction with the immune system with implications in inflammatory and autoimmune disorders, and cancer [11,12,13,14,15,16,17]. Typically, chaperones are cytoprotective but if qualitatively and/or quantitatively abnormal, they can be pathogenetic and cause diseases, namely chaperonopathies [7,12].
Among the chaperonopathies, those by mistake or collaborationism are most relevant to carcinogenesis because cancer cell growth, proliferation, metastasization, and resistance to anticancer drugs may depend on one or more chaperones. These chaperones, apparently qualitatively normal by available methodology but usually quantitatively increased, help the tumor rather than the host, “mistakenly collaborating” with the enemy. Identification of chaperonopathies by mistake as etiologic–pathogenic factors in cancer is key to the development and application of chaperonotherapy, namely the use of chaperones as agents or targets for treatment [18,19]. Therefore, it is pertinent to investigate the role of the chaperone system in thyroid carcinogenesis, beginning by mapping the chaperones in the thyroid tissue and then assessing their quantitative variations in the different tumors at various times during their development.
Here, we will survey illustrative examples of such studies, (Table 1).

2. Quantitative Changes of Molecular Chaperones and Associated Effects in TC

Molecular chaperones are involved in the maturation and stabilization of proteins, including those that are oncogenic and, therefore, chaperones are often increased in thyroid cancer tissue. Presumably, this increase is a reflection of the cancer cell’s abnormally high need for chaperones required by its high division rate, rapid growth, and dissemination mechanism, all of which require more functional proteins, correctly folded and assembled than a nonmalignant cell. In the following sections, we will briefly discuss some illustrative examples of studies describing the quantitative patterns of molecular chaperones in TC.

2.1. Hsp27

Hsp27 promotes cell survival by inhibiting apoptosis and necrosis, blocking caspase activation [28]. In TC, the Hsp27 increase is correlated with elevated estrogen (E2) levels, which promote growth, invasion, and migration, conferring resistance to apoptosis [29]. In PTC, Erα/SP1 (estrogen receptor/specificity protein 1) can upregulate Hsp27 at the mRNA and protein levels, facilitating proliferation and conferring resistance to apoptosis triggered by TNF-α through interaction with procaspase-3 [29]. Hsp27 interacts with the amino-terminal prodomain of caspase-3, thereby inhibiting the second proteolytic cleavage necessary for caspase-3 activation, Figure 1A [30,31,32]. This mechanism may be the reason why PTC is three times more common in women than in men [33]. Furthermore, in PTC, Hsp27 (HSPB1) is a scaffold protein, which promotes phosphorylation of Akt, inducing cell survival [34]. In this way, Akt, through Bax inhibition, prevents apoptosome formation, Figure 1B [35].
In one illustrative study, the level of Hsp27 was assessed in tumor and nontumor thyroid tissue samples from the same thyroid glands and cultured thyroid cells, by immunohistochemistry and Western blot, respectively [28]. Western blot detected high levels of Hsp27 on different human thyroid cancer cell lines, including the lines K1 (PTC), WRO (FTC), and FRO and KAT18 (ATC). Similarly, immunohistochemical staining showed an increase in the level of antioxidant molecules and Hsp27 in tissue samples from both malignant and benign thyroid tumors, including PTC, FTC, and follicular adenoma (FA), and multinodular goiters, when compared to normal tissue. The same pattern was observed also in human TC cells subjected to oxidative stress [20]. All these data indicate that Hsp27 is elevated in TC cells. Therefore, this chaperone can be considered a candidate biomarker of thyroid malignancy to be monitored in patients.

2.2. Hsp60

Although the level of the molecular chaperone Hsp60 in tumor tissues has been studied in various cancers [36,37,38,39], data on thyroid tumors are scarce; Hsp60 could participate in carcinogenesis after undergoing post-translational modifications, such as nitration, acetylation, oxidation, and ubiquitination, which could lead to mitochondrial dysfunction and thereby facilitate tumorigenesis [40].

2.3. Hsp70

Hsp70 levels are elevated in a variety of tumors, often correlated with tumor grade, metastases, and poor prognosis [41,42]. Members of the Hsp70 family play a variety of roles in the maintenance of protein homeostasis and, therefore, at least some of them must participate in carcinogenesis and can be used as targets for anticancer drugs. For example, VER155008 is a selective Hsc70 inhibitor that induces death of ATC cells, mediated through PI3K/Akt signaling, Figure 2A [32,43].
Mortalin (HSPA9/GRP75) plays a role in MTC cell proliferation and survival and its upregulation is associated with bad prognosis and chemoradiotherapy resistance, as is also the case for other tumors [25]. Mortalin is localized in mitochondria and its depletion is accompanied by depolarization of the mitochondrial membrane, ROS (reactive oxygen species) generation, and apoptotic cell death. Mortalin depletion induces not only cell cycle arrest by altering MEK/ERK signaling but also induces caspase-dependent apoptotic cell death, Figure 2B. These data suggest that mortalin could be a target for treatment, since it is a key regulator of cell signaling and metabolism in MTC [23,32].
Western blot showed elevated levels of mortalin in human MTC, TT, and MZ-CRC-1 cell lines when compared with normal human fibroblasts [23]. In addition, it was revealed by immunohistochemistry that mortalin was increased in human MTC, PTC, FTC, and ATC compared to normal thyroid tissues, suggesting a possible role of the chaperones in carcinogenesis, probably related to its role in maintaining the homeostasis of the proteins involved in mitochondrial bioenergetics and redox balance [23,24]. Mortalin depletion induced cell death and growth arrest in different human cell lines, including PTC (PTC-1 cell line), FTC (FTC133 cell line), and ATC (8505C, and C643 cell lines), and in mouse xenografts, revealing its role in promoting tumor cell survival and proliferation, which prompted the proposal of mortalin as therapeutic target [23,24].
Immunohistochemistry, immunocytochemistry, and immunoblotting were used to assess the levels of the estrogen receptor alpha36 (ERα36), GRP78, and GRP94 in PTC specimens [26]. Their levels were elevated in primary PTC tissues and derived cells, while no follicular cells showed positive ERα36, GRP78, or GRP94 immunostaining in normal thyroid tissues and nodular hyperplasia tissues [26]. Their high levels were found to be strongly associated with aggressive PTC, suggesting these three proteins as possible indicators of PTC aggressiveness measured by estimating extra-thyroid extension (ETE), lymph node metastasis (LNM), distant metastasis (DM), and high tumor node metastasis (TNM) stage [26]. Moreover, it was found that the levels of GRP78 and GRP94 are increased by estrogen E2 [26] as found also in other cancers [44,45,46,47], suggesting a role of E2 in the progression and metastasis of PTC.
The levels of GRP78 assessed by immunostaining in primary tumor and noncancerous thyroid tissues were found significantly higher in MTC specimens than in normal controls, while the levels of Hsp70 and Hsp90 were also elevated in cancerous tissues but not significantly [25].

