Next Article in Journal
Opportunities, Challenges and Pitfalls of Using Cannabidiol as an Adjuvant Drug in COVID-19
Previous Article in Journal
Metabolic Syndrome and Reproduction
Previous Article in Special Issue
FOXE1 Gene Dosage Affects Thyroid Cancer Histology and Differentiation In Vivo
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Cell and Molecular Biology of Thyroid Disorders 2.0

1
Department of Biomedicine, Pharmacology, Aarhus University, Ole Worms Allé 4, 8000 Aarhus C, Denmark
2
Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
Int. J. Mol. Sci. 2021, 22(4), 1990; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22041990
Submission received: 4 January 2021 / Revised: 12 February 2021 / Accepted: 15 February 2021 / Published: 17 February 2021
(This article belongs to the Special Issue Cell and Molecular Biology of Thyroid Disorders 2.0)
This issue is the second volume of the previous Special Issue, “Cell and Molecular Biology of Thyroid Disorders” [1].
The thyroid is a hormonal gland in vertebrates and is located in mammals on the neck below the larynx in front of the trachea [2]. It has the shape of a butterfly in humans. Its proper function is of highest importance for our health. Very often, iodine deficiency causes a malfunction of the thyroid and plays a role in carcinogenesis [3,4,5]. Low iodine intake is associated with an increased risk of thyroid cancer (TC). It increases the frequency of appearance for more aggressive histotypes [3]. In addition, both hyperthyroidism and hypothyroidism exhibit detrimental effects on the human organism.
Two articles of this Special Issue report about Graves’ disease (GD) [6,7], also known as Basedow’s disease. GD is an autoimmune disorder resulting in hyperthyroidism with an overproduction of thyroid hormones.
This hyperthyroidism is characterized by the presence of autoantibodies which stimulate the thyroid-stimulating hormone receptor (TSHR) [8,9]. The conventional therapeutic regimen (e.g., antithyroid medication, radioiodine (RAI), or surgery) still lacks efficacy for a large number of patients. The lifelong thyroid hormone replacement therapy after RAI or surgical treatment is also problematic. Therefore, novel therapeutic approaches are necessary. New options include biologics, small molecules, peptide immunomodulation, and TSHR-specific treatment modalities; specific, targeted therapies are currently under investigation [8].
Another disorder of the thyroid is hypothyroidism, where an underactive thyroid gland is not producing the necessary amount of thyroid hormones [1]. One form is congenital hypothyroidism (CH) which is characterized by thyroid hormone deficiency present at birth. If untreated, this disease leads to growth failure and permanent intellectual disability. Babies born with CH may be asymptomatic, or may exhibit mild symptoms that often are not recognized.
In this issue, Kalveram et al. [10] published a research article about central CH, which is a rare and severe endocrine disorder. They demonstrated a mutation in the β-subunit of thyrotropin (thyroid-stimulating-hormone-B (TSH-B)). This mutation leads to isolated TSH-deficiency and to a severe phenotype.
A main topic of this Special Issue is the cell and molecular biology of thyroid carcinomas. TC can occur anywhere in the thyroid gland. It is the most common type of endocrine tumours [11]. Depending on which part of the thyroid tissue the tumour originates from, TCs are classified into several categories: differentiated (DTC), including follicular (FTC), papillary (PTC) and Hürthle cell cancer; as well as anaplastic thyroid cancer (ATC) and medullary (MTC) [11,12]. The American Cancer Society estimated about 52,890 new cases of TC (12,720 in men and 40,170 in women) and 2180 deaths from TC (1040 men and 1140 women) for thyroid cancer in the United States during 2020 [13].
The majority of the thyroid carcinomas are differentiated tumours. PTC comprise about 80–90% of the TC types, whereas poorly differentiated TC (PDTC) show a frequency of approximately 10% [14]. PDTC are characterized by low-differentiation, worse prognosis, and missing response to RAI-therapy [11].