2.4. Hsp90

Hsp90 levels in TC tumor tissue have been found elevated by comparison with normal peritumoral tissue [48,49]. Generally, Hsp90 increased levels correlated with a poor prognosis [50,51,52]. Hsp90 is involved in the activities of β-catenin, BRAF, AKT, survivin, and some members of the Bcl-2 (B-cell lymphoma 2) complex [32]. Thus, inhibition of Hsp90 induces tumor cell apoptosis, lessening migration and invasion, Figure 3C [49].
Hsp90 regulates hypoxia-inducible factor (HIF)-1α activity, suggesting that it might have a role in PTC tumorigenesis, Figure 3A [50]. Hsp90 regulates telomerase activity in TC [53]. Probably, Hsp90, together with chaperone p23, binds and activates the human telomerase reverse transcriptase (hTERT) [54]. This activation could be an important step in tumorigenesis since the cells gain the ability to proliferate indefinitely and become immortal [55]. For this reason, increased Hsp90 levels and telomerase activity have been described as markers that correlated with malignancy and tumor progression [53,54]. Likewise, it has been shown that levels of p23 and Hsp90 mRNAs were elevated in cancerous thyroid tissue by comparison with non-malignant thyroid tissue, Figure 3B [53].
Figure 3. Hsp90 is involved in cancer development in pathways critical for the cell growth, invasiveness, and survival. Hsp90 regulates the hypoxia-inducible factor (HIF)-1α activity [50] (A), and together with chaperone p23, binds the human telomerase reverse transcriptase (hTERT) [54] (B), causing evasion from apoptosis and metastasis. (C) The components of the PI3K/AKT/mTOR pathway are Hsp90 clients, indicating the chaperone’s role in the regulation of autophagy. Therefore, autophagy has a pro-survival function [32,49]. (D) The tumor necrosis factor receptor-associated protein 1 (TRAP1) is involved in TCs: it is thought to enhance ERK phosphorylation and cell cycle progression, leading to a dampening of apoptosis by activation of the RAS/RAF/ERK pathway [56].
Figure 3. Hsp90 is involved in cancer development in pathways critical for the cell growth, invasiveness, and survival. Hsp90 regulates the hypoxia-inducible factor (HIF)-1α activity [50] (A), and together with chaperone p23, binds the human telomerase reverse transcriptase (hTERT) [54] (B), causing evasion from apoptosis and metastasis. (C) The components of the PI3K/AKT/mTOR pathway are Hsp90 clients, indicating the chaperone’s role in the regulation of autophagy. Therefore, autophagy has a pro-survival function [32,49]. (D) The tumor necrosis factor receptor-associated protein 1 (TRAP1) is involved in TCs: it is thought to enhance ERK phosphorylation and cell cycle progression, leading to a dampening of apoptosis by activation of the RAS/RAF/ERK pathway [56].
Ijms 22 04196 g003
Another member of the Hsp90 family, the TRAP1 (tumor necrosis factor receptor-associated protein 1) chaperone, is also involved in protection from apoptosis, drug resistance, and cell cycle progression [56]. TRAP1 is increased in various human malignancies, including TC, with its levels progressively increasing from normal peritumoral thyroid tissue to PTC tissue [57]. The involvement of TRAP1 in TC occurs via modulation of ERK phosphorylation and cell-cycle progression, leading to increased apoptosis by constitutive activation of the RAS/RAF/ERK pathway, Figure 3D [32,56].
Because various proteins necessary for thyroid cancer growth are Hsp90 clients, inhibition of the chaperone seems a promising anticancer therapy. For instance, the compounds 17-allylamino-17-demethoxygeldanamycin, KU711, and WGA-TA are Hsp90 inhibitors that could interrupt oncogenic pathways in TC [32,58,59]. A natural product, BTIMNP_D004, was evaluated for anticancer activity in the following TC cell lines: papillary cancer cell lines (BCPAP and TPC1), follicular cancer cell lines (FTC133, FTC236, FTC238), medullary cancer cell line (DRO81-1), anaplastic cancer cell line (SWI736), and a fibroblast cell line (MRC-5) as control. The colorimetric assay MTS was used for measuring cell viability/proliferation and evaluating the effect of BTIMNP_D004. It was observed that BTIMNP_D004 inhibited the proliferation TC cells more effectively than the proliferation of fibroblasts. Levels of Hsp90 were evaluated by Western blot and showed that in FTC133 cells treated with the drug, Hsp90α increased whereas Hsp90β decreased, but in BCPAP cells, the levels of Hsp90α decreased. Likewise, it was demonstrated that the levels of ERK decreased in BCPAP cells, and the levels of AKT decreased in FTC133 cells [60].
Another two Hsp90 inhibitors, KU711 and WGA-TA, were tested in the TC cell lines TPC1, FTC238, WRO, and ACT1 [59]. Migration and invasion assays, with the use of Boyden chambers and matrigel chambers, respectively, were conducted to test the effect of KU711 and WGA-TA on the cell lines, and it was found that the drug caused a decrease in migration and invasion. Western blot analyses of Hsp90 and its client proteins BRAF and AKT in TC cell lines treated with the drugs were also conducted. The results showed that the levels of Hsp90 were stable compared with the control, whereas the levels of AKT and BRAF decreased [59].
Immunoblot analysis to assess the levels of TRAP1 and the client proteins BRAF and ERK, with or without TRAP1 silencing, was conducted using the FTC cell lines ML1 and FTC133, the PTC cell line BCPAP, and the ATC cell lines BHT101 and CAL62. This study showed that TRAP1 silencing inhibited both BRAF and ERK pathways in TC [56].

3. Various Chaperones Assessed Simultaneously

Recently, our research group has estimated the levels of Hsp27, Hsp60, Hsp70, and Hsp90 in thyroid cancerous tissues and adjacent peritumoral tissues from patients with FA or FTC by immunohistochemistry. The level of Hsp27 was high but did not show a significant quantitative variation in cancerous tissues compared to the corresponding normal parenchyma, whereas Hsp60, Hsp70, and Hsp90 were increased in FTC compared to FA and/or to adjacent normal parenchyma [22]. In another study, the immunohistochemical levels of Hsp27, Hsp60, and Hsp90, but not Hsp70, were high in PTC as compared with benign goiters and normal peritumoral tissues [21]. Both works also reported the intracellular distribution of each Hsp, showing not only an increased level in cancerous compared to normal tissues, but also an altered localization, with accumulation in the cytoplasm and plasma cell membrane in cancerous specimens [21,22]. These results agree with other works showing a change in the cellular distribution or even extracellular secretion of Hsps during carcinogenesis processes [61,62,63,64,65].
A comprehensive immunomorphological description of the distribution of molecular chaperones in TC enhances the visualization of the malignant patterns and constitutes one of the benefits of immunohistochemistry [21,22]. For example, immunohistochemistry was used to evaluate the levels of Hsp70 and Ki-67 (a proliferation marker) and their subcellular localization in relation to clinical and morphological parameters of PTC [66]. The immunohistochemical detection of Hsp70 showed that it was differentially localized in tumor cells (cytoplasm, nucleus, nucleolus, or a combination of these different sites), and that its nuclear translocation was correlated with the stage of the carcinogenic process and with prognosis: the nuclear translocation of the protein was found higher in samples with stage IV and with an unfavorable prognosis.

4. Exosomes and TC

Almost all cell types release extracellular vesicles (EVs), such as exosomes, including cancer cells [67,68]. It is assumed that exosomes released by cancer cells can interact with cells around the tumor, both cancerous and stromal, and with distant cells through the circulation, possibly promoting cancer development. The latter could be mediated by one or more of the following mechanisms: enhancement of cell proliferation and invasiveness, stimulation of angiogenesis, induction of immune suppression, and remodeling of the tumor microenvironment to favor metastasis [69,70,71]. These mechanisms and effects are still under investigation. Despite the current uncertainties, circulating EVs and their molecular cargo can be considered a promising source of biomarkers for the early detection, diagnosis, monitoring, and follow up of different types of cancer. Unfortunately, unlike other tumors for which there is considerable information, such as glioblastoma and prostate, lung, ovarian, and breast cancers [72,73,74,75,76,77,78], only two studies have been reported on exosomal molecular chaperones in TCs and their potential applications [21,27]. The levels of Hsp27, Hsp60, and Hsp90 were increased in the exosomes from the plasma of PTC patients compared with the same patients after thyroidectomy and patients with benign goiter [21]. In addition, it was demonstrated that Hsp27 was increased in exosomes from the plasma of PTC patients with lymph node metastases, suggesting the potential of this molecular chaperone as an indicator of lymphatic metastasis [27].

5. Conclusions and Perspectives for the Future

The results discussed in the preceding paragraphs show that quantification of chaperones by immunohistochemistry can be useful for differential diagnosis and to follow their changes in relation to the evolution of the disease. Furthermore, immunohistochemistry provides information on the distribution of the chaperones inside and outside the cells with the potential for revealing patterns that distinguish different tumors from each other. In addition, the data suggest that chaperones may play an etiologic–pathogenic role and encourages research aimed at elucidating the molecular mechanisms underpinning the quantitative changes of the chaperones and their effects on the tumor cell.
Tumor classification has historically been based on the primary anatomic site in which the tumor occurs and on its morphologic and histologic phenotypes. However, histopathology alone cannot accurately predict the prognosis and treatment response of individual cancers [79]. It is necessary to quantify morphological and molecular features of cells and tissues under physiological and pathological conditions, noting that each cell unit possesses the nucleus and cytoplasm, which structurally and functionally cooperate for gene expression and cellular dynamics [80]. For this reason, molecular and cell morphological methods have increased in number and versatility in recent years. In this review, we discuss studies of the quantitative profiles of chaperones in thyroid tissue, of cancerous compared to normal. The results provide an incentive to progress toward elucidation of the molecular mechanisms underpinning the observed quantitative variations. By what mechanism do chaperones augment (or decrease) in cancer cells? Likewise, the following question arises: what role, if any, do chaperones play in tumor initiation and progression? It is possible that the changes observed in the levels of chaperones in thyroid tissue, particularly those indicating an increase in quantity of chaperone molecules inside the tumor cells, are the consequence of the special needs of the cancer cell with its high proliferation rate and metabolism in addition to its dissemination tendency. Higher quantities of correctly folded proteins and their functional assemblages are required by cancer cells as compared to normal ones, which call for higher quantities of chaperones. Thus, chaperones do play a role in carcinogenesis by assisting cancer cells so they can successfully perform key functions characteristic of malignancy, which they could not perform adequately if chaperones were scarce. Thyroid cancer, at least some of its forms, can therefore be considered chaperonopathies by mistake or collaborationism. A normal chaperone—normal in so far as can be determined by current technology—mistakenly helps, so to speak, the enemy; it collaborates with the enemy rather than the contrary. The importance of this concept resides in the fact that it alerts physicians and researchers to the possibility of developing/applying chaperonotherapy, namely the use of therapeutic strategies and compounds targeting the mistaken chaperones. This strategy is exemplified by studies with Hsp90 inhibitors, which have been proposed as anticancer drugs in different forms of TCs since inhibition of the chaperone causes the simultaneous degradation of multiple proteins that are Hsp90 clients involved in oncogenic signaling pathways [17]. For instance, the novel non-geldanamycin Hsp90 inhibitor NVP-AUY922 induced apoptosis in PTC cells and death by disrupting the complex Hsp90/survivin, thus causing the downregulation of survivin [49]. Likewise, Hsp90 inhibition has been proposed to enhance the effect of other anticancer drugs [43].