The detection of molecular gene expression changes and alterations in the proteome in TC proposes new diagnostic and prognostic molecular markers and therapeutic targets [15,16,17]. Moreover, novel drugs such as tyrosine kinase inhibitors, antibodies, and small molecules have been introduced and tested in vitro, in animals, and in clinical trials [18,19,20,21].
In this Special Issue, a total of 12 excellent papers consisting of eight original articles [6,7,10,22,23,24,25,26], and four reviews [19,27,28,29] were published, as detailed in Table 1.
This collection includes nine manuscripts focusing on TC [19,22,23,24,25,26,27,28,29], two research articles on the topic of Graves’ disease [6,7], and one article with focus on central congenital hypothyroidism [10]. The TC studies published in this issue focused on prognostic, predictive markers or biomarkers [23,24,25,27], oncogenes [22] of TC, and anti-cancer drugs [19,26,28,29].
This Special Issue covers an ex vivo study with tumour specimen of patients [23], investigating metastatic and non-metastatic samples from PTC. The transcriptome oligonucleotide microarray technology was used to detect differences between M0 and M1 PTC. Moreover, an animal study (mice) was used to the effects of FOXE1 gene dosage reduction on cancer phenotype in vivo [22]. Single cell culture studies [10,24], in vitro studies with cell-free systems using human, rat, mouse, and rabbit hepatic microsomes [26], combined in vitro and ex vivo studies (tumour samples) [25], and single in vivo clinical studies [6,7] were included in this issue.
This Special Issue covered three studies investigating benign thyroid disorders. Graves’ disease is a very common one but with an aetiology that is still not fully understood. Polak et al. [7] investigated the relationship between the expression levels of TLR-2 and TLR-4 on CD4+ and CD8+ T lymphocytes and CD19+ B lymphocytes in patients with GD and selected clinical parameters. The authors concluded that TLR-2 and TLR-4 may serve as prognostic marker for Graves’ disease. The evaluation of peripheral blood lymphocytes expressing TLR-2 and TLR-4 suggested their important role in etiopathogenesis and clinical course of GD [7].
Another group investigated whether the Epstein–Barr Virus (EBV) plays a role in the pathogenesis of GD [6]. The authors found a significantly higher presence of EBV DNA copies in peripheral blood mononuclear cells (PBMCs) in patients newly diagnosed with GD as compared with controls. They concluded a probable role for EBV in the development of GD. EBV DNA had no effect on the severity of hyperthyroidism [6]. Central congenital hypothyroidism is a rare disorder and is caused by mutations in the β-subunit of thyrotropin (TSHB).
Kalveram et al. [10] characterized in vitro the pathogenic TSH β-Subunit Variant C105Vfs114X for signalling pathway modifications. They showed that this mutation induces changes in the signalling profile at the TSHR. They detected a strong decrease in cAMP signalling induction and conclude that this together with other changes in cell signalling is responsible for a severe phenotype.
Taken together, the new knowledge gained from these basic studies may impact translational research. New prognostic factors for GD or the future development of new compounds for targeting TSHR signalling are of interest.
A main focus of this issue was thyroid cancer research. Five original papers and four reviews with focus on TC were included in this issue. The transcription factor Forkhead box E1 (FOXE1) was investigated by Credendino et al. [22] in order to study the effects of a FOXE1 gene dosage reduction on the cancer phenotype in vivo (mice). The in vivo mouse model demonstrated that lower FOXE1 expression results in the development of less differentiated carcinomas, exhibiting lower proliferation and an increase in apoptosis. The authors concluded that FOXE1 could act as a lineage-specific oncogene [22].