Author Contributions

Conceptualization, F.R. and L.P.; resources, A.P.; writing—original draft preparation, L.P.; F.R.; A.M.V.; R.S.; writing—review and editing, F.R.; C.C., G.G., E.C.d.M., and A.J.L.M.; supervision, F.B.; E.C.d.M., and A.J.L.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

A.J.L.M and E.C.d.M. were partially supported by IMET and IEMEST. This is IMET contribution number IMET 21-007.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AKTserine/threonine protein kinase B
ATCanaplastic/undifferentiated thyroid carcinoma
Bcl-2B-cell lymphoma 2
DMdistant metastasis
ERKextracellular signal-regulated kinases
Erα/SP1estrogen receptor/specificity protein 1
ERα36estrogen receptor alpha36
ETEextra-thyroid extension
EVsextracellular vesicles
FAfollicular adenoma
FNAfine-needle aspiration
FTCfollicular thyroid carcinoma
(HIF)-1αhypoxia-inducible factor-1α
hTERThuman telomerase reverse transcriptase
LNMlymph node metastasis
MEKmitogen-activated protein kinase kinase
MTCmedullary thyroid carcinoma
mTORmammalian target of rapamycin
PDTCpoorly differentiated thyroid carcinoma
PI3Kphosphatidylinositol 3-kinase
PTCpapillary thyroid carcinoma
RAFproto-oncogene serine/threonine protein kinase
ROSreactive oxygen species
TCthyroid cancer (or thyroid carcinoma)
TNMtumor node metastasis stage
TRAP1tumor necrosis factor receptor-associated protein 1

References

  1. Abooshahab, R.; Gholami, M.; Sanoie, M.; Azizi, F.; Hedayati, M. Advances in metabolomics of thyroid cancer diagnosis and metabolic regulation. Endocrine 2019, 65, 1–14. [Google Scholar] [CrossRef] [PubMed]
  2. Lebastchi, A.H.; Callender, G.G. Thyroid cancer. Curr. Probl. Cancer 2014, 38, 48–74. [Google Scholar] [CrossRef]
  3. Asa, S.L. The Current Histologic Classification of Thyroid Cancer. Endocrinol. Metab. Clin. N. Am. 2019, 48, 1–22. [Google Scholar] [CrossRef]
  4. Grogan, R.H.; Mitmaker, E.J.; Clark, O.H. The evolution of biomarkers in thyroid cancer-from mass screening to a personalized biosignature. Cancers 2010, 2, 885–912. [Google Scholar] [CrossRef] [Green Version]
  5. Feldkamp, J.; Führer, D.; Luster, M.; Musholt, T.J.; Spitzweg, C.; Schott, M. Fine Needle Aspiration in the Investigation of Thyroid Nodules. Dtsch. Aerzteblatt Online 2016, 113, 353–359. [Google Scholar] [CrossRef] [Green Version]
  6. Moses, W.; Weng, J.; Sansano, I.; Peng, M.; Khanafshar, E.; Ljung, B.-M.; Duh, Q.-Y.; Clark, O.H.; Kebebew, E. Molecular Testing for Somatic Mutations Improves the Accuracy of Thyroid Fine-needle Aspiration Biopsy. World J. Surg. 2010, 34, 2589–2594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Macario, A.J.L.; Conway de Macario, E. Sick chaperones, cellular stress, and disease. N. Engl. J. Med. 2005, 353, 1489–1501. [Google Scholar] [CrossRef]
  8. Macario, A.J.L.; Conway de Macario, E. Chaperone proteins and Chaperonopathies. In Handbook of Stress, Volume 3, Stress Physiology, Biochemistry, and Pathology; Fink, G., Ed.; Elsevier/Academic Press: Cambridge, MA, USA, 2019; Chapter 12; pp. 135–152. [Google Scholar]
  9. Kocaturk, N.M.; Gozuacik, D. Crosstalk Between Mammalian Autophagy and the Ubiquitin-Proteasome System. Front. Cell Dev. Biol. 2018, 6, 128. [Google Scholar] [CrossRef]
  10. Tekirdag, K.; Cuervo, A.M. Chaperone-mediated autophagy and endosomal microautophagy: Jointed by a chaperone. J. Biol. Chem. 2018, 293, 5414–5424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Henderson, B.; Fares, M.A.; Lund, P.A. Chaperonin 60: A paradoxical, evolutionarily conserved protein family with multiple moonlighting functions. Biol. Rev. Camb. Philos. Soc. 2013, 88, 955–987. [Google Scholar] [CrossRef]
  12. Macario, A.J.L.; Conway de Macario, E.; Cappello, F. The Chaperonopathies. Diseases with Defective Molecular Chaperones; Springer: Dordrecht, The Netherlands; Heidelberg, Germany; New York, NY, USA; London, UK, 2013. [Google Scholar]
  13. Calderwood, S.K.; Gong, J. Heat shock proteins promote cancer: It’s a protection racket. Trends Biochem. Sci. 2016, 41, 311–323. [Google Scholar] [CrossRef] [Green Version]
  14. Jeffery, C.J. Protein moonlighting: What is it, and why is it important? Philos. Trans. R. Soc. B Biol. Sci. 2018, 373, 20160523. [Google Scholar] [CrossRef] [PubMed]
  15. Saini, J.; Sharma, P.K. Clinical, Prognostic and therapeutic significance of heat shock proteins in cancer. Curr. Drug Targets 2018, 19, 1478–1490. [Google Scholar] [CrossRef] [PubMed]
  16. Milani, A.; Basirnejad, M.; Bolhassani, A. Heat-shock proteins in diagnosis and treatment: An overview of different biochemical and immunological functions. Immunotherapy 2019, 11, 215–239. [Google Scholar] [CrossRef]
  17. Yun, C.W.; Kim, H.J.; Lim, J.H.; Lee, S.H. Heat Shock Proteins: Agents of Cancer Development and Therapeutic Targets in Anti-Cancer Therapy. Cells 2019, 9, 60. [Google Scholar] [CrossRef] [Green Version]
  18. Cappello, F.; Marino Gammazza, A.; Palumbo Piccionello, A.; Campanella, C.; Pace, A.; Conway de Macario, E.; Macario, A.J.L. Hsp60 chaperonopathies and chaperonotherapy: Targets and agents. Expert Opin. Ther. Targets 2013, 18, 185–208. [Google Scholar] [CrossRef]
  19. Meng, Q.; Li, B.X.; Xiao, X. Toward developing chemical modulators of Hsp60 as potential therapeutics. Front. Mol. Biosci. 2018, 5, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Wang, S.; Yang, S.; Vlantis, A.C.; Liu, S.Y.; Ng, E.K.; Chan, A.B.; Wu, J.; Du, J.; Wei, W.; Liu, X.; et al. Expression of antioxidant molecules and heat shock protein 27 in thyroid tumors. J. Cell. Biochem. 2016, 117, 2473–2481. [Google Scholar] [CrossRef]
  21. Caruso Bavisotto, C.; Cipolla, C.; Graceffa, G.; Barone, R.; Bucchieri, F.; Bulone, D.; Cabibi, D.; Campanella, C.; Marino Gammazza, A.; Pitruzzella, A.; et al. Immunomorphological pattern of molecular chaperones in normal and pathological thyroid tissues and circulating exosomes: potential use in clinics. Int. J. Mol. Sci. 2019, 20, 4496. [Google Scholar] [CrossRef] [Green Version]
  22. Pitruzzella, A.; Paladino, L.; Vitale, A.M.; Martorana, S.; Cipolla, C.; Graceffa, G.; Cabibi, D.; David, S.; Fucarino, A.; Bucchieri, F.; et al. Quantitative immunomorphological analysis of heat shock proteins in thyroid follicular adenoma and carcinoma tissues reveals their potential for differential diagnosis and points to a role in carcinogenesis. Appl. Sci. 2019, 9, 4324. [Google Scholar] [CrossRef] [Green Version]
  23. Starenki, D.; Hong, S.-K.; Lloyd, R.V.; Park, J.-I. Mortalin (GRP75/HSPA9) upregulation promotes survival and proliferation of medullary thyroid carcinoma cells. Oncogene 2014, 34, 4624–4634. [Google Scholar] [CrossRef] [Green Version]
  24. Starenki, D.; Sosonkina, N.; Hong, S.K.; Lloyd, R.V.; Park, J.I. Mortalin (GRP75/HSPA9) promotes survival and proliferation of thyroid carcinoma cells. Int. J. Mol. Sci. 2019, 20, 2069. [Google Scholar] [CrossRef] [Green Version]
  25. Soudry, E.; Shavit, S.S.; Hardy, B.; Morgenstern, S.; Hadar, T.; Feinmesser, R. Heat shock proteins HSP90, HSP70 and GRP78 expression in medullary thyroid carcinoma. Ann. Diagn. Pathol. 2017, 26, 52–56. [Google Scholar] [CrossRef] [PubMed]
  26. Dai, Y.-J.; Qiu, Y.-B.; Jiang, R.; Xu, M.; Liao, L.-Y.; Chen, G.G.; Liu, Z.-M. Concomitant high expression of ERα36, GRP78 and GRP94 is associated with aggressive papillary thyroid cancer behavior. Cell. Oncol. 2018, 41, 269–282. [Google Scholar] [CrossRef] [PubMed]