Another study focused on the mechanisms of distant metastases (M1) in PTC [23]. It is unclear whether there are differences in gene expression profile between metastatic and non-metastatic PTC. Further investigations are necessary with hundreds of tumours. The authors detected differences in immune-related transcripts, indicating a possible role of tumour immune infiltration for metastasis [23].
PTC are diagnosed very often and have a very good prognosis, whereas ATC are rare and have a worse prognosis [11,12,24]. Tumour Suppressor Candidate 2 (TUSC2) is downregulated in ATC and PTC compared to benign thyroid tissue. Mariniello et al. [24] showed in vitro that TUSC2 overexpression reduced proliferation, migration, and the invasive potential of TC cells. In addition, TUSC2 elevated the TC cells’ sensitivity to apoptosis by induction of SMAC/DIABLO and cytochrome C proteins [24]. TUSC2 may serve as novel target and biomarker for TC.
A further study [25] investigated Prospero homeobox 1 (PROX1) which is involved in lymphangiogenesis. In a molecular in vitro study, a PROX1 knockdown resulted in an increase in various angiogenesis factors, which caused an elevation in new capillary-like structures by HUVECs and upregulated focal adhesion in the tumour cells [25]. PROX1 and FGF2 showed opposing expression levels in FTC tissues and seven thyroid tumour-derived cell lines [25]. This interesting study demonstrated the key role of PROX1 in the spreading behaviour of TC via regulation of angiogenesis [25]. PROX1 acts as a prognostic factor and is associated with patients’ outcomes [27]. An earlier study had proposed that the reactivation of PROX1 is a potential therapeutic strategy to attenuate disease progression in PTC [30]. Future cancer research is necessary to increase the current knowledge about PROX1 and to clarify whether it can support molecular-based treatment strategies in the clinic.
Vandetanib is a multikinase inhibitor (MKI) and used among others for the treatment of advanced MTC [31]. Indra et al. [26] published a pharmacological study investigating the microsomal metabolism of vandetanib. They identified human enzymes oxidizing vandetanib and explained the high efficiency of cytochrome P450 3A4 in the MKI’s oxidation. A review article supported this Special Issue with an update on MKI treatment (lenvatinib, sorafenib, sunitinib, cabozantinib, pazopanib, vandetanib) regarding the efficacy and safety profile in advanced refractory TC [19]. The application of these new drugs has shown favourable results in otherwise treatment-resistant TC.
Finally, the review by Varrichi et al. [28] completed this Special Issue. The authors reviewed novel data explaining how the immune system is involved in TC development and progression. In addition, cytokines are known to be involved in tumour growth and metastasis in FTC [32]. The authors discussed new results of treatment with monoclonal antibodies (mAbs) targeting immune checkpoints (IC) in patients with aggressive TCs. Monoclonal antibodies such as anti-cytotoxic T lymphocyte antigen 4 (anti-CTLA-4) or anti-programmed cell death protein-1/programmed cell death ligand-1 (anti-PD-1/PD-L1) had been used for tumour therapy, but 10% of the patients revealed a thyroid dysfunction. Therefore, combination strategies involving IC inhibitors with TKIs or serine/threonine protein kinase B-raf (BRAF) inhibitors are showing favourable effects in advanced TC.
Taken together, the 12 excellent publications included in this Special Issue demonstrate novel findings in the field of thyroid research.
I like to thank all the authors who supported this Special Issue. I am convinced that the application of new molecular biological technologies is helpful to improve the diagnosis and therapy of benign and malignant thyroid disorders. The detection of new biomarkers and the increasing knowledge of diagnosis, prognosis, novel targets, and new treatment strategies for TC will be important for supporting our fight against TC and contribute to reduce the mortality of advanced TC.