  27. Luo, D.; Zhan, S.; Xia, W.; Huang, L.; Ge, W.; Wang, T. Proteomics study of serum exosomes from papillary thyroid cancer patients. Endocr. Relat. Cancer 2018, 25, 879–891. [Google Scholar] [CrossRef] [PubMed]
  28. Garrido, C.; Brunet, M.; Didelot, C.; Zermati, Y.; Schmitt, E.; Kroemer, G. Heat shock proteins 27 and 70: Anti-apoptotic proteins with tumorigenic properties. Cell Cycle 2006, 5, 2592–2601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Mo, X.-M.; Li, L.; Zhu, P.; Dai, Y.-J.; Zhao, T.-T.; Liao, L.-Y.; Chen, G.G.; Liu, Z.-M. Up-regulation of Hsp27 by ERα/Sp1 facilitates proliferation and confers resistance to apoptosis in human papillary thyroid cancer cells. Mol. Cell. Endocrinol. 2016, 431, 71–87. [Google Scholar] [CrossRef]
  30. Pandey, P.M.; Farber, R.; Nakazawa, A.; Kumar, S.; Bharti, A.C.; Nalin, C.M.; Weichselbaum, R.R.; Kufe, D.; Kharbanda, S. Hsp27 functions as a negative regulator of cytochrome c-dependent activation of procaspase-3. Oncogene 2000, 19, 1975–1981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Voss, O.H.; Batra, S.; Kolattukudy, S.J.; Gonzalez-Mejia, M.E.; Smith, J.B.; Doseff, A.I. Binding of Caspase-3 Prodomain to Heat Shock Protein 27 Regulates Monocyte Apoptosis by Inhibiting Caspase-3 Proteolytic Activation. J. Biol. Chem. 2007, 282, 25088–25099. [Google Scholar] [CrossRef] [Green Version]
  32. Lettini, G.; Pietrafesa, M.; Lepore, S.; Maddalena, F.; Crispo, F.; Sgambato, A.; Esposito, F.; Landriscina, M. Heat shock proteins in thyroid malignancies: Potential therapeutic targets for poorly differentiated and anaplastic tumours? Mol. Cell. Endocrinol. 2020, 502, 110676. [Google Scholar] [CrossRef]
  33. Derwahl, M.; Nicula, D. Estrogen and its role in thyroid cancer. Endocr. Relat. Cancer 2014, 21, T273–T283. [Google Scholar] [CrossRef] [PubMed]
  34. Li, Y.; Yang, Q.; Guan, H.; Shi, B.; Ji, M.; Hou, P. ZNF677 Suppresses Akt Phosphorylation and Tumorigenesis in Thyroid Cancer. Cancer Res. 2018, 78, 5216–5228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Jutabha, P.; Kanai, Y.; Hosoyamada, M.; Chairoungdua, A.; Kim, D.K.; Iribe, Y.; Babu, E.; Kim, J.Y.; Anzai, N.; Chatsudthipong, V.; et al. Identification of a Novel Voltage-driven Organic Anion Transporter Present at Apical Membrane of Renal Proximal Tubule. J. Biol. Chem. 2003, 278, 27930–27938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Cappello, F.; Czarnecka, A.M.; La Rocca, G.; Di Stefano, A.; Zummo, G.; Macario, A.J.L. Hsp60 and Hspl0 as antitumor molecular agents. Cancer Biol. Ther. 2007, 6, 487–489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Cappello, F.; Bellafiore, M.; Palma, A.; Marciano, V.; Martorana, G.; Belfiore, P.; Martorana, A.; Farina, F.; Zummo, G.; Bucchieri, F. Expression of 60-kD heat shock proteinin creases during carcinogenesis in the uterine exocervix. Pathobiology 2002, 70, 83–88. [Google Scholar] [CrossRef] [PubMed]
  38. Cappello, F.; Bellafiore, M.; Palma, A.; David, S.; Marcianò, V.; Bartolotta, T.; Sciumè, C.; Modica, G.; Farina, F.; Zummo, G.; et al. 60kDa chaperonin (HSP60) is over-expressed during colorectal carcinogenesis. Eur. J. Histochem. 2003, 47, 105–110. [Google Scholar] [CrossRef]
  39. Cappello, F.; David, S.; Rappa, F.; Bucchieri, F.; Marasà, L.; Bartolotta, T.E.; Farina, F.; Zummo, G. The expression of HSP60 and HSP10 in large bowel carcinomas with lymph node metastase. BMC Cancer 2005, 5, 139. [Google Scholar] [CrossRef] [Green Version]
  40. Caruso Bavisotto, C.; Alberti, G.; Vitale, A.M.; Paladino, L.; Campanella, C.; Rappa, F.; Gorska, M.; Conway de Macario, E.; Cappello, F.; Macario, A.J.L.; et al. Hsp60 post-translational modifications: Functional and pathological consequences. Front. Mol. Biosci. 2020, 7, 95. [Google Scholar] [CrossRef]
  41. Jolly, C.; Morimoto, R.I. Role of the heat shock response and molecular chaperones in oncogenesis and cell death. J. Natl. Cancer Inst. 2000, 92, 1564–1572. [Google Scholar] [CrossRef] [Green Version]
  42. Yaglom, J.A.; Wang, Y.; Li, A.; Li, Z.; Monti, S.; Alexandrov, I.; Lu, X.; Sherman, M.Y. Cancer cell responses to Hsp70 inhibitor JG-98: Comparison with Hsp90 inhibitors and finding synergistic drug combinations. Sci. Rep. 2018, 8, 3010. [Google Scholar] [CrossRef] [Green Version]
  43. Kim, S.H.; Kang, J.G.; Kim, C.S.; Ihm, S.-H.; Choi, M.G.; Yoo, H.J.; Lee, S.J. Hsp70 inhibition potentiates radicicol-induced cell death in anaplastic thyroid carcinoma cells. Anticancer Res. 2014, 34, 4829–4837. [Google Scholar] [PubMed]
  44. Kiang, J.G.; Gist, I.D.; Tsokos, G.C. 17 beta-estradiol-induced increases in glucose-regulated protein 78kD and 94kD protect human breast cancer T47-D cells from thermal injury. Chin. J. Physiol. 1997, 40, 213–219. [Google Scholar] [PubMed]
  45. Zheng, H.-C.; Takahashi, H.; Li, X.-H.; Hara, T.; Masuda, S.; Guan, Y.-F.; Takano, Y. Overexpression of GRP78 and GRP94 are markers for aggressive behavior and poor prognosis in gastric carcinomas. Hum. Pathol. 2008, 39, 1042–1049. [Google Scholar] [CrossRef]
  46. Scriven, P.; Coulson, S.; Haines, R.; Balasubramanian, S.; Cross, S.; Wyld, L. Activation and clinical significance of the unfolded protein response in breast cancer. Br. J. Cancer 2009, 101, 1692–1698. [Google Scholar] [CrossRef] [Green Version]
  47. Delie, F.; Petignat, P.; Cohen, M. GRP78 Protein expression in ovarian cancer patients and perspectives for a drug-targeting approach. J. Oncol. 2012, 2012, 1–5. [Google Scholar] [CrossRef]
  48. Pearl, L.H.; Prodromou, C.; Workman, P. The Hsp90 molecular chaperone: An open and shut case for treatment. Biochem. J. 2008, 410, 439–453. [Google Scholar] [CrossRef] [Green Version]
  49. Liu, J.; Sun, W.; Dong, W.; Wang, Z.; Qin, Y.; Zhang, T.; Zhang, H. HSP90 inhibitor NVP-AUY922 induces cell apoptosis by disruption of the survivin in papillary thyroid carcinoma cells. Biochem. Biophys. Res. Commun. 2017, 487, 313–319. [Google Scholar] [CrossRef]