Funding

D.G. was funded by Deutsches Zentrum für Luft- und Raumfahrt (DLR), BMWi project 50WB1924.

Acknowledgments

I would like to thank Marcus Krüger and Markus Wehland, Otto von Guericke University Magdeburg, Germany for their help with EndNote and their important suggestions.

Conflicts of Interest

The author declares no conflict of interest.

Abbreviations

ATCAnaplastic thyroid cancer
anti-CTLA-4anti-cytotoxic T lymphocyte antigen 4
anti-PD-1anti-programmed cell death protein-1
anti-PD-L1anti-programmed cell death ligand-1
BRAFB-Raf (rapidly accelerated fibrosarcoma) proto-oncogene/threonine protein kinase B-
CAMP3′,5′-cyclic adenosine monophosphate
CHcongenital hypothyroidism
DIABLODiablo homolog
DTCDifferentiated thyroid cancer
EBVEpstein–Barr Virus
FGF2Fibroblast Growth Factor 2
FOXE1transcription factor Forkhead box E
FTCFollicular thyroid cancer
GDGraves’ Disease
HUVECsHuman umbilical vein endothelial cells
ICimmune checkpoints
MKI(s)Multi-kinase inhibitor(s)
MTCMedullary thyroid cancer
PROX1Prospero homeobox 1
PTCPapillary thyroid cancer
RAIRadioiodine
SMACsecond mitochondria-derived activator of caspases
TCThyroid cancer
TKIsTyrosine-kinase inhibitor(s)
TSHThyroid-stimulating hormone
TSHBThyroid-stimulating hormone beta
TSHRThyroid-stimulating hormone receptor
TUSC2Tumour Suppressor Candidate 2

References

  1. Grimm, D. Cell and molecular biology of thyroid disorders. Int. J. Mol. Sci. 2019, 20, 2895. [Google Scholar] [CrossRef] [Green Version]
  2. Allen, E.; Fingeret, A. Anatomy, head and neck, thyroid. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2020. [Google Scholar]
  3. Barrea, L.; Gallo, M.; Ruggeri, R.M.; Giacinto, P.D.; Sesti, F.; Prinzi, N.; Adinolfi, V.; Barucca, V.; Renzelli, V.; Muscogiuri, G.; et al. Nutritional status and follicular-derived thyroid cancer: An update. Crit. Rev. Food Sci. Nutr. 2021, 61, 25–59. [Google Scholar] [CrossRef] [PubMed]
  4. Chung, H.R. Iodine and thyroid function. Ann. Pediatr. Endocrinol. Metab. 2014, 19, 8–12. [Google Scholar] [CrossRef] [Green Version]
  5. Zimmermann, M.B.; Boelaert, K. Iodine deficiency and thyroid disorders. Lancet Diabetes Endocrinol. 2015, 3, 286–295. [Google Scholar] [CrossRef]
  6. Pyzik, A.; Grywalska, E.; Matyjaszek-Matuszek, B.; Ludian, J.; Kiszczak-Bochyńska, E.; Smoleń, A.; Roliński, J.; Pyzik, D. Does the epstein–barr virus play a role in the pathogenesis of graves’ disease? Int. J. Mol. Sci. 2019, 20, 3145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Polak, A.; Grywalska, E.; Klatka, J.; Roliński, J.; Matyjaszek-Matuszek, B.; Klatka, M. Toll-like receptors-2 and -4 in graves’ disease—key players or bystanders? Int. J. Mol. Sci. 2019, 20, 4732. [Google Scholar] [CrossRef] [Green Version]
  8. Lane, L.C.; Cheetham, T.D.; Perros, P.; Pearce, S.H.S. New therapeutic horizons for graves’ hyperthyroidism. Endocr. Rev. 2020, 41, 873–884. [Google Scholar] [CrossRef]
  9. Brent, G.A. Clinical practice. Graves’ disease. N. Engl. J. Med. 2008, 358, 2594–2605. [Google Scholar] [CrossRef]
  10. Kalveram, L.; Kleinau, G.; Szymańska, K.; Scheerer, P.; Rivero-Müller, A.; Grüters-Kieslich, A.; Biebermann, H. The pathogenic tsh β-subunit variant c105vfs114x causes a modified signaling profile at tshr. Int. J. Mol. Sci. 2019, 20, 5564. [Google Scholar] [CrossRef] [Green Version]
  11. Laursen, R.; Wehland, M.; Kopp, S.; Pietsch, J.; Infanger, M.; Grosse, J.; Grimm, D. Effects and role of multikinase inhibitors in thyroid cancer. Curr. Pharm. Des. 2016, 22, 5915–5926. [Google Scholar] [CrossRef]
  12. Grimm, D.; Bauer, J.; Wise, P.; Krüger, M.; Simonsen, U.; Wehland, M.; Infanger, M.; Corydon, T.J. The role of sox family members in solid tumours and metastasis. Semin. Cancer Biol. 2020, 67, 122–153. [Google Scholar] [CrossRef]
  13. Society, A.C. How common is thyroid cancer? Available online: https://www.cancer.org/cancer/thyroid-cancer/about/key-statistics.html (accessed on 3 January 2021).
  14. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
  15. Lin, P.; Yao, Z.; Sun, Y.; Li, W.; Liu, Y.; Liang, K.; Liu, Y.; Qin, J.; Hou, X.; Chen, L. Deciphering novel biomarkers of lymph node metastasis of thyroid papillary microcarcinoma using proteomic analysis of ultrasound-guided fine-needle aspiration biopsy samples. J. Proteom. 2019, 204, 103414. [Google Scholar] [CrossRef] [PubMed]
  16. Martínez-Aguilar, J.; Clifton-Bligh, R.; Molloy, M.P. Proteomics of thyroid tumours provides new insights into their molecular composition and changes associated with malignancy. Sci. Rep. 2016, 6, 23660. [Google Scholar] [CrossRef] [Green Version]
  17. Yip, L. Molecular markers for thyroid cancer diagnosis, prognosis, and targeted therapy. J. Surg. Oncol. 2015, 111, 43–50. [Google Scholar] [CrossRef]
  18. Ancker, O.V.; Wehland, M.; Bauer, J.; Infanger, M.; Grimm, D. The adverse effect of hypertension in the treatment of thyroid cancer with multi-kinase inhibitors. Int. J. Mol. Sci. 2017, 18, 625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Ancker, O.V.; Krüger, M.; Wehland, M.; Infanger, M.; Grimm, D. Multikinase inhibitor treatment in thyroid cancer. Int. J. Mol. Sci. 2019, 21, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Jayarangaiah, A.; Sidhu, G.; Brown, J.; Barrett-Campbell, O.; Bahtiyar, G.; Youssef, I.; Arora, S.; Skwiersky, S.; McFarlane, S.I. Therapeutic options for advanced thyroid cancer. Int. J. Clin. Endocrinol. Metab. 2019, 5, 26–34. [Google Scholar] [CrossRef] [Green Version]