  50. Mo, J.-H.; Choi, I.J.; Jeong, W.-J.; Jeon, E.-H.; Ahn, S.-H. HIF-1α and HSP90: Target molecules selected from a tumorigenic papillary thyroid carcinoma cell line. Cancer Sci. 2012, 103, 464–471. [Google Scholar] [CrossRef]
  51. Wang, J.; Cui, S.; Zhang, X.; Wu, Y.; Tang, H. High expression of heat shock protein 90 is associated with tumor aggressiveness and poor prognosis in patients with advanced gastric cancer. PLoS ONE 2013, 8, e62876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Zhang, S.; Guo, S.; Li, Z.; Li, D.; Zhan, Q. High expression of HSP90 is associated with poor prognosis in patients with colorectal cancer. PeerJ 2019, 7, e7946. [Google Scholar] [CrossRef] [PubMed]
  53. Boltze, C.; Schneider-Stock, R.; Roessner, A.; Quednow, C.; Hoang-Vu, C. Function of HSP90 and p23 in the telomerase complex of thyroid tumors. Pathol. Res. Pract. 2003, 199, 573–579. [Google Scholar] [CrossRef]
  54. Holt, S.E.; Aisner, D.L.; Baur, J.; Tesmer, V.M.; Dy, M.; Ouellette, M.; Trager, J.B.; Morin, G.B.; Toft, D.O.; Shay, J.W.; et al. Functional requirement of p23 and Hsp90 in telomerase complexes. Genes Dev. 1999, 13, 817–826. [Google Scholar] [CrossRef] [PubMed]
  55. Hoang-Vu, C.; Boltze, C.; Gimm, O.; Poremba, C.; Dockhorn-Dworniczak, B.; Köhrle, J.; Rath, F.-W.; Dralle, H. Expression of telomerase genes in thyroid carcinoma. Int. J. Oncol. 2002, 21, 265–272. [Google Scholar] [CrossRef] [PubMed]
  56. Palladino, G.; Notarangelo, T.; Pannone, G.; Piscazzi, A.; Lamacchia, O.; Sisinni, L.; Spagnoletti, G.; Toti, P.; Santoro, A.; Storto, G.; et al. TRAP1 regulates cell cycle and apoptosis in thyroid carcinoma cells. Endocr. Relat. Cancer 2016, 23, 699–709. [Google Scholar] [CrossRef] [Green Version]
  57. Rasola, A.; Neckers, L.; Picard, D. Mitochondrial oxidative phosphorylation TRAP(1)ped in tumor cells. Trends Cell Biol. 2014, 24, 455–463. [Google Scholar] [CrossRef] [PubMed]
  58. Braga-Basaria, M.; Hardy, E.; Gottfried, R.; Burman, K.D.; Saji, M.; Ringel, M.D. 17-Allylamino-17-Demethoxygeldanamycin activity against thyroid cancer cell lines correlates with heat shock protein 90 levels. J. Clin. Endocrinol. Metab. 2004, 89, 2982–2988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. White, P.T.; Subramanian, C.; Zhu, Q.; Zhang, H.; Zhao, H.; Gallagher, R.J.; Timmermann, B.N.; Blagg, B.S.J.; Cohen, M.S. Novel HSP90 inhibitors effectively target functions of thyroid cancer stem cell preventing migration and invasion. Surgery 2016, 159, 142–151. [Google Scholar] [CrossRef] [Green Version]
  60. Samadi, A.; Loo, P.; Mukerji, R.; O’Donnell, G.; Tong, X.; Timmermann, B.N.; Cohen, M.S. A novel HSP90 modulator with selective activity against thyroid cancers in vitro. Surgery 2009, 146, 1196–1207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Campanella, C.; Bucchieri, F.; Ardizzone, N.M.; Marino Gammazza, A.; Montalbano, A.; Ribbene, A.; Di Felice, V.; Bellafiore, M.; David, S.; Rappa, F.; et al. Upon oxidative stress, the antiapoptotic Hsp60/procaspase-3 complex persists in mucoepidermoid carcinoma cells. Eur. J. Histochem. 2008, 52, 221–228. [Google Scholar] [CrossRef] [Green Version]
  62. Campanella, C.; Bucchieri, F.; Merendino, A.M.; Fucarino, A.; Burgio, G.; Corona, D.F.; Barbieri, G.; David, S.; Farina, F.; Zummo, G.; et al. The odyssey of Hsp60 from tumor cells to other destinationsincludes plasma membrane-associated stages and Golgi and exosomal protein-trafficking modalities. PLoS ONE 2012, 7, e42008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Campanella, C.; Rappa, F.; Sciumè, C.; Marino Gammazza, A.; Barone, R.; Bucchieri, F.; David, S.; Curcurù, G.; Caruso Bavisotto, C.; Pitruzzella, A.; et al. Heat shock protein 60 levels in tissue and circulating exosomes in human large bowel cancer before and after ablative surgery. Cancer 2015, 121, 3230–3239. [Google Scholar] [CrossRef] [PubMed]
  64. Santos, T.G.; Martins, V.R.; Hajj, G.N.M. Unconventional secretion of heat shock proteins in cancer. Int. J. Mol. Sci. 2017, 18, 946. [Google Scholar] [CrossRef] [Green Version]
  65. Shevtsov, M.; Balogi, Z.; Khachatryan, W.; Gao, H.; Vígh, L.; Multhoff, G. Membrane-associated heat shock proteins in oncology: from basic research to new theranostic targets. Cells 2020, 9, 1263. [Google Scholar] [CrossRef] [PubMed]
  66. Avdalyan, A.M.; Ivanov, A.A.; Lushnikova, E.L.; Molodykh, O.P.; Vikhlyanov, I.V. The Relationship of immunoexpression of Ki-67 and Hsp70 with clinical and morphological parameters and prognosis of papillary thyroid cancer. Bull. Exp. Biol. Med. 2020, 168, 688–693. [Google Scholar] [CrossRef]
  67. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [Green Version]
  68. Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular vesicles: Composition, biological relevance, and methods of study. Bioscience 2015, 65, 783–797. [Google Scholar] [CrossRef] [Green Version]
  69. Vader, P.; Breakefield, X.O.; Wood, M.J. Extracellular vesicles: Emerging targets for cancer therapy. Trends Mol. Med. 2014, 20, 385–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Rappa, G.; Puglisi, C.; Santos, M.F.; Forte, S.; Memeo, L.; Lorico, A. Extracellular vesicles from thyroid carcinoma: The new frontier of liquid biopsy. Int. J. Mol. Sci. 2019, 20, 1114. [Google Scholar] [CrossRef] [Green Version]
  71. Feng, K.; Ma, R.; Zhang, L.; Li, H.; Tang, Y.; Du, G.; Niu, D.; Yin, D. The role of exosomes in thyroid cancer and their potential clinical application. Front. Oncol. 2020, 10, 596132. [Google Scholar] [CrossRef]
  72. Gobbo, J.; Marcion, G.; Cordonnier, M.; Dias, A.M.M.; Pernet, N.; Hammann, A.; Richaud, S.; Mjahed, H.; Isambert, N.; Clausse, V.; et al. Restoring anticancer immune response by targeting tumor-derived exosomes with a HSP70 peptide aptamer. J. Natl. Cancer Inst. 2015, 108. [Google Scholar] [CrossRef]