  21. Fallahi, P.; Ferrari, S.M.; Galdiero, M.R.; Varricchi, G.; Elia, G.; Ragusa, F.; Paparo, S.R.; Benvenga, S.; Antonelli, A. Molecular targets of tyrosine kinase inhibitors in thyroid cancer. Semin. Cancer Biol. 2020. [Google Scholar] [CrossRef]
  22. Credendino, S.C.; Moccia, C.; Amendola, E.; D’Avino, G.; Di Guida, L.; Clery, E.; Greco, A.; Bellevicine, C.; Brunetti, A.; De Felice, M.; et al. Foxe1 gene dosage affects thyroid cancer histology and differentiation in vivo. Int. J. Mol. Sci. 2021, 22, 25. [Google Scholar] [CrossRef]
  23. Szpak-Ulczok, S.; Pfeifer, A.; Rusinek, D.; Oczko-Wojciechowska, M.; Kowalska, M.; Tyszkiewicz, T.; Cieslicka, M.; Handkiewicz-Junak, D.; Fujarewicz, K.; Lange, D.; et al. Differences in gene expression profile of primary tumors in metastatic and non-metastatic papillary thyroid carcinoma-do they exist? Int. J. Mol. Sci. 2020, 21, 4629. [Google Scholar] [CrossRef] [PubMed]
  24. Mariniello, R.M.; Maria Orlandella, F.; De Stefano, A.E.; Iervolino, P.L.C.; Smaldone, G.; Luciano, N.; Cervone, N.; Munciguerra, F.; Esposito, S.; Mirabelli, P.; et al. The tusc2 tumour suppressor inhibits the malignant phenotype of human thyroid cancer cells via smac/diablo protein. Int. J. Mol. Sci. 2020, 21, 702. [Google Scholar] [CrossRef] [Green Version]
  25. Rudzińska, M.; Mikula, M.; Arczewska, K.D.; Gajda, E.; Sabalińska, S.; Stępień, T.; Ostrowski, J.; Czarnocka, B. Transcription factor prospero homeobox 1 (prox1) as a potential angiogenic regulator of follicular thyroid cancer dissemination. Int. J. Mol. Sci. 2019, 20, 5619. [Google Scholar] [CrossRef] [Green Version]
  26. Indra, R.; Pompach, P.; Martínek, V.; Takácsová, P.; Vavrová, K.; Heger, Z.; Adam, V.; Eckschlager, T.; Kopečková, K.; Arlt, V.M.; et al. Identification of human enzymes oxidizing the anti-thyroid-cancer drug vandetanib and explanation of the high efficiency of cytochrome p450 3a4 in its oxidation. Int. J. Mol. Sci. 2019, 20, 3392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Rudzińska, M.; Czarnocka, B. The impact of transcription factor prospero homeobox 1 on the regulation of thyroid cancer malignancy. Int. J. Mol. Sci. 2020, 21, 3220. [Google Scholar] [CrossRef] [PubMed]
  28. Varricchi, G.; Loffredo, S.; Marone, G.; Modestino, L.; Fallahi, P.; Ferrari, S.M.; de Paulis, A.; Antonelli, A.; Galdiero, M.R. The immune landscape of thyroid cancer in the context of immune checkpoint inhibition. Int. J. Mol. Sci. 2019, 20, 3934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Manzella, L.; Massimino, M.; Stella, S.; Tirrò, E.; Pennisi, M.S.; Martorana, F.; Motta, G.; Vitale, S.R.; Puma, A.; Romano, C.; et al. Activation of the igf axis in thyroid cancer: Implications for tumorigenesis and treatment. Int. J. Mol. Sci. 2019, 20, 3258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Choi, D.; Ramu, S.; Park, E.; Jung, E.; Yang, S.; Jung, W.; Choi, I.; Lee, S.; Kim, K.E.; Seong, Y.J.; et al. Aberrant activation of notch signaling inhibits prox1 activity to enhance the malignant behavior of thyroid cancer cells. Cancer Res. 2016, 76, 582–593. [Google Scholar] [CrossRef] [Green Version]
  31. Milling, R.V.; Grimm, D.; Krüger, M.; Grosse, J.; Kopp, S.; Bauer, J.; Infanger, M.; Wehland, M. Pazopanib, cabozantinib, and vandetanib in the treatment of progressive medullary thyroid cancer with a special focus on the adverse effects on hypertension. Int. J. Mol. Sci. 2018, 19, 3258. [Google Scholar] [CrossRef] [Green Version]
  32. Svejgaard, B.; Wehland, M.; Ma, X.; Kopp, S.; Sahana, J.; Warnke, E.; Aleshcheva, G.; Hemmersbach, R.; Hauslage, J.; Grosse, J.; et al. Common effects on cancer cells exerted by a random positioning machine and a 2D clinostat. PLoS ONE 2015, 10, e0135157. [Google Scholar] [CrossRef] [Green Version]
Table 1. Contributions to the Special Issue “Cell and Molecular Biology of Thyroid Disorders 2.0”.
Table 1. Contributions to the Special Issue “Cell and Molecular Biology of Thyroid Disorders 2.0”.
AuthorTitleTopicsTypeReference
Credendino S.C., et al.Foxe1 gene dosage modulates thyroid cancer histology and differentiation in vivoFOXE1 behaves as lineage-specific oncogene in follicular cell-derived TCResearch Article[22]
Szpak-Ulczok S., et al.Differences in Gene Expression Profile of Primary Tumours in Metastatic and Non-Metastatic Papillary Thyroid Carcinoma—Do They Exist?Valuable prognostic and predictive markers for distant metastases in PTCResearch article[23]
Mariniello R.M., et al.The TUSC2 Tumour Suppressor Inhibits the Malignant Phenotype of Human Thyroid Cancer Cells via SMAC/DIABLO Protein • Tumour suppressor role of TUSC2 in thyroid carcinogenesis
• promising target and biomarker for TC
Research article[24]
Rudzińska M., et al.Transcription Factor Prospero Homeobox 1 (PROX1) as a Potential Angiogenic Regulator of Follicular Thyroid Cancer Dissemination in vitro study: PROX1 is involved in the spreading of TC cells by regulation of angiogenesis.Research article[25]
Kalveram L., et al.The Pathogenic TSH β-Subunit Variant C105Vfs114X Causes a Modified Signalling Profile at TSHRCentral congenital hypothyroidism:
• TSHB mutation C105Vfs114X
• strong decrease in cAMP signalling
Research Article[10]
Polak A., et al.Toll-Like Receptors-2 and -4 in Graves’ Disease—Key Players or Bystanders?Graves’ Disease (GD): relationship between TLR-2/-4 to CD4+/CD8+ T- lymphocytes and CD19+ B- lymphocytes in patientsResearch Article[7]
Indra R., et al.Identification of Human Enzymes Oxidizing the Anti-Thyroid-Cancer Drug Vandetanib and Explanation of the High Efficiency of Cytochrome P450 3A4 in its OxidationThe metabolism of the TKI vandetanib used for treatment of symptomatic/progressive MTC, was studied using human hepatic microsomes, recombinant cytochromes P450 (CYPs) and flavin-containing monooxygenases (FMOs).Research Article[26]
Pyzik A., et al.Does the Epstein–Barr Virus Play a Role in the Pathogenesis of Graves’ Disease?Graves’ Disease: probable role of EBV in GD development. EBV infection does not affect the clinical picture of GD.Research Article[6]
Rudzińska M. and Czarnocka B.The Impact of Transcription Factor Prospero Homeobox 1 on the Regulation of Thyroid Cancer Malignancy• PROX1 as potential prognostic marker
• Its role in differentiated TC
Review[27]
Ancker O.V., et al.Multikinase Inhibitor Treatment in Thyroid CancerMultikinase inhibitors (MKIs) can be used in the treatment of advanced refractory TCs.Review[19]
Varricchi G., et al.The Immune Landscape of Thyroid Cancer in the Context of Immune Checkpoint Inhibition• Contribution of different immune cells to thyroid cancer development
• Rationale for the antitumor effects of ICIs in combination with BRAF/TK inhibitors
Review[28]
Manzella L., et al.Activation of the IGF Axis in Thyroid Cancer: Implications for Tumorigenesis and TreatmentRole of the IGF axis in thyroid tumorigenesis update on the current knowledge of IGF-targeted combination therapies for TCReview[29]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Grimm, D. Cell and Molecular Biology of Thyroid Disorders 2.0. Int. J. Mol. Sci. 2021, 22, 1990. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22041990

AMA Style

Grimm D. Cell and Molecular Biology of Thyroid Disorders 2.0. International Journal of Molecular Sciences. 2021; 22(4):1990. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22041990

Chicago/Turabian Style

Grimm, Daniela. 2021. "Cell and Molecular Biology of Thyroid Disorders 2.0" International Journal of Molecular Sciences 22, no. 4: 1990. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22041990

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