  73. Li, X.; Wang, S.; Zhu, R.; Li, H.; Han, Q.; Zhao, R.C. Lung tumor exosomes induce a pro-inflammatory phenotype in mesenchymal stem cells via NFκB-TLR signaling pathway. J. Hematol. Oncol. 2016, 9, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Stope, M.B.; Klinkmann, G.; Diesing, K.; Koensgen, D.; Burchardt, M.; Mustea, A. Heat shock protein HSP27 secretion by ovarian cancer cells is linked to intracellular expression levels, occurs independently of the endoplasmic reticulum pathway and HSP27′s phosphorylation status, and is mediated by exosome liberation. Dis. Mark. 2017, 2017, 1–12. [Google Scholar] [CrossRef]
  75. Caruso Bavisotto, C.; Graziano, F.; Rappa, F.; Marino Gammazza, A.; Logozzi, M.; Fais, S.; Maugeri, R.; Bucchieri, F.; Conway de Macario, E.; Macario, A.J.L.; et al. Exosomal chaperones and miRNAs in Gliomagenesis: State-of-Art and theranostics perspectives. Int. J. Mol. Sci. 2018, 19, 2626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Wyciszkiewicz, A.; Kalinowska-Łyszczarz, A.; Nowakowski, B.; Kaźmierczak, K.; Osztynowicz, K.; Michalak, S. Expression of small heat shock proteins in exosomes from patients with gynecologic cancers. Sci. Rep. 2019, 9, 9817. [Google Scholar] [CrossRef] [Green Version]
  77. Lorenc, T.; Klimczyk, K.; Michalczewska, I.; Słomka, M.; Kubiak-Tomaszewska, G.; Olejarz, W. Exosomes in prostate cancer diagnosis, prognosis and therapy. Int. J. Mol. Sci. 2020, 21, 2118. [Google Scholar] [CrossRef] [Green Version]
  78. Naryzhny, S.; Volnitskiy, A.; Kopylov, A.; Zorina, E.; Kamyshinsky, R.; Bairamukov, V.; Garaeva, L.; Shlikht, A.; Shtam, T. Proteome of glioblastoma-derived exosomes as a source of biomarkers. Biomedicine 2020, 8, 216. [Google Scholar] [CrossRef]
  79. Sherman, M.E.; Troester, M.A.; Hoadley, K.A.; Anderson, W.F. Morphological and molecular classification of human cancer. In Cancer Epidemiology and Prevention; Oxford University Press: Oxford, UK, 2017. [Google Scholar]
  80. Yasuda, Y.; Tokunaga, K.; Koga, T.; Sakamoto, C.; Goldberg, I.G.; Saitoh, N.; Nakao, M. Computational analysis of morphological and molecular features in gastric cancer tissues. Cancer Med. 2020, 9, 2223–2234. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Hsp27 regulates apoptosis. (A) Hsp27 can be upregulated by estrogen (E2) levels in human papillary thyroid cancer (PTC) cells that have a higher level of ERα than of ERβ. Overexpression of Hsp27 facilitates proliferation, conferring resistance to apoptosis through interaction with procaspase-3 [29,31]. (B) Hsp27(shown as HSPB1), acting as scaffold protein, modulates apoptosis through interaction with Akt. In this way, Hsp27 increases the interaction of Akt with its substrate, Bax, inhibiting Bax activation, oligomerization, and translocation to the mitochondria, thus inhibiting the release of cytochrome C. This prevents the correct formation and function of the apoptosome complex [34,35].
Figure 1. Hsp27 regulates apoptosis. (A) Hsp27 can be upregulated by estrogen (E2) levels in human papillary thyroid cancer (PTC) cells that have a higher level of ERα than of ERβ. Overexpression of Hsp27 facilitates proliferation, conferring resistance to apoptosis through interaction with procaspase-3 [29,31]. (B) Hsp27(shown as HSPB1), acting as scaffold protein, modulates apoptosis through interaction with Akt. In this way, Hsp27 increases the interaction of Akt with its substrate, Bax, inhibiting Bax activation, oligomerization, and translocation to the mitochondria, thus inhibiting the release of cytochrome C. This prevents the correct formation and function of the apoptosome complex [34,35].
Ijms 22 04196 g001
Figure 2. Hsp70 is involved in cancer development at multiple steps. (A) Hsp70 induces evasion from apoptosis, blocking PI3K/Akt signaling [43]. (B) Mortalin (also called HSPA9 or GRP75), localized in mitochondria, alters MEK/ERK signaling, ensuring cell survival [23].
Figure 2. Hsp70 is involved in cancer development at multiple steps. (A) Hsp70 induces evasion from apoptosis, blocking PI3K/Akt signaling [43]. (B) Mortalin (also called HSPA9 or GRP75), localized in mitochondria, alters MEK/ERK signaling, ensuring cell survival [23].
Ijms 22 04196 g002
Table 1. Molecular chaperone levels in thyroid tumors.
Table 1. Molecular chaperone levels in thyroid tumors.
ChaperoneTumorQuantitative LevelReference
ImmunohistochemistryHsp27
(HSPB1)
FA 1Increased[20]
FTCIncreased[20]
PTCIncreased[21]
Hsp60FTCIncreased[22]
PTCIncreased[21]
Hsp70FTCIncreased[22]
Mortalin
(HSPA9; GRP75)
ATCIncreased[23,24]
FTCIncreased
MTCIncreased
PTCIncreased
GRP78MTCIncreased[25]
PTCIncreased[26]
GRP94PTCIncreased[26]
Hsp90FTCIncreased[22]
PTCIncreased[21]
Biochemistry; immunochemistryHsp27ATCIncreased[20]
FTCIncreased
PTCIncreased
Mortalin
(HSPA9; GRP75)
MTCIncreased[23]
GRP78PTCIncreased[26]
GRP94PTCIncreased[26]
ExosomesHsp27PTCIncreased[21,27]
Hsp60PTCIncreased[21]
Hsp90PTCIncreased[21]
1 Abbreviations: FA, follicular adenoma; ATC, anaplastic thyroid carcinoma; FTC, follicular thyroid carcinoma; MTC, medullary thyroid cancer; PTC, papillary thyroid carcinoma.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Paladino, L.; Vitale, A.M.; Santonocito, R.; Pitruzzella, A.; Cipolla, C.; Graceffa, G.; Bucchieri, F.; Conway de Macario, E.; Macario, A.J.L.; Rappa, F. Molecular Chaperones and Thyroid Cancer. Int. J. Mol. Sci. 2021, 22, 4196. https://doi.org/10.3390/ijms22084196

AMA Style

Paladino L, Vitale AM, Santonocito R, Pitruzzella A, Cipolla C, Graceffa G, Bucchieri F, Conway de Macario E, Macario AJL, Rappa F. Molecular Chaperones and Thyroid Cancer. International Journal of Molecular Sciences. 2021; 22(8):4196. https://doi.org/10.3390/ijms22084196

Chicago/Turabian Style

Paladino, Letizia, Alessandra Maria Vitale, Radha Santonocito, Alessandro Pitruzzella, Calogero Cipolla, Giuseppa Graceffa, Fabio Bucchieri, Everly Conway de Macario, Alberto J. L. Macario, and Francesca Rappa. 2021. "Molecular Chaperones and Thyroid Cancer" International Journal of Molecular Sciences 22, no. 8: 4196. https://doi.org/10.3390/ijms22084196

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop